Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.630
Filter
1.
Clin Lab Med ; 44(3): 465-477, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39089752

ABSTRACT

Multiparameter flow cytometry (MPF) is an essential component of the diagnostic workup of hematologic malignancies. Recently developed tools have expanded the utility of MPF in detecting T-cell clonality and myelomonocytic dysplasia. Minimal/measurable residual disease analysis has long been established as critical in the management of B-lymphoblastic leukemia and is emerging as a useful tool in myeloid malignancies. With the continued increased complexity of MPF assays, emerging tools for data collection and analysis will allow users to take full advantage of MPF in the diagnosis of hematologic disease.


Subject(s)
Flow Cytometry , Hematologic Neoplasms , Humans , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/pathology , Immunophenotyping , Neoplasm, Residual/diagnosis
2.
J Med Chem ; 67(16): 14016-14039, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39089850

ABSTRACT

HDAC8 can mediate signals by using its enzymatic or nonenzymatic functions, which are expected to be critical for various types of cancer. Herein, we employed proteolysis targeting chimera (PROTAC) technology to target the enzymatic as well as the nonenzymatic functions of HDAC8. A potent and selective HDAC8 PROTAC Z16 (CZH-726) with low nanomolar DC50 values in various cell lines was identified. Interestingly, Z16 induced structural maintenance of chromosomes protein 3 (SMC3) hyperacetylation at low concentrations and histone hyperacetylation at high concentrations, which can be explained by HDAC8 degradation and off-target HDAC inhibition, respectively. Notably, Z16 potently inhibited proliferation of various cancer cell lines and the antiproliferative mechanisms proved to be cell-type-dependent, which, to a large extent, is due to off-target HDAC inhibition. In conclusion, we report a hydrazide-based HDAC8 PROTAC Z16, which can be used as a probe to investigate the biological functions of HDAC8.


Subject(s)
Cell Proliferation , Histone Deacetylase Inhibitors , Histone Deacetylases , Hydrazines , Proteolysis , Repressor Proteins , Humans , Histone Deacetylases/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/chemical synthesis , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Proteolysis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Hydrazines/pharmacology , Hydrazines/chemistry , Hydrazines/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/pathology , Neoplasms/drug therapy , Neoplasms/pathology , Structure-Activity Relationship , Drug Screening Assays, Antitumor , Proteolysis Targeting Chimera
3.
Biomed Pharmacother ; 178: 117281, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39137651

ABSTRACT

Extensive research has shown that PR domain 16 (PRDM16) plays a critical role in adipose tissue metabolism, including processes such as browning and thermogenesis of adipocytes, beigeing of adipocytes, and adipogenic differentiation of myoblasts. These functions have been associated with diseases such as obesity and diabetes. Additionally, PRDM16 has been correlated with various other conditions, including migraines, heterochromatin abnormalities, metabolic syndrome, cardiomyopathy, sarcopenia, nonsyndromic cleft lip, and essential hypertension, among others. However, there is currently no systematic or comprehensive conclusion regarding the mechanism of PRDM16 in human tumours, including haematologic and solid tumours. The aim of this review is to provide an overview of the research progress on PRDM16 in haematologic and solid tumours by incorporating recent literature findings. Furthermore, we explore the prospects of PRDM16 in the precise diagnosis and treatment of human haematologic and solid tumours.


Subject(s)
DNA-Binding Proteins , Neoplasms , Transcription Factors , Humans , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Animals , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology
4.
JCO Glob Oncol ; 10: e2300419, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38991185

ABSTRACT

PURPOSE: There is no consensus on how to identify patients with multiple-level spinal metastases who would benefit from surgery. Previous studies have revealed that patients with hematologic malignancies have a significantly longer median survival time than those with solid tumor spinal metastases. We aimed to compare predictors and survival data between patients with spinal metastases, including hematologic malignancies (all-malignancies group), with only those with nonhematologic malignancies (nonhematologic malignancies group). MATERIALS AND METHODS: This single-center retrospective study included all patients age >18 years who underwent surgery to treat spinal metastases in our department from 2008 to 2018. The patients' baseline characteristics, treatment modalities, and laboratory results were analyzed. Survival was calculated from the date of surgery to the date of confirmed death. Cox regression analysis was used to identify independent predictors of survival. RESULTS: The study cohort comprised 186 patients with a mean age of 57.1 ± 13.4 years, 101 of whom were male and 18 of whom had hematologic malignancies. The median survival time was 201 days in the all-malignancies group and 168 days in the nonhematologic malignancies group. Independent predictors of survival differed between the two groups. Eastern Cooperative Oncology Group status and response to preoperative chemotherapy were identified as independent factors in both groups. However, radiosensitivity and CNS metastases were identified only in the all-malignancies group, and tumor growth potential, albumin status, and number of vertebrae were identified only in the nonhematologic malignancies group. The receiver operating characteristics were comparable in the two groups: 0.75 in the all-malignancies group and 0.77 in the nonhematologic malignancies group. CONCLUSION: Longer survival in patients with hematologic malignancies influences the overall prediction of survival. Tumor-specific prognostic factors may improve the prediction of survival in patients with spinal metastases.


