Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Nature ; 618(7966): 834-841, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37286599

ABSTRACT

Tumours most often arise from progression of precursor clones within a single anatomical niche. In the bone marrow, clonal progenitors can undergo malignant transformation to acute leukaemia, or differentiate into immune cells that contribute to disease pathology in peripheral tissues1-4. Outside the marrow, these clones are potentially exposed to a variety of tissue-specific mutational processes, although the consequences of this are unclear. Here we investigate the development of blastic plasmacytoid dendritic cell neoplasm (BPDCN)-an unusual form of acute leukaemia that often presents with malignant cells isolated to the skin5. Using tumour phylogenomics and single-cell transcriptomics with genotyping, we find that BPDCN arises from clonal (premalignant) haematopoietic precursors in the bone marrow. We observe that BPDCN skin tumours first develop at sun-exposed anatomical sites and are distinguished by clonally expanded mutations induced by ultraviolet (UV) radiation. A reconstruction of tumour phylogenies reveals that UV damage can precede the acquisition of alterations associated with malignant transformation, implicating sun exposure of plasmacytoid dendritic cells or committed precursors during BPDCN pathogenesis. Functionally, we find that loss-of-function mutations in Tet2, the most common premalignant alteration in BPDCN, confer resistance to UV-induced cell death in plasmacytoid, but not conventional, dendritic cells, suggesting a context-dependent tumour-suppressive role for TET2. These findings demonstrate how tissue-specific environmental exposures at distant anatomical sites can shape the evolution of premalignant clones to disseminated cancer.


Subject(s)
Cell Transformation, Neoplastic , Dendritic Cells , Leukemia, Myeloid, Acute , Skin Neoplasms , Skin , Ultraviolet Rays , Humans , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Cells/radiation effects , Cell Death/radiation effects , Cell Lineage/genetics , Cell Lineage/radiation effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cell Transformation, Neoplastic/radiation effects , Clone Cells/metabolism , Clone Cells/pathology , Clone Cells/radiation effects , Dendritic Cells/metabolism , Dendritic Cells/pathology , Dendritic Cells/radiation effects , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation/radiation effects , Organ Specificity , Single-Cell Gene Expression Analysis , Skin Neoplasms/etiology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Ultraviolet Rays/adverse effects , Skin/pathology , Skin/radiation effects
2.
Cancer Discov ; 12(2): 522-541, 2022 02.
Article in English | MEDLINE | ID: mdl-34615655

ABSTRACT

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is an aggressive leukemia of plasmacytoid dendritic cells (pDC). BPDCN occurs at least three times more frequently in men than in women, but the reasons for this sex bias are unknown. Here, studying genomics of primary BPDCN and modeling disease-associated mutations, we link acquired alterations in RNA splicing to abnormal pDC development and inflammatory response through Toll-like receptors. Loss-of-function mutations in ZRSR2, an X chromosome gene encoding a splicing factor, are enriched in BPDCN, and nearly all mutations occur in males. ZRSR2 mutation impairs pDC activation and apoptosis after inflammatory stimuli, associated with intron retention and inability to upregulate the transcription factor IRF7. In vivo, BPDCN-associated mutations promote pDC expansion and signatures of decreased activation. These data support a model in which male-biased mutations in hematopoietic progenitors alter pDC function and confer protection from apoptosis, which may impair immunity and predispose to leukemic transformation. SIGNIFICANCE: Sex bias in cancer is well recognized, but the underlying mechanisms are incompletely defined. We connect X chromosome mutations in ZRSR2 to an extremely male-predominant leukemia. Aberrant RNA splicing induced by ZRSR2 mutation impairs dendritic cell inflammatory signaling, interferon production, and apoptosis, revealing a sex- and lineage-related tumor suppressor pathway.This article is highlighted in the In This Issue feature, p. 275.


