1
|
Ainciburu M, Ezponda T, Berastegui N, Alfonso-Pierola A, Vilas-Zornoza A, San Martin-Uriz P, Alignani D, Lamo-Espinosa J, San-Julian M, Jiménez-Solas T, Lopez F, Muntion S, Sanchez-Guijo F, Molero A, Montoro J, Serrano G, Diaz-Mazkiaran A, Lasaga M, Gomez-Cabrero D, Diez-Campelo M, Valcarcel D, Hernaez M, Romero JP, Prosper F. Uncovering perturbations in human hematopoiesis associated with healthy aging and myeloid malignancies at single-cell resolution. eLife 2023; 12:79363. [PMID: 36629404 PMCID: PMC9904760 DOI: 10.7554/elife.79363] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 01/10/2023] [Indexed: 01/12/2023] Open
Abstract
Early hematopoiesis is a continuous process in which hematopoietic stem and progenitor cells (HSPCs) gradually differentiate toward specific lineages. Aging and myeloid malignant transformation are characterized by changes in the composition and regulation of HSPCs. In this study, we used single-cell RNA sequencing (scRNA-seq) to characterize an enriched population of human HSPCs obtained from young and elderly healthy individuals. Based on their transcriptional profile, we identified changes in the proportions of progenitor compartments during aging, and differences in their functionality, as evidenced by gene set enrichment analysis. Trajectory inference revealed that altered gene expression dynamics accompanied cell differentiation, which could explain aging-associated changes in hematopoiesis. Next, we focused on key regulators of transcription by constructing gene regulatory networks (GRNs) and detected regulons that were specifically active in elderly individuals. Using previous findings in healthy cells as a reference, we analyzed scRNA-seq data obtained from patients with myelodysplastic syndrome (MDS) and detected specific alterations of the expression dynamics of genes involved in erythroid differentiation in all patients with MDS such as TRIB2. In addition, the comparison between transcriptional programs and GRNs regulating normal HSPCs and MDS HSPCs allowed identification of regulons that were specifically active in MDS cases such as SMAD1, HOXA6, POU2F2, and RUNX1 suggesting a role of these transcription factors (TFs) in the pathogenesis of the disease. In summary, we demonstrate that the combination of single-cell technologies with computational analysis tools enable the study of a variety of cellular mechanisms involved in complex biological systems such as early hematopoiesis and can be used to dissect perturbed differentiation trajectories associated with perturbations such as aging and malignant transformation. Furthermore, the identification of abnormal regulatory mechanisms associated with myeloid malignancies could be exploited for personalized therapeutic approaches in individual patients.
Collapse
Affiliation(s)
- Marina Ainciburu
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
| | - Teresa Ezponda
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
| | - Nerea Berastegui
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
| | - Ana Alfonso-Pierola
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
- Clinica Universidad de NavarraPamplonaSpain
| | - Amaia Vilas-Zornoza
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
| | - Patxi San Martin-Uriz
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
| | - Diego Alignani
- Flow Cytometry Core, Universidad de NavarraPamplonaSpain
| | | | | | | | - Felix Lopez
- Hospital Universitario de SalamancaSalamancaSpain
| | - Sandra Muntion
- Hospital Universitario de SalamancaSalamancaSpain
- Red de Investigación Cooperativa en Terapia Celular TerCel, ISCIII.MadridSpain
| | - Fermin Sanchez-Guijo
- Hospital Universitario de SalamancaSalamancaSpain
- Red de Investigación Cooperativa en Terapia Celular TerCel, ISCIII.MadridSpain
| | - Antonieta Molero
- Department of Hematology, Vall d'Hebron Hospital UniversitariBarcelonaSpain
| | - Julia Montoro
- Department of Hematology, Vall d'Hebron Hospital UniversitariBarcelonaSpain
| | | | - Aintzane Diaz-Mazkiaran
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
- Computational Biology Program, Universidad de NavarraPamplonaSpain
| | - Miren Lasaga
- Translational Bioinformatics Unit, NavarraBiomedPamplonaSpain
| | - David Gomez-Cabrero
- Translational Bioinformatics Unit, NavarraBiomedPamplonaSpain
- Biological & Environmental Sciences & Engineering Division, King Abdullah University of Science and TechnologyThuwalSaudi Arabia
| | | | - David Valcarcel
- Department of Hematology, Vall d'Hebron Hospital UniversitariBarcelonaSpain
| | - Mikel Hernaez
- Computational Biology Program, Universidad de NavarraPamplonaSpain
| | - Juan P Romero
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
| | - Felipe Prosper
- Area de Hemato-Oncología, Centro de Investigación Médica Aplicada, Universidad de Navarra, Instituto de investigación sanitaria de Navarra (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de CáncerMadridSpain
- Clinica Universidad de NavarraPamplonaSpain
- Red de Investigación Cooperativa en Terapia Celular TerCel, ISCIII.MadridSpain
| |
Collapse
|
2
|
Berastegui N, Ainciburu M, Romero JP, Garcia-Olloqui P, Alfonso-Pierola A, Philippe C, Vilas-Zornoza A, San Martin-Uriz P, Ruiz-Hernández R, Abarrategi A, Ordoñez R, Alignani D, Sarvide S, Castro-Labrador L, Lamo-Espinosa JM, San-Julian M, Jimenez T, López-Cadenas F, Muntion S, Sanchez-Guijo F, Molero A, Montoro MJ, Tazón B, Serrano G, Diaz-Mazkiaran A, Hernaez M, Huerga S, Bewicke-Copley F, Rio-Machin A, Maurano MT, Díez-Campelo M, Valcarcel D, Rouault-Pierre K, Lara-Astiaso D, Ezponda T, Prosper F. The transcription factor DDIT3 is a potential driver of dyserythropoiesis in myelodysplastic syndromes. Nat Commun 2022; 13:7619. [PMID: 36494342 PMCID: PMC9734135 DOI: 10.1038/s41467-022-35192-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are hematopoietic stem cell (HSC) malignancies characterized by ineffective hematopoiesis, with increased incidence in older individuals. Here we analyze the transcriptome of human HSCs purified from young and older healthy adults, as well as MDS patients, identifying transcriptional alterations following different patterns of expression. While aging-associated lesions seem to predispose HSCs to myeloid transformation, disease-specific alterations may trigger MDS development. Among MDS-specific lesions, we detect the upregulation of the transcription factor DNA Damage Inducible Transcript 3 (DDIT3). Overexpression of DDIT3 in human healthy HSCs induces an MDS-like transcriptional state, and dyserythropoiesis, an effect associated with a failure in the activation of transcriptional programs required for normal erythroid differentiation. Moreover, DDIT3 knockdown in CD34+ cells from MDS patients with anemia is able to restore erythropoiesis. These results identify DDIT3 as a driver of dyserythropoiesis, and a potential therapeutic target to restore the inefficient erythroid differentiation characterizing MDS patients.
Collapse
Affiliation(s)
- Nerea Berastegui
- Department of Hematology-Oncology, CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Marina Ainciburu
- Department of Hematology-Oncology, CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan P Romero
- Department of Hematology-Oncology, CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Paula Garcia-Olloqui
- Department of Hematology-Oncology, CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Alfonso-Pierola
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Hematology, Clínica Universidad de Navarra, Universidad de Navarra and CCUN, 31008, Pamplona, Spain
| | - Céline Philippe
- Department of Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, England, UK
| | - Amaia Vilas-Zornoza
- Department of Hematology-Oncology, CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Patxi San Martin-Uriz
- Department of Hematology-Oncology, CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - Raquel Ruiz-Hernández
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), San Sebastian, Spain
| | - Ander Abarrategi
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Raquel Ordoñez
- Institute for Systems Genetics, NYU School of Medicine, New York, NY, USA
| | - Diego Alignani
- Department of Hematology-Oncology, CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Sarai Sarvide
- Department of Hematology-Oncology, CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Castro-Labrador
- Department of Hematology-Oncology, CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - José M Lamo-Espinosa
- Department of Orthopedic Surgery and Traumatology, Clínica Universidad de Navarra, Universidad de Navarra and CCUN, 31008, Pamplona, Spain
| | - Mikel San-Julian
- Department of Orthopedic Surgery and Traumatology, Clínica Universidad de Navarra, Universidad de Navarra and CCUN, 31008, Pamplona, Spain
| | - Tamara Jimenez
- Department of Hematology, Hospital Universitario de Salamanca-IBSAL, Universidad de Salamanca, Salamanca, Spain
| | - Félix López-Cadenas
- Department of Hematology, Hospital Universitario de Salamanca-IBSAL, Universidad de Salamanca, Salamanca, Spain
| | - Sandra Muntion
- Department of Hematology, Hospital Universitario de Salamanca-IBSAL, Universidad de Salamanca, Salamanca, Spain
| | - Fermin Sanchez-Guijo
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Hematology, Hospital Universitario de Salamanca-IBSAL, Universidad de Salamanca, Salamanca, Spain
| | - Antonieta Molero
- Department of Hematology, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Maria Julia Montoro
- Department of Hematology, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Bárbara Tazón
- Department of Hematology, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Guillermo Serrano
- Computational Biology Program, Institute for data science and artificial intelligence (datai), CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Navarra, Spain
| | - Aintzane Diaz-Mazkiaran
- Department of Hematology-Oncology, CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Computational Biology Program, Institute for data science and artificial intelligence (datai), CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Navarra, Spain
| | - Mikel Hernaez
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Computational Biology Program, Institute for data science and artificial intelligence (datai), CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Navarra, Spain
| | - Sofía Huerga
- Department of Hematology, Clínica Universidad de Navarra, Universidad de Navarra and CCUN, 31008, Pamplona, Spain
| | - Findlay Bewicke-Copley
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Ana Rio-Machin
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Matthew T Maurano
- Institute for Systems Genetics, NYU School of Medicine, New York, NY, USA
- Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - María Díez-Campelo
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Hematology, Hospital Universitario de Salamanca-IBSAL, Universidad de Salamanca, Salamanca, Spain
| | - David Valcarcel
- Department of Hematology, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Kevin Rouault-Pierre
- Department of Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, England, UK
| | - David Lara-Astiaso
- Department of Hematology-Oncology, CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - Teresa Ezponda
- Department of Hematology-Oncology, CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| | - Felipe Prosper
- Department of Hematology-Oncology, CIMA Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
- Department of Hematology, Clínica Universidad de Navarra, Universidad de Navarra and CCUN, 31008, Pamplona, Spain.