Subject(s)
Hematologic Neoplasms , Spinal Neoplasms , Humans , Male , Middle Aged , Female , Spinal Neoplasms/secondary , Spinal Neoplasms/mortality , Spinal Neoplasms/surgery , Retrospective Studies , Hematologic Neoplasms/mortality , Hematologic Neoplasms/therapy , Hematologic Neoplasms/surgery , Hematologic Neoplasms/pathology , Aged , Adult , Prognosis
5.
J Hematol Oncol ; 17(1): 51, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38978094

ABSTRACT

In 2022, two updated classification systems for lymphoid neoplasms were published by the World Health Organization (WHO Classification of Haematolymphoid Tumours, 5th edition, referred to hereafter as WHO-HAEM5) and the International Consensus Conference (ICC) (Alaggio et al. in Leukemia 36(7):1720-1748, 2022; Campo et al. in Blood 140(11):1229-1253, 2022). Both classifications were conceived by both pathologists and clinicians with expertise in the field. The reasons for this have been reviewed previously (Arber et al. in Virchows Arch 482(1):1-9, 2023; Cree in Leukemia 36(7):1701-1702, 2022, Leukemia 36(11):2750, 2022). Given that both groups were using data-driven processes and consensus and used the revised 4th edition of the WHO Classification of Haematolymphoid Tumours (WHO-HAEM4R) as a starting point, it is not entirely surprising that the resulting classifications are quite similar. However, they are not identical and reflect preferences or approaches for certain unsettled areas as well as preferred terminology. In this review, we will compare nomenclature of the WHO-HAEM5 and ICC classifications, focusing on lymphoid neoplasms and lymphoproliferative disorders (LPDs).


Subject(s)
Consensus , World Health Organization , Humans , Neoplasms, Plasma Cell/classification , Neoplasms, Plasma Cell/diagnosis , Neoplasms, Plasma Cell/pathology , Killer Cells, Natural/pathology , Killer Cells, Natural/immunology , Hematologic Neoplasms/classification , Hematologic Neoplasms/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
6.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000208

ABSTRACT

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is an aggressive hematological malignancy derived from the precursors of plasmacytoid dendritic cells. Although disease awareness has increased over time, BPDCN represents a rare disease with an aggressive clinical course and a dismal prognosis. Due to the overlap in clinical and histological features with a large spectrum of inflammatory and neoplastic diseases, BPDCN is difficult to diagnose. Furthermore, given the rarity of the disease, treatment options for BPDCN are limited, sometimes changing by practitioner and hospitals. Treatment options range from conventional chemotherapy to the recently approved biologic agent tagraxofusp and stem cell transplantation. Therefore, a multidisciplinary approach with coordination among dermatologists, pathologists, and hematologists is ultimately imperative to reach the correct diagnosis and management of BPDCN.


Subject(s)
Dendritic Cells , Hematologic Neoplasms , Skin Neoplasms , Humans , Dendritic Cells/pathology , Hematologic Neoplasms/pathology , Skin Neoplasms/pathology , Prognosis
7.
Zhonghua Bing Li Xue Za Zhi ; 53(7): 651-654, 2024 Jul 08.
Article in Chinese | MEDLINE | ID: mdl-38955693

ABSTRACT

The fifth edition of the World Health Organization (WHO) classification of lymphohematopoietic system tumors updated the terminology, types of lesions, diagnostic criteria, nomenclature, and other aspects of lymphoid proliferations and lymphomas associated with immune deficiency and dysregulation. The important updates and main changes in this section were briefly introduced, in order to guide the precise classification of lymphoid proliferations and lymphomas associated with immune deficiency and dysregulation, and standardize pathological reports.