Subject(s)
Dendritic Cells/metabolism , Myeloproliferative Disorders/genetics , Ribonucleoproteins/genetics , Apoptosis , Female , Gender Identity , Humans , Male , Mutation
3.
Blood Adv ; 4(6): 1006-1011, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32182365

ABSTRACT

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a hematologic malignancy believed to originate from plasmacytoid dendritic cells (pDCs), the immune cells responsible for producing type 1 interferons during infection. Nearly all patients with BPDCN have prominent skin involvement, with cutaneous infiltration occupying the dermis and subcutis. One half of patients present with BPDCN cells only in the skin, with no evidence of disease elsewhere. Because normal pDCs are rare or absent in cutaneous sites, and they only traffic to the skin after activation by pathogen or inflammation, our aim was to determine if a microorganism is associated with BPDCN. We performed RNA sequencing in BPDCN skin and bone marrow, with cutaneous T-cell lymphoma (CTCL) and normal skin as controls. GATK-PathSeq was used to identify known microbial sequences. Bacterial reads in BPDCN skin were components of normal flora and did not distinguish BPDCN from controls. We then developed a new computational tool, virID (Viral Identification and Discovery; https://github.com/jnoms/virID), for identification of microbial-associated reads remaining unassigned after GATK-PathSeq. We found no evidence for a known or novel virus in BPDCN skin or bone marrow, despite confirming that virID could identify Merkel cell polyomavirus in Merkel cell carcinoma, human papillomavirus in head and neck squamous cell carcinoma, and Kaposi's sarcoma herpesvirus in Kaposi's sarcoma in a blinded fashion. Thus, at the level of sensitivity used here, we found no clear pathogen linked to BPDCN.


Subject(s)
Hematologic Neoplasms , Lymphoma, T-Cell, Cutaneous , Myeloproliferative Disorders , Skin Neoplasms , Dendritic Cells , Humans
4.
Nat Commun ; 11(1): 1406, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32179749

ABSTRACT

Chromatin organization is a highly orchestrated process that influences gene expression, in part by modulating access of regulatory factors to DNA and nucleosomes. Here, we report that the chromatin accessibility regulator HMGN1, a target of recurrent DNA copy gains in leukemia, controls myeloid differentiation. HMGN1 amplification is associated with increased accessibility, expression, and histone H3K27 acetylation of loci important for hematopoietic stem cells (HSCs) and leukemia, such as HoxA cluster genes. In vivo, HMGN1 overexpression is linked to decreased quiescence and increased HSC activity in bone marrow transplantation. HMGN1 overexpression also cooperates with the AML-ETO9a fusion oncoprotein to impair myeloid differentiation and enhance leukemia stem cell (LSC) activity. Inhibition of histone acetyltransferases CBP/p300 relieves the HMGN1-associated differentiation block. These data nominate factors that modulate chromatin accessibility as regulators of HSCs and LSCs, and suggest that targeting HMGN1 or its downstream effects on histone acetylation could be therapeutically active in AML.


Subject(s)
Chromatin/metabolism , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , Acetylation , Animals , Cell Differentiation , Cell Survival , Female , HMGN1 Protein/genetics , HMGN1 Protein/metabolism , Hematopoietic Stem Cells/cytology , Histones/genetics , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism
5.
J Clin Invest ; 129(11): 5005-5019, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31437130

ABSTRACT

The interleukin-3 receptor α subunit, CD123, is expressed in many hematologic malignancies including acute myeloid leukemia (AML) and blastic plasmacytoid dendritic cell neoplasm (BPDCN). Tagraxofusp (SL-401) is a CD123-targeted therapy consisting of interleukin-3 fused to a truncated diphtheria toxin payload. Factors influencing response to tagraxofusp other than CD123 expression are largely unknown. We interrogated tagraxofusp resistance in patients and experimental models and found that it was not associated with CD123 loss. Rather, resistant AML and BPDCN cells frequently acquired deficiencies in the diphthamide synthesis pathway, impairing tagraxofusp's ability to ADP-ribosylate cellular targets. Expression of DPH1, encoding a diphthamide pathway enzyme, was reduced by DNA CpG methylation in resistant cells. Treatment with the DNA methyltransferase inhibitor azacitidine restored DPH1 expression and tagraxofusp sensitivity. We also developed a drug-dependent ADP-ribosylation assay in primary cells that correlated with tagraxofusp activity and may represent an additional novel biomarker. As predicted by these results and our observation that resistance also increased mitochondrial apoptotic priming, we found that the combination of tagraxofusp and azacitidine was effective in patient-derived xenografts treated in vivo. These data have important implications for clinical use of tagraxofusp and led to a phase 1 study combining tagraxofusp and azacitidine in myeloid malignancies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Dendritic Cells/metabolism , Drug Delivery Systems , Hematologic Neoplasms , Interleukin-3 Receptor alpha Subunit/metabolism , Leukemia, Myeloid, Acute , Neoplasm Proteins/metabolism , Animals , Azacitidine/pharmacology , Cell Line, Tumor , DNA Methylation , Dendritic Cells/pathology , Female , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Nude , Minor Histocompatibility Antigens/metabolism , Recombinant Fusion Proteins/pharmacology , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
6.
Cancer Discov ; 7(2): 156-164, 2017 02.
Article in English | MEDLINE | ID: mdl-27986708

ABSTRACT

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is an aggressive hematologic malignancy with dismal outcomes for which no standard therapy exists. We found that primary BPDCN cells were dependent on the antiapoptotic protein BCL2 and were uniformly sensitive to the BCL2 inhibitor venetoclax, as measured by direct cytotoxicity, apoptosis assays, and dynamic BH3 profiling. Animals bearing BPDCN patient-derived xenografts had disease responses and improved survival after venetoclax treatment in vivo Finally, we report on 2 patients with relapsed/refractory BPDCN who received venetoclax off-label and experienced significant disease responses. We propose that venetoclax or other BCL2 inhibitors undergo expedited clinical evaluation in BPDCN, alone or in combination with other therapies. In addition, these data illustrate an example of precision medicine to predict treatment response using ex vivo functional assessment of primary tumor tissue, without requiring a genetic biomarker. SIGNIFICANCE: Therapy for BPDCN is inadequate, and survival in patients with the disease is poor. We used primary tumor cell functional profiling to predict BCL2 antagonist sensitivity as a common feature of BPDCN, and demonstrated in vivo clinical activity of venetoclax in patient-derived xenografts and in 2 patients with relapsed chemotherapy-refractory disease. Cancer Discov; 7(2); 156-64. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 115.


Subject(s)
Antineoplastic Agents/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Dendritic Cells/pathology , Hematologic Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/administration & dosage , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dendritic Cells/metabolism , Hematologic Neoplasms/metabolism , Humans , Male , Mice , Precision Medicine , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology , Survival Analysis , Treatment Outcome , Xenograft Model Antitumor Assays
7.
J Biochem ; 159(1): 17-25, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26590301

ABSTRACT

Recent progress in high-speed sequencing technology has revealed that tumors harbor novel mutations in a variety of genes including those for molecules involved in epigenetics and splicing, some of which were not categorized to previously thought malignancy-related genes. However, despite thorough identification of mutations in solid tumors and hematological malignancies, how these mutations induce cell transformation still remains elusive. In addition, each tumor usually contains multiple mutations or sometimes consists of multiple clones, which makes functional analysis difficult. Fifteen years ago, it was proposed that combination of two types of mutations induce acute leukemia; Class I mutations induce cell growth or inhibit apoptosis while class II mutations block differentiation, co-operating in inducing acute leukemia. This notion has been proven using a variety of mouse models, however most of recently found mutations are not typical class I/II mutations. Although some novel mutations have been found to functionally work as class I or II mutation in leukemogenesis, the classical class I/II theory seems to be too simple to explain the whole story. We here overview the molecular basis of hematological malignancies based on clinical and experimental results, and propose a new working hypothesis for leukemogenesis.


Subject(s)
Carcinogenesis/genetics , Hematologic Neoplasms/genetics , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Translocation, Genetic , Animals , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Transgenic , Mutation , Oncogene Proteins, Fusion/genetics , Phenotype
8.
Exp Hematol ; 43(4): 300-8.e1, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25534203

ABSTRACT

Two types of CCAAT-enhancer-binding protein α (C/EBPα) mutants are found in acute myeloid leukemia (AML) patients: N-terminal frame-shift mutants (C/EBPα-N(m)) generating p30 as a dominant form and C-terminal basic leucine zipper domain mutants (C/EBPα-C(m)). We have previously shown that C/EBPα-K304_R323dup belonging to C/EBPα-C(m), but not C/EBPα-T60fsX159 belonging to C/EBPα-N(m), alone induced AML in mouse bone marrow transplantation (BMT) models. Here we show that various C/EBPα-C(m) mutations have a similar, but not identical, potential in myeloid leukemogenesis. Notably, like C/EBPα-K304_R323dup, any type of C/EBPα-C(m) tested (C/EBPα-S299_K304dup, K313dup, or N321D) by itself induced AML, albeit with different latencies after BMT; C/EBPα-N321D induced AML with the shortest latency. By analyzing the gene expression profiles of C/EBPα-N321D- and mock-transduced c-kit(+)Sca-1(+)Lin(-) cells, we identified Csf1r as a gene downregulated by C/EBPα-N321D. In addition, leukemic cells expressing C/EBPα-C(m) exhibited low levels of colony stimulating factor 1 receptor in mice. On the other hand, transduction with C/EBPα-N(m) did not influence Csf1r expression in c-kit(+)Sca-1(+)Lin(-) cells, implying a unique role for C/EBPα-C(m) in downregulating Csf1r. Importantly, Csf1r overexpression collaborated with C/EBPα-N321D to induce fulminant AML with leukocytosis in mouse BMT models to a greater extent than did C/EBPα-N321D alone. Collectively, these results suggest that C/EBPα-C(m)-mediated downregulation of Csf1r has a negative, rather than a positive, impact on the progression of AML involving C/EBPα-C(m), which might possibly be accelerated by additional genetic and/or epigenetic alterations inducing Csf1r upregulation.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/physiology , Down-Regulation/physiology , Leukemia, Myeloid, Acute/pathology , Mutation , Receptor, Macrophage Colony-Stimulating Factor/physiology , Animals , Base Sequence , CCAAT-Enhancer-Binding Protein-alpha/genetics , DNA Primers , Disease Progression , Flow Cytometry , Humans , Leukemia, Myeloid, Acute/physiopathology , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
9.
Proc Jpn Acad Ser B Phys Biol Sci ; 90(10): 389-404, 2014.
Article in English | MEDLINE | ID: mdl-25504228

ABSTRACT

Myeloid malignancies consist of acute myeloid leukemia (AML), myelodysplastic syndromes (MDS) and myeloproliferative neoplasm (MPN). The latter two diseases have preleukemic features and frequently evolve to AML. As with solid tumors, multiple mutations are required for leukemogenesis. A decade ago, these gene alterations were subdivided into two categories: class I mutations stimulating cell growth or inhibiting apoptosis; and class II mutations that hamper differentiation of hematopoietic cells. In mouse models, class I mutations such as the Bcr-Abl fusion kinase induce MPN by themselves and some class II mutations such as Runx1 mutations induce MDS. Combinations of class I and class II mutations induce AML in a variety of mouse models. Thus, it was postulated that hematopoietic cells whose differentiation is blocked by class II mutations would autonomously proliferate with class I mutations leading to the development of leukemia. Recent progress in high-speed sequencing has enabled efficient identification of novel mutations in a variety of molecules including epigenetic factors, splicing factors, signaling molecules and proteins in the cohesin complex; most of these are not categorized as either class I or class II mutations. The functional consequences of these mutations are now being extensively investigated. In this article, we will review the molecular basis of hematological malignancies, focusing on mouse models and the interfaces between these models and clinical findings, and revisit the classical class I/II hypothesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic , Hematologic Neoplasms/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Myelodysplastic Syndromes/genetics , Animals , Cell Proliferation/genetics , Cell Transformation, Neoplastic/metabolism , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Hematologic Neoplasms/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Myelodysplastic Syndromes/metabolism
11.
Blood ; 123(25): 3932-42, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24825862

ABSTRACT

High levels of HES1 expression are frequently found in BCR-ABL(+) chronic myelogenous leukemia in blast crisis (CML-BC). In mouse bone marrow transplantation (BMT) models, co-expression of BCR-ABL and Hes1 induces CML-BC-like disease; however, the underlying mechanism remained elusive. Here, based on gene expression analysis, we show that MMP-9 is upregulated by Hes1 in common myeloid progenitors (CMPs). Analysis of promoter activity demonstrated that Hes1 upregulated MMP-9 by activating NF-κB. Analysis of 20 samples from CML-BC patients showed that MMP-9 was highly expressed in three, with two exhibiting high levels of HES1 expression. Interestingly, MMP-9 deficiency impaired the cobblestone area-forming ability of CMPs expressing BCR-ABL and Hes1 that were in conjunction with a stromal cell layer. In addition, CMPs expressing BCR-ABL and Hes1 secreted MMP-9, promoting the release of soluble Kit-ligand (sKitL) from stromal cells, thereby enhancing proliferation of the leukemic cells. In accordance, mice transplanted with CMPs expressing BCR-ABL and Hes1 exhibited high levels of sKitL as well as MMP-9 in the serum. Importantly, MMP-9 deficiency impaired the development of CML-BC-like disease induced by BCR-ABL and Hes1 in mouse BMT models. The present results suggest that Hes1 promotes the development of CML-BC, partly through MMP-9 upregulation in leukemic cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Blast Crisis/genetics , Gene Expression Regulation, Leukemic , Homeodomain Proteins/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Matrix Metalloproteinase 9/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blast Crisis/metabolism , Bone Marrow Transplantation/methods , Cell Movement/genetics , Cell Proliferation , Flow Cytometry , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Homeodomain Proteins/metabolism , Humans , Kaplan-Meier Estimate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Models, Genetic , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcription Factor HES-1 , Up-Regulation
12.
Exp Hematol ; 42(5): 369-379.e3, 2014 May.
Article in English | MEDLINE | ID: mdl-24486648

ABSTRACT

We have previously shown that elevated expression of Hairy enhancer of split 1 (Hes1) contributes to blast crisis transition in Bcr-Abl-positive chronic myelogenous leukemia. Here we investigate whether Hes1 is involved in the development of other myeloid neoplasms. Notably, Hes1 expression was elevated in only a few cases of 65 samples with different types of myeloid neoplasms. Interestingly, elevated expression of Hes1 was found in two of five samples of Fip1-like1 platelet-derived growth factor receptor-α (FIP1L1-PDGFA)-positive myeloid neoplasms associated with eosinophilia. Whereas FIP1L1-PDGFRα alone induced acute T-cell leukemia or myeloproliferative neoplasms in mouse bone marrow transplantation models, mice transplanted with bone marrow cells expressing both Hes1 and FIP1L1-PDGFRα developed acute leukemia characterized by an expansion of myeloid blasts and leukemic cells without eosinophilic granules. FIP1L1-PDGFRα conferred cytokine-independent growth to Hes1-transduced common myeloid progenitors, interleukin-3-dependent cells. Imatinib inhibited the growth of common myeloid progenitors expressing Hes1 with FIP1L1-PDGFRα, but not with imatinib-resistant FIP1L1-PDGFRα mutants harboring T674I or D842V. In contrast, ponatinib efficiently eradicated leukemic cells expressing Hes1 and the imatinib-resistant FLP1L1-PDGFRΑ mutant in vitro and in vivo. Thus, we have established mouse models of FIP1L1-PDGFRA-positive leukemia in myeloid blast crisis, which will help elucidate the pathogenesis of the disease and develop a new treatment for it.


Subject(s)
Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Blast Crisis/metabolism , Gene Expression Regulation, Leukemic , Homeodomain Proteins/biosynthesis , Leukemia, Myeloid, Acute/mortality , Neoplasms, Experimental/metabolism , Oncogene Proteins, Fusion/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Amino Acid Substitution , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Benzamides/pharmacology , Blast Crisis/genetics , Blast Crisis/pathology , Female , Homeodomain Proteins/genetics , Humans , Imatinib Mesylate , Interleukin-3/biosynthesis , Interleukin-3/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred BALB C , Mutation, Missense , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Oncogene Proteins, Fusion/genetics , Piperazines/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Transcription Factor HES-1 , mRNA Cleavage and Polyadenylation Factors/genetics
13.
Sci Rep ; 4: 4012, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24500246

ABSTRACT

The quiescent (G0) phase of the cell cycle is the reversible phase from which the cells exit from the cell cycle. Due to the difficulty of defining the G0 phase, quiescent cells have not been well characterized. In this study, a fusion protein consisting of mVenus and a defective mutant of CDK inhibitor, p27 (p27K(-)) was shown to be able to identify and isolate a population of quiescent cells and to effectively visualize the G0 to G1 transition. By comparing the expression profiles of the G0 and G1 cells defined by mVenus-p27K(-), we have identified molecular features of quiescent cells. Quiescence is also an important feature of many types of stem cells, and mVenus-p27K(-)-transgenic mice enabled the detection of the quiescent cells with muscle stem cell markers in muscle in vivo. The mVenus-p27K(-) probe could be useful in investigating stem cells as well as quiescent cells.


Subject(s)
Bacterial Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , G1 Phase/genetics , Luminescent Proteins/genetics , Recombinant Fusion Proteins/genetics , Resting Phase, Cell Cycle/genetics , 3T3 Cells , Animals , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Gene Expression , Intracellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , MicroRNAs/biosynthesis , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Recombinant Fusion Proteins/biosynthesis , Retinoblastoma Protein/biosynthesis , Selenium-Binding Proteins/biosynthesis , Selenium-Binding Proteins/genetics , Stem Cells/cytology , Tumor Suppressor Protein p53/biosynthesis
14.
J Clin Invest ; 123(11): 4627-40, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24216483

ABSTRACT

Recurrent mutations in the gene encoding additional sex combs-like 1 (ASXL1) are found in various hematologic malignancies and associated with poor prognosis. In particular, ASXL1 mutations are common in patients with hematologic malignancies associated with myelodysplasia, including myelodysplastic syndromes (MDSs), and chronic myelomonocytic leukemia. Although loss-of-function ASXL1 mutations promote myeloid transformation, a large subset of ASXL1 mutations is thought to result in stable truncation of ASXL1. Here we demonstrate that C-terminal­truncating Asxl1 mutations (ASXL1-MTs) inhibited myeloid differentiation and induced MDS-like disease in mice. ASXL1-MT mice displayed features of human-associated MDS, including multi-lineage myelodysplasia, pancytopenia, and occasional progression to overt leukemia. ASXL1-MT resulted in derepression of homeobox A9 (Hoxa9) and microRNA-125a (miR-125a) expression through inhibition of polycomb repressive complex 2­mediated (PRC2-mediated) methylation of histone H3K27. miR-125a reduced expression of C-type lectin domain family 5, member a (Clec5a), which is involved in myeloid differentiation. In addition, HOXA9 expression was high in MDS patients with ASXL1-MT, while CLEC5A expression was generally low. Thus, ASXL1-MT­induced MDS-like disease in mice is associated with derepression of Hoxa9 and miR-125a and with Clec5a dysregulation. Our data provide evidence for an axis of MDS pathogenesis that implicates both ASXL1 mutations and miR-125a as therapeutic targets in MDS.


Subject(s)
Mutant Proteins/genetics , Mutation , Myelodysplastic Syndromes/genetics , Repressor Proteins/genetics , Animals , Cell Line , Disease Models, Animal , Histones/metabolism , Homeodomain Proteins/genetics , Humans , Lectins, C-Type/genetics , Methylation , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Myelopoiesis/genetics , Peptide Fragments/genetics , Receptors, Cell Surface/genetics
15.
Int J Hematol ; 96(5): 638-48, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23097187

ABSTRACT

Activating mutations of c-Kit are frequently found in acute myeloid leukemia (AML) patients harboring t(8;21) chromosomal translocation generating a fusion protein AML1-ETO. Here we show that an active mutant of c-Kit cooperates with AML1-ETO to induce AML in mouse bone marrow transplantation models. Leukemic cells expressing AML1-ETO with c-Kit(D814V) were serially transplantable. Transplantation experiments indicated that lineage(-)c-Kit(+)Sca-1(+) (KSL) leukemic cells, but not lineage(+) leukemic cells, were enriched for leukemia stem cells (LSCs). Comparison of gene expression profiles between KSL leukemic and normal cells delineated that CD200R1 was highly expressed in KSL leukemic cells as compared with KSL normal cells. Upregulation of CD200R1 was verified in lineage(-) leukemic cells, but not in lineage(+) leukemic cells. CD200R1 expression in the lineage(-) leukemic cells was not correlated with the frequency of LSCs, indicating that CD200R1 is not a useful marker for LSCs in these models. Interestingly, CD200R1 was upregulated in KSL cells transduced with AML1-ETO, but not with other leukemogenic mutants, including c-Kit(D814V), AML1(D171N), and AML1(S291fsX300). Consistently, upregulation of CD200R1 in lineage(-) leukemic cells was observed only in the BM of mice suffering from AML1-ETO-positive leukemia. In conclusion, AML1-ETO upregulated CD200R1 in lineage(-) cells, which was characteristic of AML1-ETO-positive leukemia in mice.


Subject(s)
Antigens, Surface/biosynthesis , Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Leukemic , Leukemia/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Cell Surface/biosynthesis , Transcription Factors/metabolism , Up-Regulation , Amino Acid Substitution , Animals , Antigens, Surface/genetics , Cell Line, Tumor , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/genetics , Leukemia/genetics , Leukemia/pathology , Mice , Mutation, Missense , Oncogene Proteins, Fusion/genetics , Orexin Receptors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Rats , Receptors, Cell Surface/genetics , Transcription Factors/genetics
17.
J Immunol ; 189(4): 1773-9, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22772446

ABSTRACT

Leukocyte mono-Ig-like receptor 5 (LMIR5, also called CD300b) is an activating receptor expressed in myeloid cells. We have previously demonstrated that T cell Ig mucin 1 works as a ligand for LMIR5 in mouse ischemia/reperfusion injury of the kidneys. In this article, we show that LMIR5 is implicated in LPS-induced sepsis in mice. Notably, neutrophils constitutively released a soluble form of LMIR5 (sLMIR5) through proteolytic cleavage of surface LMIR5. Stimulation with TLR agonists augmented the release of sLMIR5. LPS administration or peritonitis induction increased serum levels of sLMIR5 in mice, which was substantially inhibited by neutrophil depletion. Thus, neutrophils were the main source of LPS-induced sLMIR5 in vivo. On the other hand, i.p. administration of LMIR5-Fc, a surrogate of sLMIR5, bound to resident macrophages (M) and stimulated transient inflammation in mice. Consistently, LMIR5-Fc induced in vitro cytokine production of peritoneal M via its unknown ligand. Interestingly, LMIR5 deficiency profoundly reduced systemic cytokine production and septic mortality in LPS-administered mice, although it did not affect in vitro cytokine production of LPS-stimulated peritoneal M. Importantly, the resistance of LMIR5-deficient mice to LPS- or peritonitis-induced septic death was decreased by LMIR5-Fc administration, implicating sLMIR5 in LPS responses in vivo. Collectively, neutrophil-derived sLMIR5 amplifies LPS-induced lethal inflammation.


Subject(s)
Neutrophils/immunology , Receptors, Immunologic/immunology , Sepsis/immunology , Animals , Blotting, Western , Cytokines/biosynthesis , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Inflammation/chemically induced , Inflammation/immunology , Lipopolysaccharides/immunology , Lipopolysaccharides/toxicity , Macrophages, Peritoneal/immunology , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptors, Immunologic/genetics , Sepsis/chemically induced , Solubility , Transfection
18.
Int J Hematol ; 95(2): 167-75, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22189847

ABSTRACT

The Bcr-Abl oncogene causes human Philadelphia chromosome-positive (Ph(+)) leukemias, including B-cell acute lymphoblastic leukemia (B-ALL) and chronic myeloid leukemia (CML) with chronic phase (CML-CP) to blast crisis (CML-BC). Previous studies have demonstrated that Src family kinases are required for the induction of B-ALL, but not for CML, which is induced by Bcr-Abl in mice. In contrast, it has been reported that Fyn is up-regulated in human CML-BC compared with CML-CP, implicating Fyn in the blast crisis transition. Here, we aimed to delineate the exact role of Fyn in the induction/progression of Ph(+) leukemias. We found that Fyn is expressed in mouse hematopoietic cells at varying stages of development, including c-kit(+)Sca-1(+)Lin(-) cells. Notably, Fyn is highly expressed in some of human lymphomas, but not in human Ph(+) leukemias including CML-BC. In mouse bone marrow transplantation models, mice transplanted with wild-type or Fyn-deficient bone marrow cells transduced with Bcr-Abl showed no differences in the development of B-ALL or CML-like diseases. Similarly, Fyn deficiency failed to impact the development of myeloid CML-BC induced by Bcr-Abl and Hes1. Elevated expression of Fyn was not found in mouse samples of Bcr-Abl-mediated CML- and CML-BC-like diseases. Thus, Fyn is not required for the pathogenesis of Bcr-Abl-mediated leukemias.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic/physiology , Hematopoietic Stem Cells/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-fyn/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Marrow Transplantation , Disease Models, Animal , Homeodomain Proteins/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/physiopathology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/physiopathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/physiopathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Proto-Oncogene Proteins c-fyn/physiology , Transcription Factor HES-1
19.
Biol Blood Marrow Transplant ; 17(6): 841-51, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20849969

ABSTRACT

To evaluate whether rescue with cord blood transplantation (CBT) could improve the poor survival after graft failure (GF), we surveyed the data of 80 adult patients (median age, 51 years) who received CBT within 3 months of GF (primary 64, secondary 16), with fludarabine-based reduced-intensity regimens with or without melphalan, busulfan, cyclophosphamide, and/or 2-4 Gy total-body irradiation (TBI). A median number of 2.4 × 10(7)/kg total nucleated cells (TNC) were infused, and among the 61 evaluable patients who survived for more than 28 days, 45 (74%) engrafted. The median follow-up of surviving patients was 325 days, and the 1-year overall survival rate was 33% despite poor performance status (2-4, 60%), carryover organ toxicities (grade 3/4, 14%), and infections (82%) prior to CBT. Day 100 transplantation-related mortality was 45%, with 60% related to infectious complications. Multivariate analysis showed that the infusion of TNC ≥2.5 × 10(7)/kg and an alkylating agent-containing regimen were associated with a higher probability of engraftment, and that high risk-status at the preceding transplantation and grade 3/4 organ toxicities before CBT were associated with an increased risk of mortality. In conclusion, in an older population of patients, our data support the feasibility of CBT with a reduced-intensity conditioning regimen for GF.


Subject(s)
Cord Blood Stem Cell Transplantation/mortality , Graft Rejection/pathology , Graft Rejection/prevention & control , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Transplantation Conditioning/methods , Adult , Busulfan/administration & dosage , Cyclophosphamide/administration & dosage , Disease-Free Survival , Female , Graft Rejection/immunology , Graft Rejection/mortality , Graft Survival/immunology , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Humans , Longitudinal Studies , Lymphocyte Count , Male , Melphalan/administration & dosage , Multivariate Analysis , Salvage Therapy/methods , Treatment Outcome , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
20.
Blood ; 117(1): 221-33, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-20884804

ABSTRACT

Two types of mutations of a transcription factor CCAAT-enhancer binding protein α (C/EBPα) are found in leukemic cells of 5%-14% of acute myeloid leukemia (AML) patients: N-terminal mutations expressing dominant negative p30 and C-terminal mutations in the basic leucine zipper domain. Our results showed that a mutation of C/EBPα in one allele was observed in AML after myelodysplastic syndrome, while the 2 alleles are mutated in de novo AML. Unlike an N-terminal frame-shift mutant (C/EBPα-N(m))-transduced cells, a C-terminal mutant (C/EBPα-C(m))-transduced cells alone induced AML with leukopenia in mice 4-12 months after bone marrow transplantation. Coexpression of both mutants induced AML with marked leukocytosis with shorter latencies. Interestingly, C/EBPα-C(m) collaborated with an Flt3-activating mutant Flt3-ITD in inducing AML. Moreover, C/EBPα-C(m) strongly blocked myeloid differentiation of 32Dcl3 cells, suggesting its class II mutation-like role in leukemogenesis. Although C/EBPα-C(m) failed to inhibit transcriptional activity of wild-type C/EBPα, it suppressed the synergistic effect between C/EBPα and PU.1. On the other hand, C/EBPα-N(m) inhibited C/EBPα activation in the absence of PU.1, despite low expression levels of p30 protein generated by C/EBPα-N(m). Thus, 2 types of C/EBPα mutations are implicated in leukemo-genesis, involving different and cooperating molecular mechanisms.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/genetics , Disease Models, Animal , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/pathology , Mutation/genetics , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/pathology , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Blotting, Western , Bone Marrow Transplantation , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation , Cell Proliferation , Electrophoretic Mobility Shift Assay , Female , Flow Cytometry , Humans , Leukemia, Myeloid, Acute/therapy , Luciferases/metabolism , Male , Mice , Middle Aged , Myelodysplastic Syndromes/therapy , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , RNA, Messenger/genetics , Survival Rate , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...