| |
Collapse
|
3
|
Im H, Rao V, Sridhar K, Bentley J, Mishra T, Chen R, Hall J, Graber A, Zhang Y, Li X, Mias GI, Snyder MP, Greenberg PL. Distinct transcriptomic and exomic abnormalities within myelodysplastic syndrome marrow cells. Leuk Lymphoma 2018; 59:2952-2962. [PMID: 29616851 DOI: 10.1080/10428194.2018.1452210] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
To provide biologic insights into mechanisms underlying myelodysplastic syndromes (MDS) we evaluated the CD34+ marrow cells transcriptome using high-throughput RNA sequencing (RNA-Seq). We demonstrated significant differential gene expression profiles (GEPs) between MDS and normal and identified 41 disease classifier genes. Additionally, two main clusters of GEPs distinguished patients based on their major clinical features, particularly between those whose disease remained stable versus patients who transformed into acute myeloid leukemia within 12 months. The genes whose expression was associated with disease outcome were involved in functional pathways and biologic processes highly relevant for MDS. Combined with exomic analysis we identified differential isoform usage of genes in MDS mutational subgroups, with consequent dysregulation of distinct biologic functions. This combination of clinical, transcriptomic and exomic findings provides valuable understanding of mechanisms underlying MDS and its progression to a more aggressive stage and also facilitates prognostic characterization of MDS patients.
Collapse
Affiliation(s)
- Hogune Im
- a Department of Genetics , Stanford University School of Medicine , Stanford , CA , USA
| | - Varsha Rao
- a Department of Genetics , Stanford University School of Medicine , Stanford , CA , USA
| | - Kunju Sridhar
- b Hematology Division , Stanford University School of Medicine, Stanford Cancer Institute , Stanford , CA , USA
| | - Jason Bentley
- c Quantitative Science Unit , Stanford University , Stanford , CA , USA
| | - Tejaswini Mishra
- a Department of Genetics , Stanford University School of Medicine , Stanford , CA , USA
| | - Rui Chen
- a Department of Genetics , Stanford University School of Medicine , Stanford , CA , USA
| | - Jeff Hall
- d Genoptix Inc. , Carlsbad , CA , USA
| | | | - Yan Zhang
- e Department of Hematology , Jiaotong University, 6th Hospital , Shanghai , China
| | - Xiao Li
- e Department of Hematology , Jiaotong University, 6th Hospital , Shanghai , China
| | - George I Mias
- f Department of Biochemistry and Molecular Biology , Michigan State University , East Lansing , MI , USA
| | - Michael P Snyder
- a Department of Genetics , Stanford University School of Medicine , Stanford , CA , USA
| | - Peter L Greenberg
- b Hematology Division , Stanford University School of Medicine, Stanford Cancer Institute , Stanford , CA , USA
| |
Collapse
|
4
|
Lee JS, Choi HJ, Baek SH. Sumoylation and Its Contribution to Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 963:283-298. [PMID: 28197919 DOI: 10.1007/978-3-319-50044-7_17] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Post-translational modifications play an important role in regulating protein activity by altering their functions. Sumoylation is a highly dynamic process which is tightly regulated by a fine balance between conjugating and deconjugating enzyme activities. It affects intracellular localization and their interaction with their binding partners, thereby changing gene expression. Consequently, these changes in turn affect signaling mechanisms that regulate many cellular functions, such as cell growth, proliferation, apoptosis , DNA repair , and cell survival. It is becoming apparent that deregulation in the SUMO pathway contributes to oncogenic transformation by affecting sumoylation/desumoylation of many oncoproteins and tumor suppressors. Loss of balance between sumoylation and desumoylation has been reported in a number of studies in a variety of disease types including cancer. This chapter summarizes the mechanisms and functions of the deregulated SUMO pathway affecting oncogenes and tumor suppressor genes.
Collapse
Affiliation(s)
- Jason S Lee
- Department of Biological Sciences, Seoul National University, Seoul, 151-742, South Korea
| | - Hee June Choi
- Department of Biological Sciences, Seoul National University, Seoul, 151-742, South Korea
| | - Sung Hee Baek
- Department of Biological Sciences, Seoul National University, Seoul, 151-742, South Korea.
| |
Collapse
|
5
|
Valent P, Orazi A, Steensma DP, Ebert BL, Haase D, Malcovati L, van de Loosdrecht AA, Haferlach T, Westers TM, Wells DA, Giagounidis A, Loken M, Orfao A, Lübbert M, Ganser A, Hofmann WK, Ogata K, Schanz J, Béné MC, Hoermann G, Sperr WR, Sotlar K, Bettelheim P, Stauder R, Pfeilstöcker M, Horny HP, Germing U, Greenberg P, Bennett JM. Proposed minimal diagnostic criteria for myelodysplastic syndromes (MDS) and potential pre-MDS conditions. Oncotarget 2017; 8:73483-73500. [PMID: 29088721 PMCID: PMC5650276 DOI: 10.18632/oncotarget.19008] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/26/2017] [Indexed: 12/13/2022] Open
Abstract
Myelodysplastic syndromes (MDS) comprise a heterogeneous group of myeloid neoplasms characterized by peripheral cytopenia, dysplasia, and a variable clinical course with about 30% risk to transform to secondary acute myeloid leukemia (AML). In the past 15 years, diagnostic evaluations, prognostication, and treatment of MDS have improved substantially. However, with the discovery of molecular markers and advent of novel targeted therapies, new challenges have emerged in the complex field of MDS. For example, MDS-related molecular lesions may be detectable in healthy individuals and increase in prevalence with age. Other patients exhibit persistent cytopenia of unknown etiology without dysplasia. Although these conditions are potential pre-phases of MDS they may also transform into other bone marrow neoplasms. Recently identified molecular, cytogenetic, and flow-based parameters may add in the delineation and prognostication of these conditions. However, no generally accepted integrated classification and no related criteria are as yet available. In an attempt to address this challenge, an international consensus group discussed these issues in a working conference in July 2016. The outcomes of this conference are summarized in the present article which includes criteria and a proposal for the classification of pre-MDS conditions as well as updated minimal diagnostic criteria of MDS. Moreover, we propose diagnostic standards to delineate between ´normal´, pre-MDS, and MDS. These standards and criteria should facilitate diagnostic and prognostic evaluations in clinical studies as well as in clinical practice.