Subject(s)
Lymphoma , World Health Organization , Humans , Lymphoma/pathology , Lymphoma/classification , Lymphoproliferative Disorders/pathology , Lymphoproliferative Disorders/classification , Immunologic Deficiency Syndromes/classification , Immunologic Deficiency Syndromes/pathology , Terminology as Topic , Hematologic Neoplasms/pathology , Hematologic Neoplasms/classification
8.
Klin Onkol ; 38(3): 164-177, 2024.
Article in English | MEDLINE | ID: mdl-38960672

ABSTRACT

BACKGROUND: Histiocytoses are rare disorders characterized by the accumulation of macrophages, dendritic cells, or monocyte-derived cells in various tissues and organs of children and adults, with a wide range of clinical manifestations, presentations, and histology. The histiocytoses are classified according to the WHO Classification, the last version of which was published in 2022, or according to the Histiocyte Society Classification, with the last version published in 2016. PURPOSE: This text provides an overview of histiocytoses as described in the WHO Classification 2022.


Subject(s)
World Health Organization , Humans , Histiocytosis/pathology , Histiocytosis/classification , Histiocytosis/diagnosis , Hematologic Neoplasms/classification , Hematologic Neoplasms/pathology , Dendritic Cells/pathology
9.
Adv Exp Med Biol ; 1459: 291-320, 2024.
Article in English | MEDLINE | ID: mdl-39017849

ABSTRACT

Genetic alterations of the repressive ETS family transcription factor gene ETV6 are recurrent in several categories of hematopoietic malignancy, including subsets of B-cell and T-cell acute lymphoblastic leukemias (B-ALL and T-ALL), myeloid neoplasms, and mature B-cell lymphomas. ETV6 is essential for adult hematopoietic stem cells (HSCs), contributes to specific functions of some mature immune cells, and plays a key role in thrombopoiesis as demonstrated by familial ETV6 mutations associated with thrombocytopenia and predisposition to hematopoietic cancers, particularly B-ALL. ETV6 appears to have a tumor suppressor role in several hematopoietic lineages, as demonstrated by recurrent somatic loss-of-function (LoF) and putative dominant-negative alterations in leukemias and lymphomas. ETV6 rearrangements contribute to recurrent fusion oncogenes such as the B-ALL-associated transcription factor (TF) fusions ETV6::RUNX1 and PAX5::ETV6, rare drivers such as ETV6::NCOA6, and a spectrum of tyrosine kinase gene fusions encoding hyperactive signaling proteins that self-associate via the ETV6 N-terminal pointed domain. Another subset of recurrent rearrangements involving the ETV6 gene locus appear to function primarily to drive overexpression of the partner gene. This review surveys what is known about the biochemical and genome regulatory properties of ETV6 as well as our current understanding of how alterations in these functions contribute to hematopoietic and nonhematopoietic cancers.


Subject(s)
ETS Translocation Variant 6 Protein , Hematologic Neoplasms , Proto-Oncogene Proteins c-ets , Repressor Proteins , Humans , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Animals , Mutation , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism
10.
Exp Hematol ; 136: 104583, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39059457

ABSTRACT

Embryonic and fetal hematopoietic stem and progenitor cells differ in some key properties from cells that are part of the adult hematopoietic system. These include higher proliferation and self-renewal capacity, different globin gene usage, and differing lineage biases. Although these evolved to cover specific requirements of embryonic development, they can have serious consequences for the pathogenesis of hematologic malignancies that initiate prebirth in fetal blood cells and may result in a particularly aggressive disease that does not respond well to treatments that have been designed for adult leukemias. This indicates that these infant/pediatric leukemias should be considered developmental diseases, where a thorough understanding of their unique biology is essential for designing more effective therapies. In this review, we will highlight some of these unique fetal properties and detail the underlying molecular drivers of these phenotypes. We specifically focus on those that are pertinent to disease pathogenesis and that may therefore reveal disease vulnerabilities. We have also included an extensive description of the origins, phenotypes, and key molecular drivers of the main infant and pediatric leukemias that have a known prenatal origin. Importantly, successes in recent years in generating faithful models of these malignancies in which cellular origins, key drivers, and potential vulnerabilities can be investigated have resulted in uncovering potential, new therapeutic avenues.


Subject(s)
Hematopoietic Stem Cells , Humans , Child , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Hematologic Diseases/pathology , Hematologic Diseases/therapy , Hematologic Diseases/genetics , Hematologic Diseases/etiology , Animals , Infant , Hematologic Neoplasms/pathology , Hematologic Neoplasms/genetics , Hematologic Neoplasms/therapy , Leukemia/pathology , Leukemia/genetics , Leukemia/etiology , Leukemia/therapy
13.
Expert Opin Ther Pat ; 34(5): 365-382, 2024 May.
Article in English | MEDLINE | ID: mdl-38842051

ABSTRACT

INTRODUCTION: PIM Kinases (PIM-1, PIM-2, and PIM-3) have been reported to play crucial role in signaling cascades that govern cell survival, proliferation, and differentiation. Over-expression of these kinases leads to hematological malignancies such as diffuse large B cell lymphomas (DLBCL), multiple myeloma, leukemia, lymphoma and prostate cancer etc. PIM kinases as biomarkers and potential therapeutic targets have shown promise toward precision cancer therapy. The selective PIM-1, PIM-2, and/or PIM-3 isoform inhibitors have shown significant results in patients with advanced stages of cancer including relapsed/refractory cancer. AREAS COVERED: A comprehensive literature review of PIM Kinases (PIM-1, PIM-2, and PIM-3) in oncogenesis, the patented PIM kinase inhibitors (2016-Present), and their pharmacological and structural insights have been highlighted. EXPERT OPINION: Recently, PIM kinases viz. PIM-1, PIM-2, and PIM-3 (members of the serine/threonine protein kinase family) as therapeutic targets have attracted considerable interest in oncology especially in hematological malignancies. The patented PIM kinase inhibitors comprised of heterocyclic (fused)ring structure(s) like indole, pyridine, pyrazine, pyrazole, pyridazine, piperazine, thiazole, oxadiazole, quinoline, triazolo-pyridine, pyrazolo-pyridine, imidazo-pyridazine, oxadiazole-thione, pyrazolo-pyrimidine, triazolo-pyridazine, imidazo-pyridazine, pyrazolo-quinazoline and pyrazolo-pyridine etc. showed promising results in cancer chemotherapy.


Subject(s)
Antineoplastic Agents , Neoplasms , Patents as Topic , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-pim-1 , Humans , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/metabolism , Antineoplastic Agents/pharmacology , Animals , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/enzymology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/pathology , Molecular Targeted Therapy , Drug Development , Drug Design , Protein Serine-Threonine Kinases
14.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38928344

ABSTRACT

The association between leukemic stem cells (LSCs) and leukemia development has been widely established in the context of genetic alterations, epigenetic pathways, and signaling pathway regulation. Hematopoietic stem cells are at the top of the bone marrow hierarchy and can self-renew and progressively generate blood and immune cells. The microenvironment, niche cells, and complex signaling pathways that regulate them acquire genetic mutations and epigenetic alterations due to aging, a chronic inflammatory environment, stress, and cancer, resulting in hematopoietic stem cell dysregulation and the production of abnormal blood and immune cells, leading to hematological malignancies and blood cancer. Cells that acquire these mutations grow at a faster rate than other cells and induce clone expansion. Excessive growth leads to the development of blood cancers. Standard therapy targets blast cells, which proliferate rapidly; however, LSCs that can induce disease recurrence remain after treatment, leading to recurrence and poor prognosis. To overcome these limitations, researchers have focused on the characteristics and signaling systems of LSCs and therapies that target them to block LSCs. This review aims to provide a comprehensive understanding of the types of hematopoietic malignancies, the characteristics of leukemic stem cells that cause them, the mechanisms by which these cells acquire chemotherapy resistance, and the therapies targeting these mechanisms.


Subject(s)
Hematologic Neoplasms , Neoplastic Stem Cells , Humans , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Hematologic Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Hematopoietic Stem Cells/metabolism , Leukemia/pathology , Leukemia/genetics , Leukemia/metabolism , Signal Transduction , Animals , Tumor Microenvironment/genetics , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Mutation
15.
J Med Chem ; 67(13): 11326-11353, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38913763