Collapse
Affiliation(s)
- Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Attilio Orazi
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - David P Steensma
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Benjamin L Ebert
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Detlef Haase
- Clinic of Hematology and Medical Oncology, Universitymedicine Göttingen, Göttingen, Germany
| | - Luca Malcovati
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Arjan A van de Loosdrecht
- Department of Hematology Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Theresia M Westers
- Department of Hematology Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | - Alberto Orfao
- Servicio Central de Citometría, Centro de Investigación del Cáncer (IBMCC, CSIC-USAL) and IBSAL, Universidad de Salamanca, Salamanca, Spain
| | - Michael Lübbert
- Department of Medicine I, Medical Center-University of Freiburg, Freiburg, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Kiyoyuki Ogata
- Metropolitan Research and Treatment Center for Blood Disorders (MRTC Japan), Tokyo, Japan
| | - Julie Schanz
- Clinic of Hematology and Medical Oncology, Universitymedicine Göttingen, Göttingen, Germany
| | - Marie C Béné
- Laboratoire d'Hématologie CHU de Nantes, Nantes, France
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Wolfgang R Sperr
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Karl Sotlar
- Institute of Pathology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | | | - Reinhard Stauder
- Department of Internal Medicine V (Haematology and Oncology) Innsbruck Medical University, Innsbruck, Austria
| | | | - Hans-Peter Horny
- Institute of Pathology, Ludwig-Maximilians University, Munich, Germany
| | - Ulrich Germing
- Department of Hematology, Oncology, and Clinical Immunology, Heinrich-Heine-University, Düsseldorf, Germany
| | | | - John M Bennett
- Department of Pathology, Hematopathology Unit and James P Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
6
|
Abstract
SUMOylation is a key post-translational modification that regulates crucial cellular functions and pathological processes. Recently, Small Ubiquitin-related MOdifier (SUMO) modification has emerged as a fundamental route that may drive different steps of human tumorigenesis. Indeed, alteration in expression or activity of one of the different SUMO pathway components may completely subvert cellular properties through fine-tuning modulation of protein(s) involved in carcinogenic pathways, leading to altered cell proliferation, apoptosis resistance and metastatic potential. Here we describe some of the most interesting findings pointing to a clear link between SUMO pathway and human malignancies. Importantly, a putative role for SUMO enzymes to predict cancer behavior can be speculated, and thus the possible application of alterations in SUMO pathway components as tumor biomarkers is discussed.
Collapse
Affiliation(s)
- Domenico Mattoscio
- 1Department of Experimental Oncology, European Institute of Oncology@ IFOM-IEO Campus, Via Adamello 16, 20139 Milan, Italy
| | - Susanna Chiocca
- 1Department of Experimental Oncology, European Institute of Oncology@ IFOM-IEO Campus, Via Adamello 16, 20139 Milan, Italy
| |
Collapse
|
7
|
Zhao W, Zhao W, Zhao J, Wang D, Li J. Screening of potential target genes for cataract by analyzing mRNA expression profile of mouse Hsf4-null lens. BMC Ophthalmol 2015; 15:76. [PMID: 26187041 PMCID: PMC4506420 DOI: 10.1186/s12886-015-0066-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 07/08/2015] [Indexed: 01/18/2023] Open
Abstract
Background Hsf4 is closely related to the development of cataract. However, the molecular mechanisms remain unknown. This study aimed to explore the molecular mechanisms that how Hsf4 mutations influence development of lens and thus lead to cataract in mouse. Methods The mRNA expression profile of mouse tissue samples from Hsf4-null and wile-type lenses was downloaded from Gene Expression Omnibus database. Then the LIMMA package was used to screen differentially expressed genes (DEGs) and DAVID was applied to identify the significantly enriched Gene Ontology (GO) categories for DEGs. Furthermore, the protein-protein interaction (PPI) network of DEGs was constructed using Cytoscape and the key modules were selected from the PPI network based on the MCODE analysis. Results A total of 216 DEGs were screened, including 51 up- and 165 down-regulated genes. Meanwhile, nine GO terms were obtained, and DEGs such as SGK1, CRY2 and REV1 were enriched in response to DNA damage stimulus. Furthermore, 89 DEGs and 99 gene pairs were mapped into the PPI network and Ubc was the hob node. Two key modules, which contained the genes (e.g. Ubc, Egr1, Ptgs2, Hmox1, Cd44, Btg2, Cyr61 and Fos) were related to response to DNA damage stimulus. Conclusions The deletion of Hsf4 affects the expression of many genes, such as Ubc, Ptgs2, Egr1 and Fos. These genes may be involved in the development of cataract and could be used as therapeutic targets for cataract.
Collapse
Affiliation(s)
- Wenjuan Zhao
- Department of Ophthalmology, Qilu Hospital of Shandong University, Wenhuaxi Road 107, Jinan, Shandong, 250012, China. .,Department of Ophthalmology, Shandong University Affiliated Jinan Central Hospital, Jinan, 250013, China.
| | - Wenqing Zhao
- Department of Neurosurgery, The 5th People's Hospital of Jinan, Jinan, 250022, China.
| | - Jun Zhao
- Health Examination Center, Jinan 2nd People's Hospital, Jinan, 250001, China.
| | - Dong Wang
- School of Management Science and Engineering, Shandong University of Finance and Economics, Jinan, 250014, China.
| | - Jinghai Li
- Department of Ophthalmology, Qilu Hospital of Shandong University, Wenhuaxi Road 107, Jinan, Shandong, 250012, China.
| |
Collapse
|
8
|
Targeting JAK kinase in solid tumors: emerging opportunities and challenges. Oncogene 2015; 35:939-51. [DOI: 10.1038/onc.2015.150] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 03/24/2015] [Accepted: 03/24/2015] [Indexed: 02/07/2023]
|
9
|
Bellido M, Stirewalt DL, Zhao LP, Radich JP. Use of gene expression microarrays for the study of acute leukemia. Expert Rev Mol Diagn 2014; 6:733-47. [PMID: 17009907 DOI: 10.1586/14737159.6.5.733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Genetic lesions found in acute leukemia drive the pathology of the disease in addition to forming reliable classifications of prognosis. However, there is still a reasonable heterogeneity of response among cases with the same genetic lesion. Moreover, many leukemia cases have no detectable genetic marker and these cases have marked heterogeneity of response. How can we learn more about the genes and pathways involved with leukemogenesis and response in the midst of such complexity? Gene expression microarrays are experimental platforms that allow for the simultaneous evaluation of the thousands of mRNA transcripts (the 'transcriptome'). This technology has revolutionized the study of leukemia, giving insight into genes and pathways involved in disease response and the biology involved in specific translocations.
Collapse
Affiliation(s)
- Mar Bellido
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Public Health Sciences Division, 1100 Fairview Ave N., Seattle, WA 98109, USA.
| | | | | | | |
Collapse
|
10
|
Cherian S, Bagg A. The genetics of the myelodysplastic syndromes: Classical cytogenetics and recent molecular insights. Hematology 2013; 11:1-13. [PMID: 16522543 DOI: 10.1080/10245330500276691] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are a complex group of clonal hematopoietic disorders with an attendant diverse array of associated genetic changes. Conventional cytogenetics plays a prominent and well-established role in determining the contemporary diagnosis and prognosis of these disorders. More recently, molecular approaches have been useful in further characterizing this group of diseases, albeit in a largely experimental context, with the detection of changes at the single gene level including mutations, amplification and epigenetic phenomena. Nevertheless, we remain largely ignorant of the genetic underpinnings of MDS. Here we briefly review the established role of cytogenetics in MDS, and emphasize recent advances in unraveling the genetics of MDS, with a view towards how such findings might facilitate our ability to understand, diagnose and treat these disorders in a more rational manner.
Collapse
Affiliation(s)
- Sindhu Cherian
- University of Washington Medical Center, Department of Laboratory Medicine, Seattle, 98195, USA.
| | | |
Collapse
|
11
|
Pellagatti A, Fidler C, Wainscoat JS, Boultwood J. Gene expression profiling in the myelodysplastic syndromes. Hematology 2013; 10:281-7. [PMID: 16085540 DOI: 10.1080/10245330500065680] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The myelodysplastic syndromes (MDS) are a heterogeneous group of haematopoietic malignancies, characterized by blood cytopenias, ineffective hematopoiesis and hypercellular bone marrow. Several genetic alterations have been reported in MDS but these are not MDS-specific and the underlying molecular causes of the disease remain poorly understood. Gene expression microarray technology allows the simultaneous parallel analysis of many thousands of genes and has already provided novel insights into cancer pathogenesis. In this review we discuss the results of several recent studies which utilize the enormous power of microarray technology for the study of MDS. Several exciting findings have emerged from these early studies that highlight the potential of this technology to further our understanding of the molecular pathogenesis of this disorder. It is clear, however, that these findings should be confirmed in larger sets of MDS patients.
Collapse
Affiliation(s)
- Andrea Pellagatti
- Leukaemia Research Fund Molecular Haematology Unit, Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | | | | | | |
Collapse
|
12
|
Baratti MO, Moreira YB, Traina F, Costa FF, Verjovski-Almeida S, Olalla-Saad ST. Identification of protein-coding and non-coding RNA expression profiles in CD34+ and in stromal cells in refractory anemia with ringed sideroblasts. BMC Med Genomics 2010; 3:30. [PMID: 20633296 PMCID: PMC2914047 DOI: 10.1186/1755-8794-3-30] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Accepted: 07/15/2010] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Myelodysplastic syndromes (MDS) are a group of clonal hematological disorders characterized by ineffective hematopoiesis with morphological evidence of marrow cell dysplasia resulting in peripheral blood cytopenia. Microarray technology has permitted a refined high-throughput mapping of the transcriptional activity in the human genome. Non-coding RNAs (ncRNAs) transcribed from intronic regions of genes are involved in a number of processes related to post-transcriptional control of gene expression, and in the regulation of exon-skipping and intron retention. Characterization of ncRNAs in progenitor cells and stromal cells of MDS patients could be strategic for understanding gene expression regulation in this disease. METHODS In this study, gene expression profiles of CD34+ cells of 4 patients with MDS of refractory anemia with ringed sideroblasts (RARS) subgroup and stromal cells of 3 patients with MDS-RARS were compared with healthy individuals using 44 k combined intron-exon oligoarrays, which included probes for exons of protein-coding genes, and for non-coding RNAs transcribed from intronic regions in either the sense or antisense strands. Real-time RT-PCR was performed to confirm the expression levels of selected transcripts. RESULTS In CD34+ cells of MDS-RARS patients, 216 genes were significantly differentially expressed (q-value <or= 0.01) in comparison to healthy individuals, of which 65 (30%) were non-coding transcripts. In stromal cells of MDS-RARS, 12 genes were significantly differentially expressed (q-value <or= 0.05) in comparison to healthy individuals, of which 3 (25%) were non-coding transcripts. CONCLUSIONS These results demonstrated, for the first time, the differential ncRNA expression profile between MDS-RARS and healthy individuals, in CD34+ cells and stromal cells, suggesting that ncRNAs may play an important role during the development of myelodysplastic syndromes.
Collapse
Affiliation(s)
- Mariana O Baratti
- Department of Internal Medicine, School of Medical Science, Hematology and Hemotherapy Center, University of Campinas, 13083-970 Campinas, SP, Brazil
| | | | | | | | | | | |
Collapse
|
13
|
Vasikova A, Belickova M, Budinska E, Cermak J. A distinct expression of various gene subsets in CD34+ cells from patients with early and advanced myelodysplastic syndrome. Leuk Res 2010; 34:1566-72. [PMID: 20303173 DOI: 10.1016/j.leukres.2010.02.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Revised: 02/08/2010] [Accepted: 02/19/2010] [Indexed: 11/16/2022]
Abstract
Gene expression profiles of CD34+ cells were compared between 51 MDS patients and 7 controls. The most up-regulated genes in patients included HBG2, HBG1, CYBRD1, HSPA1B, ANGPT, and MYC, while 13 genes related to B-lymphopoiesis showed down-regulation. We observed in advanced MDS patients decreased expression of genes involved in cell cycle control, DNA repair and increased expression of proto-oncogenes, angiogenic and anti-apoptic genes. The results suggest that increased cell proliferation and resistance to apoptosis together with a loss of cell cycle control, damaged DNA repair and altered immune response may play an important role in malignant clone expansion in MDS.
Collapse
Affiliation(s)
- Alzbeta Vasikova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | | | | | | |
Collapse
|
14
|
Sridhar K, Ross DT, Tibshirani R, Butte AJ, Greenberg PL. Relationship of differential gene expression profiles in CD34+ myelodysplastic syndrome marrow cells to disease subtype and progression. Blood 2009; 114:4847-58. [PMID: 19801443 PMCID: PMC2786292 DOI: 10.1182/blood-2009-08-236422] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 09/13/2009] [Indexed: 01/12/2023] Open
Abstract
Microarray analysis with 40 000 cDNA gene chip arrays determined differential gene expression profiles (GEPs) in CD34(+) marrow cells from myelodysplastic syndrome (MDS) patients compared with healthy persons. Using focused bioinformatics analyses, we found 1175 genes significantly differentially expressed by MDS versus normal, requiring a minimum of 39 genes to separately classify these patients. Major GEP differences were demonstrated between healthy and MDS patients and between several MDS subgroups: (1) those whose disease remained stable and those who subsequently transformed (tMDS) to acute myeloid leukemia; (2) between del(5q) and other MDS patients. A 6-gene "poor risk" signature was defined, which was associated with acute myeloid leukemia transformation and provided additive prognostic information for International Prognostic Scoring System Intermediate-1 patients. Overexpression of genes generating ribosomal proteins and for other signaling pathways was demonstrated in the tMDS patients. Comparison of del(5q) with the remaining MDS patients showed 1924 differentially expressed genes, with underexpression of 1014 genes, 11 of which were within the 5q31-32 commonly deleted region. These data demonstrated (1) GEPs distinguishing MDS patients from healthy and between those with differing clinical outcomes (tMDS vs those whose disease remained stable) and cytogenetics [eg, del(5q)]; and (2) molecular criteria refining prognostic categorization and associated biologic processes in MDS.
Collapse
Affiliation(s)
- Kunju Sridhar
- Hematology Division,Stanford University Medical Center, 875 Blake Wilbur Drive, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
15
|
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | | |
Collapse
|
16
|
Benekli M, Baumann H, Wetzler M. Targeting signal transducer and activator of transcription signaling pathway in leukemias. J Clin Oncol 2009; 27:4422-32. [PMID: 19667270 DOI: 10.1200/jco.2008.21.3264] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Signal transducer and activator of transcription (STAT) proteins comprise a seven-member family of latent cytoplasmic transcription factors that are activated through tyrosine phosphorylation by a variety of cytokines and growth factors. Aberrant activation of STATs accompanies malignant cellular transformation with resultant leukemogenesis. Constitutive activation of STATs has been demonstrated in various leukemias. A better understanding of the mechanisms of dysregulation of the STAT pathway and understanding of the cause and effect relationship in leukemogenesis may serve as a basis for designing novel therapeutic strategies directed against STATs. Mechanisms of STAT activation, the potential role of STAT signaling in leukemogenesis, and recent advances in drug discovery targeting the STAT pathway are the focus of this review.
Collapse
Affiliation(s)
- Mustafa Benekli
- Department of Medical Oncology, Gazi University Faculty of Medicine, Ankara, Turkey
| | | | | |
Collapse
|
17
|
Abstract
Myelodysplastic syndrome (MDS) is an enigmatic disorder characterized by ineffective hematopoiesis and dysplastic morphology of blood cells. The clinical course of MDS consists of distinct stages, with early stages often progressing to advanced ones or to acute myeloid leukemia (AML). Little is known of the molecular pathogenesis of MDS or of the mechanism of its stage progression. DNA micro-array analysis, which allows simultaneous monitoring of the expression levels of tens of thousands of genes, has the potential to provide insight into the pathophysiology of MDS. Several studies have applied this new technology to compare gene expression profiles either between MDS and the healthy condition, among the different stages of MDS or between MDS-derived AML and de novo AML. Selection of an appropriate hematopoietic fraction is important for such studies, which to date have been performed with differentiated granulocytes, CD34+ progenitors and CD133+ immature cells. These studies have revealed that each stage of MDS has its own 'molecular signature', indicating the feasibility of differential diagnosis of MDS based on gene expression profile. They have also demonstrated that the current clinical diagnosis of MDS results in the misclassification of patients with regard to these molecular signatures.
Collapse
Affiliation(s)
- Hiroyuki Mano
- Division of Functional Genomics, Jichi Medical School, Kawachigun, Tochigi, Japan.
| |
Collapse
|
18
|
Clinical implications of gene expression profiling in myelodysplastic syndromes: Recognition of ribosomal and translational gene dysregulation and development of predictive assays. Best Pract Res Clin Haematol 2009; 22:223-37. [DOI: 10.1016/j.beha.2009.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
19
|
Ou JJ, Bagg A. Diagnostic challenges in the myelodysplastic syndromes: the current and future role of genetic and immunophenotypic studies. EXPERT OPINION ON MEDICAL DIAGNOSTICS 2009; 3:275-91. [PMID: 23488463 DOI: 10.1517/17530050902813947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Myelodysplastic syndromes (MDS) comprise a clinically and pathologically diverse collection of hematopoietic neoplasms, most commonly presenting with peripheral cytopenias typically in the context of bone marrow hypercellularity. Mechanistically, at least in the early phases of the disease, this apparently paradoxical picture is primarily due to ineffective hematopoiesis, which is accompanied by a variety of morphologic abnormalities in hematopoietic cells. The identification of recurrent, clinically relevant cytogenetic defects in MDS has spurred the research of molecular mechanisms that contribute to its inception as well as to the development of heterogeneous subtypes. Although conventional cytogenetic analyses remain a diagnostic mainstay in MDS, the application of contemporary techniques including molecular cytogenetics, microarray technologies and multiparametric flow cytometry may ultimately reveal new diagnostic parameters that are theoretically more objective and sensitive than current morphologic approaches. This review aims to outline the role of genetic and immunophenotypic studies in the evaluation of MDS, including findings that may potentially influence future diagnostic classifications, which could refine prognostication and ultimately facilitate the growth of targeted therapies.
Collapse
Affiliation(s)
- Joyce J Ou
- University of Pennsylvania, Department of Pathology and Laboratory Medicine, 3400 Spruce Street, 6 Founders Pavilion, PA 19406-4283, USA
| | | |
Collapse
|
20
|
Differential gene expression of bone marrow-derived CD34+ cells is associated with survival of patients suffering from myelodysplastic syndrome. Int J Hematol 2009; 89:173-187. [DOI: 10.1007/s12185-008-0242-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 11/30/2008] [Accepted: 12/04/2008] [Indexed: 01/24/2023]
|
21
|
Kracmarova A, Cermak J, Brdicka R, Bruchova H. High expression of ERCC1, FLT1, NME4 and PCNA associated with poor prognosis and advanced stages in myelodysplastic syndrome. Leuk Lymphoma 2008; 49:1297-305. [PMID: 18604718 DOI: 10.1080/10428190802129918] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Myelodysplastic syndrome (MDS) represents a good model for research of prognostic/progression markers due to frequent transformation into acute myeloid leukemia (AML). We analysed expression profiles of 26 MDS and 6 AML patients using cDNA arrays comprising 588 gene probes. The array data were validated in a larger set of 46 patients by qRT-PCR. Data analysis identified differently expressed genes in MDS and the cluster of four genes (ERCC1, FLT1, NME4 and PCNA) whose expression was correlated with MDS subtypes. High expression of these genes was associated with poor prognosis and/or unfavorable outcome. Furthermore, PCNA expression was correlated with peripheral blood blast percentage (r = 0.71, p < 0.05), while the other genes showed non-significant correlation. Our findings demonstrate the progressive up-regulation of the genes along the sequence of 5q-syndrome/RCMD/RAEB/de novo AML, suggesting their association with disease progression.
Collapse
Affiliation(s)
- Alzbeta Kracmarova
- Department of Molecular Genetics, Institute of Hematology and Blood Transfusion, U Nemocnice 1, Prague, Czech Republic
| | | | | | | |
Collapse
|
22
|
Cortelezzi A, Colombo G, Pellegrini C, Silvestris I, Moronetti Mazzeo L, Bosari S, Lambertenghi Deliliers G, Fracchiolla NS. Bone marrow glycophorin-positive erythroid cells of myelodysplastic patients responding to high-dose rHuEPO therapy have a different gene expression pattern from those of nonresponders. Am J Hematol 2008; 83:531-9. [PMID: 18383321 DOI: 10.1002/ajh.21178] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The main clinical problems of low-risk patients with myelodysplastic syndromes (MDS), as defined by the International Prognostic Scoring System, are infections and the need for frequent transfusions due to ineffective myelopoiesis and peripheral blood cytopenia. Promising results in treating MDS-related anemia have been obtained using high-dose recombinant human erythropoietin (rhEPO). To evaluate the molecular basis of the response to rhEPO, we used commercially available macro-arrays to investigate gene expression profiles in the glycophorin-expressing (Gly+) bone marrow (BM) erythroid cells of five responders (ERs) and five non-responders (ENRs) to rhEPO treatment. The cells were separated by means of positive selection using an immunomagnetic procedure, after which flow cytometry showed that their purity was more than 97% in all cases. The array data were validated by means of real time RT-PCR. The results showed that the genes responsible for proliferation/differentiation and DNA repair/stability were repressed in the BM Gly+ erythroid cells of the ENRs, but almost normally expressed in the ERs. Furthermore, the expression of genes involved in signal transduction suggested that the activity of the MAPK signaling pathway is inhibited in ERs. The different gene expression profiles of ERs and ENRs may provide a basis for early gene testing as a means of predicting the response to rhEPO of MDS patients with low endogenous EPO levels.
Collapse
Affiliation(s)
- Agostino Cortelezzi
- Hematology-Bone Marrow Transplant Unit, Fondazione Ospedale Maggiore Maggiore Policlinico, Mangiagalli, Regina Elena IRCCS, Milan.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Bernimoulin M, Stern M, Tichelli A, Jotterand M, Gratwohl A, Nissen C. Leukemic cluster growth in culture is an independent risk factor for acute myeloid leukemia and short survival in patients with myelodysplastic syndrome. Acta Haematol 2008; 119:226-35. [PMID: 18566541 DOI: 10.1159/000140675] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2008] [Accepted: 04/11/2008] [Indexed: 11/19/2022]
Abstract
In patients with myelodysplastic syndrome (MDS) precursor cell cultures (colony-forming unit cells, CFU-C) can provide an insight into the growth potential of malignant myeloid cells. In a retrospective single-center study of 73 untreated MDS patients we assessed whether CFU-C growth patterns were of prognostic value in addition to established criteria. Abnormalities were classified as qualitative (i.e. leukemic cluster growth) or quantitative (i.e. strongly reduced/absent growth). Thirty-nine patients (53%) showed leukemic growth, 26 patients (36%) had strongly reduced/absent colony growth, and 12 patients showed both. In a univariate analysis the presence of leukemic growth was associated with strongly reduced survival (at 10 years 4 vs. 34%, p = 0.004), and a high incidence of transformation to AML (76 vs. 32%, p = 0.01). Multivariate analysis identified leukemic growth as a strong and independent predictor of early death (relative risk 2.12, p = 0.03) and transformation to AML (relative risk 2.63, p = 0.04). Quantitative abnormalities had no significant impact on the disease course. CFU-C assays have a significant predictive value in addition to established prognostic factors in MDS. Leukemic growth identifies a subpopulation of MDS patients with poor prognosis.
Collapse
|
24
|
Iwasaki T, Murakami M, Sugisaki C, Sobue S, Ohashi H, Asano H, Suzuki M, Nakamura S, Ito M, Murate T. Characterization of myelodysplastic syndrome and aplastic anemia by immunostaining of p53 and hemoglobin F and karyotype analysis: Differential diagnosis between refractory anemia and aplastic anemia. Pathol Int 2008; 58:353-60. [DOI: 10.1111/j.1440-1827.2008.02236.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
25
|
Raponi M, Harousseau JL. Development of genomic markers that predict response to molecularly targeted antileukemic therapy. EXPERT OPINION ON MEDICAL DIAGNOSTICS 2008; 2:361-72. [PMID: 23495705 DOI: 10.1517/17530059.2.4.361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The cancer genome is characterized by the accumulation of multiple mutations and alterations that ultimately result in the deregulation of various cell-signaling pathways. Knowledge of these genetic alterations has provided a unique opportunity to develop therapies targeted against these pathways and to identify which patients are likely to benefit from them. OBJECTIVE The progress that has been made in identifying genomic biomarkers that can predict response to antileukemic therapies is highlighted. METHODS Global gene expression profiling approaches utilizing tipifarnib in acute myeloid leukemia as an in-depth example are focused on. The challenges in developing associated theranostic molecular assays are discussed. CONCLUSION The integration of validated genomic-based assays with common morphological tests may allow for improved prediction of antileukemic drug response.
Collapse
Affiliation(s)
- Mitch Raponi
- Veridex, LLC, a Johnson & Johnson Company, 3210 Merryfield Row, San Diego, CA 92121, USA +1 858 320 3319 ; +1 858 784 3182 ;
| | | |
Collapse
|
26
|
Spatial interplay between PIASy and FIP200 in the regulation of signal transduction and transcriptional activity. Mol Cell Biol 2008; 28:2771-81. [PMID: 18285457 DOI: 10.1128/mcb.01210-07] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The members of the protein inhibitor of activated STAT (PIAS) family of proteins are implicated in fundamental cellular processes, including transcriptional regulation, either through action as E3 SUMO ligases or through SUMO-independent effects. We report here the identification of FIP200 (focal adhesion kinase family-interacting protein of 200 kDa) as a new PIASy-interacting protein. We show that the interaction depends on the integrity of the RING finger of PIASy and the carboxy terminus of FIP200. Both in vitro and in vivo sumoylation assays failed to reveal any sumoylation of FIP200, suggesting that FIP200 is not a bona fide SUMO substrate. Immunofluorescence microscopy and subcellular fractionation, either upon forced PIASy expression or in the absence of PIASy, revealed that interaction with PIASy redistributes FIP200 from the cytoplasm to the nucleus, correlating with abrogation of FIP200 regulation of TSC/S6K signaling. Conversely, FIP200 enhances the transcriptional activation of the p21 promoter by PIASy whereas PIASy transcription activity is severely reduced upon FIP200 depletion by RNA interference. Chromatin immunoprecipitation analysis demonstrates that endogenous PIASy and FIP200 are corecruited to the p21 promoter. Altogether, these results provide the first evidence for the existence of a close-spatially controlled-mode of regulation of FIP200 and PIASy nucleocytoplasmic functions.
Collapse
|
27
|
Ghoshal Gupta S, Baumann H, Wetzler M. Epigenetic regulation of signal transducer and activator of transcription 3 in acute myeloid leukemia. Leuk Res 2008; 32:1005-14. [PMID: 18192010 DOI: 10.1016/j.leukres.2007.11.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Revised: 11/25/2007] [Accepted: 11/26/2007] [Indexed: 12/22/2022]
Abstract
We have demonstrated that constitutive signal transducer and activator of transcription (STAT) 3 activity, observed in approximately 50% of acute myeloid leukemia (AML) cases, is associated with adverse treatment outcome. Constitutive STAT3 activation may result from the expression of oncogenic protein tyrosine kinases or from autocrine stimulation by hematopoietic growth factors. These causes are generally neither necessary nor sufficient for leukemogenesis; additional transforming events or growth stimulatory processes are needed. Here we review the literature addressing epigenetic regulation as a mechanism controlling STAT3 signaling in AML. A better understanding of mechanisms of dysregulation of STAT signaling pathways may serve as a basis for designing novel therapeutic strategies that target these pathways in leukemia cells.
Collapse
Affiliation(s)
- Sampa Ghoshal Gupta
- Department of Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | | | | |
Collapse
|
28
|
Rujkijyanont P, Beyene J, Wei K, Khan F, Dror Y. Leukaemia-related gene expression in bone marrow cells from patients with the preleukaemic disorder Shwachman?Diamond syndrome. Br J Haematol 2007; 137:537-44. [PMID: 17539775 DOI: 10.1111/j.1365-2141.2007.06608.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Shwachman-Diamond syndrome (SDS) is an inherited bone marrow failure disorder with cytopenia and a high propensity for myelodysplastic syndrome (MDS) and leukaemia, particularly acute myeloid leukaemia. The mechanism of leukaemogenesis in SDS is unknown. In accordance to the multi-hit theory of carcinogenesis, it is likely that several molecular and cellular hits occur before MDS/leukaemia become apparent. This study used oligonucleotide microarray to identify gene expression patterns, which were shown to be associated with leukaemogenesis, in marrow mononuclear cells of nine SDS patients without overt transformation compared to healthy controls. Among 154 known leukaemia-related genes, several oncogenes were found to be upregulated, including LARG, TAL1 and MLL, and of several tumour suppressor genes were downregulated, including DLEU1, RUNX1, FANCD2 and DKC1. Real time polymerase chain reaction confirmed statistically higher expression of LARG and TAL1 in SDS marrows. We conclude that SDS marrow mononuclear cells exhibit abnormal gene expression patterns, which might result in continuous stimulation favouring evolution or progression of malignant clones. Additional molecular and cytogenetic events are probably necessary for the malignant process to be irreversible and complete.
Collapse
Affiliation(s)
- Piya Rujkijyanont
- Marrow Failure and Myelodysplasia Program, Division of Haematology/Oncology, Department of Paediatrics, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
29
|
Roela RA, Carraro DM, Brentani HP, Kaiano JHL, Simão DF, Guarnieiro R, Lopes LF, Borojevic R, Brentani MM. Gene stage-specific expression in the microenvironment of pediatric myelodysplastic syndromes. Leuk Res 2006; 31:579-89. [PMID: 17092558 DOI: 10.1016/j.leukres.2006.10.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2006] [Revised: 09/11/2006] [Accepted: 10/10/2006] [Indexed: 01/19/2023]
Abstract
Using cDNA microarray assays we have observed a clear difference in the gene expression pattern between bone marrow stromal cells obtained from healthy children (CT) and from pediatric patients with either myelodysplastic syndromes (MDS) or acute myeloid leukemia (AML) associated with MDS (MDS-AML). The global gene function profiling analysis indicated that in the pediatric MDS microenvironment the disease stages may be characterized mainly by underexpression of genes associated with biological processes such as transport. Furthermore, a subset of downregulated genes related to endocytosis and protein secretion was able to discriminate MDS from MDS-AML.
Collapse
Affiliation(s)
- Rosimeire A Roela
- Faculdade de Medicina da Universidade de São Paulo, Departamento de Radiologia (LIM24), Av Dr Arnaldo 455 sala 4112, São Paulo 01246-903, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Mahadevan D, DiMento J, Croce KD, Riley C, George B, Fuchs D, Mathews T, Wilson C, Lobell M. Transcriptosome and serum cytokine profiling of an atypical case of myelodysplastic syndrome with progression to acute myelogenous leukemia. Am J Hematol 2006; 81:779-86. [PMID: 16838325 DOI: 10.1002/ajh.20690] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
A Native American-Indian female presenting with anemia and thrombocytosis was diagnosed with myelodysplastic syndrome (MDS, refractory anemia). Over the course of 5 years she developed cytopenias and periods of leukocytosis with normal bone marrow (BM) blast counts, features of an unclassifiable MDS/MPS syndrome. The patient ultimately progressed to acute myelogenous leukemia (AML, FAB M2) and had a normal karyotype throughout her course. The episodes of leukocytosis were associated with infectious complications. Transformation to AML was characterized by a BM blast percentage of 49%. Peripheral blood and BM samples were obtained for serum protein analysis and gene expression profiling (GEP) to elucidate her disease process. An ELISA assay of the serum analyzed approximately 80 cytokines, which demonstrated that hepatocyte growth factor/scatter factor and insulin-like growth factor binding protein 1 were markedly elevated compared to normal. GEP demonstrated a unique "tumor molecular profile," which included overexpression of oncogenes (HOXA9, N-MYC, KOC1), proliferative genes (PAWR, DLG5, AKR1C3), invasion/metastatic genes (FN1, N-CAM-1, ITGB5), pro-angiogenesis genes (c-Kit), and down regulation of tumor suppressor genes (SUI1, BARD1) and anti-apoptotic genes (PGLYRP, SERPINB2, MPO). Hence, a biomics approach has provided insight into elucidating disease mechanisms, molecular prognostic factors, and discovery of novel targets for therapeutic intervention.
Collapse
MESH Headings
- Aged
- Biomarkers, Tumor/blood
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
- Cytokines/blood
- Disease Progression
- Enzyme-Linked Immunosorbent Assay/methods
- Fatal Outcome
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic/genetics
- Hepatocyte Growth Factor/blood
- Humans
- Insulin-Like Growth Factor Binding Protein 1/blood
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Myelodysplastic Syndromes/blood
- Myelodysplastic Syndromes/diagnosis
- Myelodysplastic Syndromes/genetics
- Oligonucleotide Array Sequence Analysis/methods
- Reverse Transcriptase Polymerase Chain Reaction/methods
Collapse
Affiliation(s)
- Daruka Mahadevan
- University of Arizona Cancer Center, Tucson, Arizona 85724, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Ubiquitin and ubiquitin-like proteins (Ubls) are signalling messengers that control many cellular functions, such as cell proliferation, apoptosis, the cell cycle and DNA repair. It is becoming apparent that the deregulation of ubiquitin pathways results in the development of human diseases, including many types of tumours. Here we summarize the common principles and specific features of ubiquitin and Ubls in the regulation of cancer-relevant pathways, and discuss new strategies to target ubiquitin signalling in drug discovery.
Collapse
Affiliation(s)
- Daniela Hoeller
- Institute of Biochemistry II, Goethe University School of Medicine, University Hospital, Building 75, Theodour-Stern-Kai 7, D-60590 Frankfurt, Germany
| | | | | |
Collapse
|
32
|
Abstract
The production of blood cells is regulated by a number of protein growth factors and cytokines that influence cell survival, proliferation and differentiation. Many of these molecules bind to cell surface receptors, which belong to a family of closely related cytokine receptors that lack intrinsic catalytic activity but are intimately associated with tyrosine kinases of the Janus kinase (JAK) family. Ligand binding induces the activation of JAKs, which sit at the apex of a signalling cascade in which a key role is played by members of the signal transducers and activators of transcription (STAT) group. Congenital deficiencies in JAK-STAT signalling are associated with immunodeficiency states and acquired activating mutations and translocations are involved in the pathophysiology of haematological malignancy. The latter findings have raised hopes that drugs that target aberrant JAK-STAT signalling may be useful for the treatment of human disease.
Collapse
Affiliation(s)
- Asim Khwaja
- Department of Haematology, Royal Free and University College Medical School, London, UK.
| |
Collapse
|
33
|
Vawter MP, Tomita H, Meng F, Bolstad B, Li J, Evans S, Choudary P, Atz M, Shao L, Neal C, Walsh DM, Burmeister M, Speed T, Myers R, Jones EG, Watson SJ, Akil H, Bunney WE. Mitochondrial-related gene expression changes are sensitive to agonal-pH state: implications for brain disorders. Mol Psychiatry 2006; 11:615, 663-79. [PMID: 16636682 PMCID: PMC3098558 DOI: 10.1038/sj.mp.4001830] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mitochondrial defects in gene expression have been implicated in the pathophysiology of bipolar disorder and schizophrenia. We have now contrasted control brains with low pH versus high pH and showed that 28% of genes in mitochondrial-related pathways meet criteria for differential expression. A majority of genes in the mitochondrial, chaperone and proteasome pathways of nuclear DNA-encoded gene expression were decreased with decreased brain pH, whereas a majority of genes in the apoptotic and reactive oxygen stress pathways showed an increased gene expression with a decreased brain pH. There was a significant increase in mitochondrial DNA copy number and mitochondrial DNA gene expression with increased agonal duration. To minimize effects of agonal-pH state on mood disorder comparisons, two classic approaches were used, removing all subjects with low pH and agonal factors from analysis, or grouping low and high pH as a separate variable. Three groups of potential candidate genes emerged that may be mood disorder related: (a) genes that showed no sensitivity to pH but were differentially expressed in bipolar disorder or major depressive disorder; (b) genes that were altered by agonal-pH in one direction but altered in mood disorder in the opposite direction to agonal-pH and (c) genes with agonal-pH sensitivity that displayed the same direction of changes in mood disorder. Genes from these categories such as NR4A1 and HSPA2 were confirmed with Q-PCR. The interpretation of postmortem brain studies involving broad mitochondrial gene expression and related pathway alterations must be monitored against the strong effect of agonal-pH state. Genes with the least sensitivity to agonal-pH could present a starting point for candidate gene search in neuropsychiatric disorders.
Collapse
Affiliation(s)
- M P Vawter
- Department of Psychiatry, University of California, Irvine, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Albor A, El-Hizawi S, Horn EJ, Laederich M, Frosk P, Wrogemann K, Kulesz-Martin M. The interaction of Piasy with Trim32, an E3-ubiquitin ligase mutated in limb-girdle muscular dystrophy type 2H, promotes Piasy degradation and regulates UVB-induced keratinocyte apoptosis through NFkappaB. J Biol Chem 2006; 281:25850-66. [PMID: 16816390 DOI: 10.1074/jbc.m601655200] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Protein inhibitors of activated STATs (PIAS) family members are ubiquitin-protein isopeptide ligase-small ubiquitin-like modifier ligases for diverse transcription factors. However, the regulation of PIAS protein activity in cells is poorly understood. Previously, we reported that expression of Trim32, a RING domain ubiquitin-protein isopeptide ligase-ubiquitin ligase mutated in human limb-girdle muscular dystrophy type 2H (LGMD2H) and Bardet-Biedl syndrome, is elevated during mouse skin carcinogenesis, protecting keratinocytes from apoptosis induced by UVB and tumor necrosis factor-alpha (TNFalpha). Here we report that Trim32 interacts with Piasy and promotes Piasy ubiquitination and degradation. Ubiquitination of Piasy by Trim32 could be reproduced in vitro using purified components. Their interaction was induced by treatment with UVB/TNFalpha and involved redistribution of Piasy from the nucleus to the cytoplasm, where it accumulated in cytoplasmic granules that colocalized with Trim32. Piasy destabilization and ubiquitination required an intact RING domain in Trim32. The LGMD2H-associated missense point mutation prevented Trim32 binding to Piasy, and human Piasy failed to colocalize with human Trim32 in fibroblasts isolated from an LGMD2H patient. Trim32 expression increased the transcriptional activity of NFkappaB in epidermal keratinocytes, both under basal treatment and after UVB/TNFalpha treatment. Conversely, Piasy inhibited NFkappaB activity under the same conditions and sensitized keratinocytes to apoptosis induced by TNFalpha and UVB. Our results indicate that, by controlling Piasy stability, Trim32 regulates UVB-induced keratinocyte apoptosis through induction of NFkappaB and suggests loss of function of Trim32 in LGMD2H.
Collapse
Affiliation(s)
- Amador Albor
- Department of Dermatology and Program in Cell and Molecular Biology, Oregon Health and Science University, Portland, Oregon 97239, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Bennett JM, Komrokji RS. The myelodysplastic syndromes: diagnosis, molecular biology and risk assessment. ACTA ACUST UNITED AC 2006; 10 Suppl 1:258-69. [PMID: 16188686 DOI: 10.1080/10245330512331390311] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Myelodysplastic syndromes (MDS) are heterogeneous group of neoplastic clonal stem cell diseases characterized by dysplastic morphological features and clinical bone marrow failure. The FAB (French-American-British) system served as the gold standard for MDS classification for more than two decades. The WHO classification, built on the backbone of FAB classification, is an attempt to further improve the prognostic value of MDS classification as well as establish its clinical utility as a tool to select different treatments. In this article we review the epidemiology, pathogenesis, molecular biology, diagnosis and classification of MDS. We highlight the major differences between the FAB classification and the WHO MDS classification. We discuss in more detail the experience of using the new WHO classification since its publication and review the studies that tried to validate the prognostic value of the new classification or apply it to predict clinical responses to various treatments.
Collapse
Affiliation(s)
- John M Bennett
- Department of Medicine and the James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA.
| | | |
Collapse
|
36
|
Valentino L, Pierre J. JAK/STAT signal transduction: regulators and implication in hematological malignancies. Biochem Pharmacol 2006; 71:713-21. [PMID: 16426581 DOI: 10.1016/j.bcp.2005.12.017] [Citation(s) in RCA: 194] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Revised: 12/07/2005] [Accepted: 12/09/2005] [Indexed: 01/12/2023]
Abstract
Signal transducers and activators of transcription (STATs) comprise a family of several transcription factors that are activated by a variety of cytokines, hormones and growth factors. STATs are activated through tyrosine phosphorylation, mainly by JAK kinases, which lead to their dimerization, nuclear translocation and regulation of target genes expression. Stringent mechanisms of signal attenuation are essential for insuring appropriate, controlled cellular responses. Among them phosphotyrosine phosphatases (SHPs, CD45, PTP1B/TC-PTP), protein inhibitors of activated STATs (PIAS) and suppressors of cytokine signaling (SOCS) inhibit specific and distinct aspects of cytokine signal transduction. SOCS proteins bind through their SH2 domain to phosphotyrosine residues in either cytokine receptors or JAK and thus can suppress cytokine signaling. Many recent findings indicate that SOCS proteins act, in addition, as adaptors that regulate the turnover of certain substrates by interacting with and activating an E3 ubiquitin ligase. Thus, SOCS proteins act as negative regulators of JAK/STAT pathways and may represent tumour suppressor genes. The discovery of oncogenic partner in this signaling pathway, more especially in diverse hematologic malignancies support a prominent role of deregulated pathways in the pathogenesis of diseases. Fusion proteins implicating the JH1 domain of JAK2 (TEL-JAK2, BCR-JAK2), leading to deregulated activity of JAK2, have been described as the result of translocation. Somatic point mutation in JH2 domain of JAK2 (JAK2V617F), leading also to constitutive tyrosine phosphorylation of JAK2 and its downstream effectors was reported in myeloproliferative disorders. Furthermore, silencing of socs-1 and shp-1 expression by gene methylation is observed in some cancer cells.
Collapse
Affiliation(s)
- Lyne Valentino
- Inserm U749, Faculté de Pharmacie, 5, rue JB Clément, 92296 Châtenay-Malabry, France
| | | |
Collapse
|
37
|
Nishino HT, Chang CC. Myelodysplastic syndromes: clinicopathologic features, pathobiology, and molecular pathogenesis. Arch Pathol Lab Med 2006; 129:1299-310. [PMID: 16196520 DOI: 10.5858/2005-129-1299-mscfpa] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT Myelodysplastic syndromes (MDSs) are clonal stem cell diseases characterized by ineffective hematopoiesis, multilineage dysplasia, and peripheral cytopenias with normocellular or hypercellular marrow. They represent a heterogeneous group of disorders with a varied spectrum of clinical, morphologic, biologic, and genetic characteristics. This heterogeneity in disease characterization has led to evolving classification systems, developing prognostic models, and continuing research efforts to elucidate its pathobiology and pathogenesis. OBJECTIVE To summarize updated information and provide a general overview of the clinicopathologic features, pathobiology, and cytogenetic and molecular pathogenesis of MDSs. DATA SOURCES Relevant articles indexed in PubMed (National Library of Medicine) between 1982 and 2005 and reference medical texts. CONCLUSIONS Although MDSs remain a relatively poorly defined disease entity, recent advancements in cytogenetic and molecular studies have significantly contributed to our present knowledge of MDSs. Novel strategies for studying the pathogenesis and evolution of MDSs continue to shape our understanding of this disease and guide our approaches to diagnosis and treatment.
Collapse
Affiliation(s)
- Ha Thanh Nishino
- Department of Pathology, The Methodist Hospital, Houston, TX 77030, USA
| | | |
Collapse
|
38
|
Steensma DP, List AF. Genetic testing in the myelodysplastic syndromes: molecular insights into hematologic diversity. Mayo Clin Proc 2005; 80:681-98. [PMID: 15887439 DOI: 10.4065/80.5.681] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The myelodysplastic syndromes (MDS) are associated with a diverse set of acquired somatic genetic abnormalities. Bone marrow karyotyping provides important diagnostic and prognostic information and should be attempted in all patients who are suspected of having MDS. Fluorescent in situ hybridization (FISH) studies on blood or marrow may also be valuable in selected cases, such as patients who may have 5q- syndrome or those who have undergone hematopoletic stem cell transplantation. The MDS-associated cytogenetic abnormalities that have been defined by karyotyping and FISH studies have already contributed substantially to our current understanding of the biology of malignant myeloid disorders, but the pathobiological meaning of common, recurrent chromosomal lesions such as del(5q), del(20q), and monosomy 7 is still unknown. The great diversity of the cytogenetic findings described in MDS highlights the molecular heterogeneity of this cluster of diseases. We review the common and pathophysiologically interesting genetic abnormalities associated with MDS, focusing on the clinical utility of conventional cytogenetic assays and selected FISH studies. In addition, we discuss a series of well-defined MDS-associated point mutations and outline the potential for further insights from newer techniques such as global gene expression profiling and array-based comparative genomic hybridization.
Collapse
Affiliation(s)
- David P Steensma
- Department of Internal Medicine and Division of Hematology, Mayo Clinic College of Medicine, Rochester, Minn 55905, USA
| | | |
Collapse
|
39
|
Abstract
During the past 15 years, important progress has been made in the understanding of the biology and prognosis of myelodysplastic syndrome (MDS). MDS is a clonal disorder characterized by ineffective hematopoiesis, which can lead to either fatal cytopenias or acute myelogenous leukemia (AML). Risk-adapted treatment strategies were established because of the high median age (60-75 years) of the MDS patients and the individual history of the disease (number of cytopenias, cytogenetic changes, transfusion requirements). Allogeneic bone marrow transplantation currently offers the only potentially curative treatment, but this form of therapy is not available for the typical MDS patient, who is >60 years of age. Therapy with erythropoietin and G-CSF has improved the quality of life of selected patients. The development of small molecules directed against specific molecular targets with minimal adverse effects is the hope for the future. Innovative uses of immunomodulatory agents and the optimizing of cytotoxic treatment should continue to help in the treatment of MDS.
Collapse
Affiliation(s)
- Wolf-K Hofmann
- Department of Hematology and Oncology and Transfusion Medicine, University Hospital Benjamin Franklin, 12200 Berlin, Germany.
| | | |
Collapse
|
40
|
Abstract
Lymphoid tumours comprise the acute and chronic leukaemias, the broad spectrum of lymphomas, including Hodgkin's disease, and multiple myeloma. The subdivision of the acute leukaemias according to the proliferating type of white blood cells has had a major impact on the care of these patients. More recently, specific chromosomal translocations have been used to identify patients who may benefit from more intensive therapies. The novel high-throughput genomic technologies, such as microarrays, provide new avenues for the molecular diagnosis of the haematological malignancies. Rapid advances in genome sequencing and gene expression profiling provide unprecedented opportunities to identify specific genes involved in complex biological processes, including tumorigenesis. The features of microarray technology and the variety of experimental approaches to elucidate lymphoid malignancies are discussed. Microarray technology has the potential to lead to more accurate prognostic assessment for patients and is expected to ultimately allow the clinician to select therapies optimally suited to each patient.
Collapse
Affiliation(s)
- Ursula R Kees
- Division of Children's Leukaemia and Cancer Research, Telethon Institute for Child Health Research and Centre for Child Health Research, The University of Western Australia, Perth, Australia.
| |
Collapse
|
41
|
Tsutsumi C, Ueda M, Miyazaki Y, Yamashita Y, Choi YL, Ota J, Kaneda R, Koinuma K, Fujiwara SI, Kisanuki H, Ishikawa M, Ozawa K, Tomonaga M, Mano H. DNA microarray analysis of dysplastic morphology associated with acute myeloid leukemia. Exp Hematol 2004; 32:828-35. [PMID: 15345284 DOI: 10.1016/j.exphem.2004.06.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2004] [Revised: 05/17/2004] [Accepted: 06/05/2004] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Acute myeloid leukemia (AML) develops de novo or secondarily to either myelodysplastic syndrome (MDS) or anticancer treatment (therapy-related leukemia, TRL). Prominent dysplasia of blood cells is apparent in individuals with MDS-related AML as well as in some patients with TRL or even with de novo AML. The clinical entity of AML with multilineage dysplasia (AML-MLD) is likely to be an amalgamation of MDS-related AML and de novo AML-MLD. The aim of this study was to clarify, by the use of high-density oligonucleotide microarrays, whether these subcategories of AML are intrinsically distinct from each other. MATERIALS AND METHODS The AC133+ hematopoietic stem cell-like fractions were purified from the bone marrow of individuals with de novo AML without dysplasia (n = 15), AML-MLD (n = 11), MDS-related AML (n = 11), or TRL (n = 2), and were subjected to the synthesis of cRNA which was subsequently hybridized to microarray harboring oligonucleotide corresponding to more than 12,000 probe sets. RESULTS We could identify many genes whose expression was specific to these various subcategories of AML. Furthermore, with the correspondence analysis/three-dimensional projection strategy, we were able to visualize the independent, yet partially overlapping, nature of current AML subcategories on the basis of their transcriptomes. CONCLUSION Our data indicate the possibility of subclassification of AML based on gene expression profiles of leukemic blasts.
Collapse
Affiliation(s)
- Chizuko Tsutsumi
- Department of Hematology and Molecular Medicine Unit, Nagasaki University, Nagasaki, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Chen G, Zeng W, Miyazato A, Billings E, Maciejewski JP, Kajigaya S, Sloand EM, Young NS. Distinctive gene expression profiles of CD34 cells from patients with myelodysplastic syndrome characterized by specific chromosomal abnormalities. Blood 2004; 104:4210-8. [PMID: 15315976 DOI: 10.1182/blood-2004-01-0103] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aneuploidy, especially monosomy 7 and trisomy 8, is a frequent cytogenetic abnormality in the myelodysplastic syndromes (MDSs). Patients with monosomy 7 and trisomy 8 have distinctly different clinical courses, responses to therapy, and survival probabilities. To determine disease-specific molecular characteristics, we analyzed the gene expression pattern in purified CD34 hematopoietic progenitor cells obtained from MDS patients with monosomy 7 and trisomy 8 using Affymetrix GeneChips. Two methods were employed: standard hybridization and a small-sample RNA amplification protocol for the limited amounts of RNA available from individual cases; results were comparable between these 2 techniques. Microarray data were confirmed by gene amplification and flow cytometry using individual patient samples. Genes related to hematopoietic progenitor cell proliferation and blood cell function were dysregulated in CD34 cells of both monosomy 7 and trisomy 8 MDS. In trisomy 8, up-regulated genes were primarily involved in immune and inflammatory responses, and down-regulated genes have been implicated in apoptosis inhibition. CD34 cells in monosomy 7 showed up-regulation of genes inducing leukemia transformation and tumorigenesis and apoptosis and down-regulation of genes controlling cell growth and differentiation. These results imply distinct molecular mechanisms for monosomy 7 and trisomy 8 MDS and implicate specific pathogenic pathways.
Collapse
Affiliation(s)
- Guibin Chen
- Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
The mechanisms underlying hematopoietic stem cell or progenitor cell abnormalities in myelodysplastic syndromes (MDSs) remain poorly characterized. Current evidence exists for multiple intrinsic and extrinsic influences upon the stem cell in these disorders. These influences are outlined in this review and include: stem cell characteristics in MDSs, as compared with those in acute myelogenous leukemia; the role of increased apoptosis; the role of signaling pathway abnormalities; the influences of immune modulation; and the effect of stromal cells and stromal cell cytokine production. Despite numerous studies that have examined these factors, how they converge to produce a situation in which accelerated proliferation and accelerated death occur simultaneously remains largely an unexplored area. It is anticipated that future studies that focus on well-characterized and purified progenitor populations in these disorders will elucidate the process by which ineffective hematopoiesis results from the influences of stem cell abnormalities versus abnormalities in the stem cell's microenvironmental and immunologic milieu.
Collapse
Affiliation(s)
- Jane L Liesveld
- Leukemia/Blood and Marrow Transplant Program, Rochester, NY, USA.
| | | | | |
Collapse
|