ABSTRACT

BRD9 is a pivotal epigenetic factor involved in cancers and inflammatory diseases. Still, the limited selectivity and poor phenotypic activity of targeted agents make it an atypically undruggable target. PROTAC offers an alternative strategy for overcoming the issue. In this study, we explored diverse E3 ligase ligands for the contribution of BRD9 PROTAC degradation. Through molecular docking, binding affinity analysis, and structure-activity relationship study, we identified a highly potent PROTAC E5, with excellent BRD9 degradation (DC50 = 16 pM) and antiproliferation in MV4-11 cells (IC50 = 0.27 nM) and OCI-LY10 cells (IC50 = 1.04 nM). E5 can selectively degrade BRD9 and induce cell cycle arrest and apoptosis. Moreover, the therapeutic efficacy of E5 was confirmed in xenograft tumor models, accompanied by further RNA-seq analysis. Therefore, these results may pave the way and provide the reference for the discovery and investigation of highly effective PROTAC degraders.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Molecular Docking Simulation , Ubiquitin-Protein Ligases , Humans , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Structure-Activity Relationship , Cell Proliferation/drug effects , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , Mice , Drug Discovery , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/pathology , Hematologic Neoplasms/metabolism , Transcription Factors/metabolism , Transcription Factors/antagonists & inhibitors , Apoptosis/drug effects , Proteolysis/drug effects , Mice, Nude , Mice, Inbred BALB C , Xenograft Model Antitumor Assays , Drug Screening Assays, Antitumor , Bromodomain Containing Proteins
16.
Am J Physiol Cell Physiol ; 327(2): C372-C378, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38912739

ABSTRACT

Heparan sulfate proteoglycans are a family of glycoproteins that modulate cell signaling by binding growth factors and changing their bioavailability. Syndecans are a specific family of transmembrane heparan sulfate proteoglycans that regulate cell adhesion, migration, and signaling. In this review, we will summarize emerging evidence for the functions of syndecans in the normal and malignant blood systems and their microenvironments. More specifically, we detail the known functions of syndecans within normal hematopoietic stem cells. Furthermore, we discuss the functions of syndecans in hematological malignancies, including myeloid malignancies, lymphomas, and bleeding disorders. As normal and malignant hematopoietic cells require cues from their microenvironments to function, we also summarize the roles of syndecans in cells of the stromal, endothelial, and osteolineage compartments. Syndecan biology is a rapidly evolving field; a comprehensive understanding of these molecules and their place in the hematopoietic system promises to improve our grasp on disease processes and better predict the efficacies of growth factor-targeting therapies.


Subject(s)
Hematopoietic Stem Cells , Stem Cell Niche , Syndecans , Humans , Hematopoietic Stem Cells/metabolism , Animals , Syndecans/metabolism , Syndecans/genetics , Signal Transduction , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Hematopoiesis/physiology
19.
Leukemia ; 38(8): 1787-1798, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38937548

ABSTRACT

Germline heterozygous mutations in DDX41 predispose individuals to hematologic malignancies in adulthood. Most of these DDX41 mutations result in a truncated protein, leading to loss of protein function. To investigate the impact of these mutations on hematopoiesis, we generated mice with hematopoietic-specific knockout of one Ddx41 allele. Under normal steady-state conditions, there was minimal effect on lifelong hematopoiesis, resulting in a mild yet persistent reduction in red blood cell counts. However, stress induced by transplantation of the Ddx41+/- BM resulted in hematopoietic stem/progenitor cell (HSPC) defects and onset of hematopoietic failure upon aging. Transcriptomic analysis of HSPC subsets from the transplanted BM revealed activation of cellular stress responses, including upregulation of p53 target genes in erythroid progenitors. To understand how the loss of p53 affects the phenotype of Ddx41+/- HSPCs, we generated mice with combined Ddx41 and Trp53 heterozygous deletions. The reduction in p53 expression rescued the fitness defects in HSPC caused by Ddx41 heterozygosity. However, the combined Ddx41 and Trp53 mutant mice were prone to developing hematologic malignancies that resemble human myelodysplastic syndrome and acute myeloid leukemia. In conclusion, DDX41 heterozygosity causes dysregulation of the response to hematopoietic stress, which increases the risk of transformation with a p53 mutation.


Subject(s)
DEAD-box RNA Helicases , Haploinsufficiency , Hematologic Neoplasms , Hematopoiesis , Mutation , Tumor Suppressor Protein p53 , Animals , Tumor Suppressor Protein p53/genetics , DEAD-box RNA Helicases/genetics , Mice , Hematopoiesis/genetics , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Hematologic Neoplasms/etiology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Mice, Knockout , Humans , Stress, Physiological/genetics , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL