Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 168(5): 749-750, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235190

ABSTRACT

Targeting of the RAS pathway has long been a critical therapeutic challenge in oncology. Burgess et al. examine how the relative expression of mutant and wild-type KRAS modulates clonal fitness and sensitivity to MEK inhibitors in a model of KrasG12D mutant acute myeloid leukemia and propose its use as a predictive biomarker.


Subject(s)
Mutation/drug effects , Protein Kinase Inhibitors/pharmacology , Genes, ras/drug effects , Humans , Leukemia, Myeloid, Acute , ras Proteins/genetics
2.
Cell ; 165(2): 303-16, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27058663

ABSTRACT

Leukemia stem cells (LSCs) have the capacity to self-renew and propagate disease upon serial transplantation in animal models, and elimination of this cell population is required for curative therapies. Here, we describe a series of pooled, in vivo RNAi screens to identify essential transcription factors (TFs) in a murine model of acute myeloid leukemia (AML) with genetically and phenotypically defined LSCs. These screens reveal the heterodimeric, circadian rhythm TFs Clock and Bmal1 as genes required for the growth of AML cells in vitro and in vivo. Disruption of canonical circadian pathway components produces anti-leukemic effects, including impaired proliferation, enhanced myeloid differentiation, and depletion of LSCs. We find that both normal and malignant hematopoietic cells harbor an intact clock with robust circadian oscillations, and genetic knockout models reveal a leukemia-specific dependence on the pathway. Our findings establish a role for the core circadian clock genes in AML.


Subject(s)
ARNTL Transcription Factors/genetics , CLOCK Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , Animals , Circadian Rhythm , Disease Models, Animal , Gene Knockout Techniques , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells/metabolism , RNA Interference , RNA, Small Interfering/metabolism
3.
Cell ; 146(5): 697-708, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21884932

ABSTRACT

AKT activation is associated with many malignancies, where AKT acts, in part, by inhibiting FOXO tumor suppressors. We show a converse role for AKT/FOXOs in acute myeloid leukemia (AML). Rather than decreased FOXO activity, we observed that FOXOs are active in ∼40% of AML patient samples regardless of genetic subtype. We also observe this activity in human MLL-AF9 leukemia allele-induced AML in mice, where either activation of Akt or compound deletion of FoxO1/3/4 reduced leukemic cell growth, with the latter markedly diminishing leukemia-initiating cell (LIC) function in vivo and improving animal survival. FOXO inhibition resulted in myeloid maturation and subsequent AML cell death. FOXO activation inversely correlated with JNK/c-JUN signaling, and leukemic cells resistant to FOXO inhibition responded to JNK inhibition. These data reveal a molecular role for AKT/FOXO and JNK/c-JUN in maintaining a differentiation blockade that can be targeted to inhibit leukemias with a range of genetic lesions.


Subject(s)
Forkhead Transcription Factors/metabolism , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Antigens, CD34/metabolism , Apoptosis , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Line, Tumor , Cells, Cultured , Disease Models, Animal , Forkhead Box Protein O3 , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism
4.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Article in English | MEDLINE | ID: mdl-35149558

ABSTRACT

Immune evasion is a significant contributor to tumor evolution, and the immunoinhibitory axis PD-1/PD-L1 is a frequent mechanism employed to escape tumor immune surveillance. To identify cancer drivers involved in immune evasion, we performed a CRISPR-Cas9 screen of tumor suppressor genes regulating the basal and interferon (IFN)-inducible cell surface levels of PD-L1. Multiple regulators of PD-L1 were identified, including IRF2, ARID2, KMT2D, and AAMP. We also identified CTCF and the cohesin complex proteins, known regulators of chromatin architecture and transcription, among the most potent negative regulators of PD-L1 cell surface expression. Additionally, loss of the cohesin subunit RAD21 was shown to up-regulate PD-L2 and MHC-I surface expression. PD-L1 and MHC-I suppression by cohesin were shown to be conserved in mammary epithelial and myeloid cells. Comprehensive examination of the transcriptional effect of STAG2 deficiency in epithelial and myeloid cells revealed an activation of strong IFN and NF-κB expression signatures. Inhibition of JAK-STAT or NF-κB pathways did not result in rescue of PD-L1 up-regulation in RAD21-deficient cells, suggesting more complex or combinatorial mechanisms at play. Discovery of the PD-L1 and IFN up-regulation in cohesin-mutant cells expands our understanding of the biology of cohesin-deficient cells as well as molecular regulation of the PD-L1 molecule.


Subject(s)
B7-H1 Antigen/metabolism , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation, Neoplastic/physiology , Neoplasms/metabolism , B7-H1 Antigen/genetics , CCCTC-Binding Factor/genetics , Cell Cycle Proteins/genetics , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Humans , Janus Kinases/genetics , Janus Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Up-Regulation , Cohesins
5.
Blood ; 138(8): 649-661, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34157074

ABSTRACT

Cohesin is a multisubunit protein complex that forms a ring-like structure around DNA. It is essential for sister chromatid cohesion, chromatin organization, transcriptional regulation, and DNA damage repair and plays a major role in dynamically shaping the genome architecture and maintaining DNA integrity. The core complex subunits STAG2, RAD21, SMC1, and SMC3, as well as its modulators PDS5A/B, WAPL, and NIPBL, have been found to be recurrently mutated in hematologic and solid malignancies. These mutations are found across the full spectrum of myeloid neoplasia, including pediatric Down syndrome-associated acute megakaryoblastic leukemia, myelodysplastic syndromes, chronic myelomonocytic leukemia, and de novo and secondary acute myeloid leukemias. The mechanisms by which cohesin mutations act as drivers of clonal expansion and disease progression are still poorly understood. Recent studies have described the impact of cohesin alterations on self-renewal and differentiation of hematopoietic stem and progenitor cells, which are associated with changes in chromatin and epigenetic state directing lineage commitment, as well as genomic integrity. Herein, we review the role of the cohesin complex in healthy and malignant hematopoiesis. We discuss clinical implications of cohesin mutations in myeloid malignancies and discuss opportunities for therapeutic targeting.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Hematologic Neoplasms , Leukemia, Myeloid , Mutation , Myeloproliferative Disorders , Neoplasm Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Leukemia, Myeloid/therapy , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Myeloproliferative Disorders/therapy , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Cohesins
6.
Int J Mol Sci ; 24(10)2023 May 09.
Article in English | MEDLINE | ID: mdl-37239828

ABSTRACT

The erythropoietin receptor (EPOR) is a transmembrane type I receptor with an essential role in the proliferation and differentiation of erythroid progenitors. Besides its function during erythropoiesis, EPOR is expressed and has protective effect in various non-hematopoietic tissues, including tumors. Currently, the advantageous aspect of EPOR related to different cellular events is still under scientific investigation. Besides its well-known effect on cell proliferation, apoptosis and differentiation, our integrative functional study revealed its possible associations with metabolic processes, transport of small molecules, signal transduction and tumorigenesis. Comparative transcriptome analysis (RNA-seq) identified 233 differentially expressed genes (DEGs) in EPOR overexpressed RAMA 37-28 cells compared to parental RAMA 37 cells, whereas 145 genes were downregulated and 88 upregulated. Of these, for example, GPC4, RAP2C, STK26, ZFP955A, KIT, GAS6, PTPRF and CXCR4 were downregulated and CDH13, NR0B1, OCM2, GPM6B, TM7SF3, PARVB, VEGFD and STAT5A were upregulated. Surprisingly, two ephrin receptors, EPHA4 and EPHB3, and EFNB1 ligand were found to be upregulated as well. Our study is the first demonstrating robust differentially expressed genes evoked by simple EPOR overexpression without the addition of erythropoietin ligand in a manner which remains to be elucidated.


Subject(s)
Adenocarcinoma , Erythropoietin , Rats , Animals , Receptors, Erythropoietin/metabolism , Ligands , Erythropoietin/pharmacology , Signal Transduction , Cell Proliferation/genetics
7.
Int J Mol Sci ; 22(13)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34281163

ABSTRACT

Erythropoietin (EPO) acts on multiple tissues through its receptor EPOR, a member of a cytokine class I receptor superfamily with pleiotropic effects. The interaction of EPO and EPOR triggers the activation of several signaling pathways that induce erythropoiesis, including JAK2/STAT5, PI3K/AKT, and MAPK. The canonical EPOR/JAK2/STAT5 pathway is a known regulator of differentiation, proliferation, and cell survival of erythroid progenitors. In addition, its role in the protection of other cells, including cancer cells, is under intense investigation. The involvement of EPOR/JAK2/STAT5 in other processes such as mRNA splicing, cytoskeleton reorganization, and cell metabolism has been recently described. The transcriptomics, proteomics, and epigenetic studies reviewed in this article provide a detailed understanding of EPO signalization. Advances in this area of research may be useful for improving the efficacy of EPO therapy in hematologic disorders, as well as in cancer treatment.


Subject(s)
Erythropoietin/metabolism , STAT5 Transcription Factor/metabolism , STAT5 Transcription Factor/physiology , Animals , Cell Differentiation/drug effects , Epigenomics/methods , Erythropoiesis/drug effects , Erythropoietin/physiology , Humans , Janus Kinase 2/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Erythropoietin/metabolism , Receptors, Erythropoietin/physiology , STAT5 Transcription Factor/genetics , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcriptome/genetics
8.
Int J Mol Sci ; 22(14)2021 Jul 19.
Article in English | MEDLINE | ID: mdl-34299300

ABSTRACT

Erythropoietin (EPO) is a glycoprotein cytokine known for its pleiotropic effects on various types of cells and tissues. EPO and its receptor EPOR trigger signaling cascades JAK2/STAT5, MAPK, and PI3K/AKT that are interconnected and irreplaceable for cell survival. In this article, we describe the role of the MAPK and PI3K/AKT signaling pathways during red blood cell formation as well as in non-hematopoietic tissues and tumor cells. Although the central framework of these pathways is similar for most of cell types, there are some stage-specific, tissue, and cell-lineage differences. We summarize the current state of research in this field, highlight the novel members of EPO-induced PI3K and MAPK signaling, and in this respect also the differences between erythroid and non-erythroid cells.


Subject(s)
Erythropoiesis/physiology , Erythropoietin/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Humans , MAP Kinase Signaling System , Models, Biological , Neoplasms/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Erythropoietin/physiology , Signal Transduction
9.
Nat Chem Biol ; 9(12): 840-848, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24161946

ABSTRACT

Efforts to develop more effective therapies for acute leukemia may benefit from high-throughput screening systems that reflect the complex physiology of the disease, including leukemia stem cells (LSCs) and supportive interactions with the bone marrow microenvironment. The therapeutic targeting of LSCs is challenging because LSCs are highly similar to normal hematopoietic stem and progenitor cells (HSPCs) and are protected by stromal cells in vivo. We screened 14,718 compounds in a leukemia-stroma co-culture system for inhibition of cobblestone formation, a cellular behavior associated with stem-cell function. Among those compounds that inhibited malignant cells but spared HSPCs was the cholesterol-lowering drug lovastatin. Lovastatin showed anti-LSC activity in vitro and in an in vivo bone marrow transplantation model. Mechanistic studies demonstrated that the effect was on target, via inhibition of HMG-CoA reductase. These results illustrate the power of merging physiologically relevant models with high-throughput screening.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Leukemia , Neoplastic Stem Cells/drug effects , Cell Line, Tumor , Hematopoietic Stem Cells , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/physiology
10.
Cancer Cell ; 12(4): 367-80, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17936561

ABSTRACT

Despite their known transforming properties, the effects of leukemogenic FLT3-ITD mutations on hematopoietic stem and multipotent progenitor cells and on hematopoietic differentiation are not well understood. We report a mouse model harboring an ITD in the murine Flt3 locus that develops myeloproliferative disease resembling CMML and further identified FLT3-ITD mutations in a subset of human CMML. These findings correlated with an increase in number, cell cycling, and survival of multipotent stem and progenitor cells in an ITD dose-dependent manner in animals that exhibited alterations within their myeloid progenitor compartments and a block in normal B cell development. This model provides insights into the consequences of constitutive signaling by an oncogenic tyrosine kinase on hematopoietic progenitor quiescence, function, and cell fate.


Subject(s)
Cell Proliferation , Hematopoietic Stem Cells/metabolism , Leukemia, Myelomonocytic, Chronic/metabolism , Multipotent Stem Cells/metabolism , Mutation , Myeloproliferative Disorders/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Cell Differentiation , Cell Survival , Cells, Cultured , Exons , Gene Expression Regulation, Neoplastic , Genotype , Hematopoietic Stem Cells/pathology , Humans , Kaplan-Meier Estimate , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Multipotent Stem Cells/pathology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Phenotype , Signal Transduction , fms-Like Tyrosine Kinase 3/genetics
11.
J Intensive Care Med ; 30(7): 401-12, 2015 Oct.
Article in English | MEDLINE | ID: mdl-24407034

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) comprises a heterogeneous group of diseases that are characterized by a hyperinflammatory state due to uncontrolled T cell, macrophage, and histiocyte activation, accompanied by excessive cytokine production. This rare condition is almost uniformly fatal unless promptly recognized and treated. Much progress has been made in the last two decades in our understanding of the mechanisms underlying familial, and to a lesser extent, acquired cases of HLH. Recurrent mutations in more than 10 different genes have now been identified, involving biological pathways converging on intracellular vesicle trafficking and cytolytic granule exocytosis. Mechanisms underlying the majority of patients with acquired HLH, however, remain elusive, hampering both diagnostic evaluation and therapeutic management of these patients. Given that the majority of intensive care unit (ICU) patients with sepsis or multiorgan failure share many features of HLH, it is especially critical for pediatric and adult intensivists to be able to recognize patients with bona fide HLH and initiate treatment without delay. In this article, we review our current understanding of the pathophysiology, clinical testing, diagnosis, and treatment of patients with HLH, especially as it pertains to the care of critically ill patients in pediatric and medical ICUs.


Subject(s)
Critical Care , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/therapy , Antineoplastic Agents/therapeutic use , Diagnosis, Differential , Hematopoietic Stem Cell Transplantation , Humans , Immunosuppressive Agents/therapeutic use , Intensive Care Units , Lymphohistiocytosis, Hemophagocytic/classification , Lymphohistiocytosis, Hemophagocytic/physiopathology , Salvage Therapy
12.
BMC Complement Med Ther ; 24(1): 96, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383414

ABSTRACT

DNA topoisomerases regulate conformational changes in DNA topology during normal cell growth, such as replication, transcription, recombination, and repair, and may be targeted for anticancer drugs. A DNA topology assay was used to investigate DNA-damaging/protective activities of extracts from Habanero Red (HR), Habanero Maya Red (HMR), Trinidad Moruga Scorpion (TMS), Jalapeno (J), Serrano pepper (SP), Habanero Red Savina (HRS), Bhut Jolokia (BJ), and Jamaica Rosso (JR) peppers, demonstrating their inhibitory effect on the relaxation of pBR by Topo I. DNA topoisomerase II (Topo II) is proven therapeutic target of anticancer drugs. Complete inhibition of Topo II was observed for samples TMS, HR, and HMR. Extracts J and SP had the lowest capsaicin and dihydrocapsaicin content compared to other peppers. HR, HMR, TMS, J, S, HRS, BJ, JR extracts showed the anticancer effect, examined by MTS and xCell assay on the in vitro culture of human colon carcinoma cell line HCT116.


Subject(s)
Antineoplastic Agents , Capsaicin/analogs & derivatives , Capsicum , Humans , Capsaicin/pharmacology , Capsicum/genetics , Capsicum/metabolism , Antineoplastic Agents/pharmacology , DNA
13.
Nat Rev Cancer ; 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39384951

ABSTRACT

Splicing factors are affected by recurrent somatic mutations and copy number variations in several types of haematologic and solid malignancies, which is often seen as prima facie evidence that splicing aberrations can drive cancer initiation and progression. However, numerous spliceosome components also 'moonlight' in DNA repair and other cellular processes, making their precise role in cancer difficult to pinpoint. Still, few would deny that dysregulated mRNA splicing is a pervasive feature of most cancers. Correctly interpreting these molecular fingerprints can reveal novel tumour vulnerabilities and untapped therapeutic opportunities. Yet multiple technological challenges, lingering misconceptions, and outstanding questions hinder clinical translation. To start with, the general landscape of splicing aberrations in cancer is not well defined, due to limitations of short-read RNA sequencing not adept at resolving complete mRNA isoforms, as well as the shallow read depth inherent in long-read RNA-sequencing, especially at single-cell level. Although individual cancer-associated isoforms are known to contribute to cancer progression, widespread splicing alterations could be an equally important and, perhaps, more readily actionable feature of human cancers. This is to say that in addition to 'repairing' mis-spliced transcripts, possible therapeutic avenues include exacerbating splicing aberration with small-molecule spliceosome inhibitors, targeting recurrent splicing aberrations with synthetic lethal approaches, and training the immune system to recognize splicing-derived neoantigens.

14.
Leukemia ; 38(9): 1992-2002, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39033241

ABSTRACT

Mutations in the cohesin complex components (STAG2, RAD21, SMC1A, SMC3, and PDS5B) are recurrent genetic drivers in myelodysplastic neoplasm (MDS) and acute myeloid leukemia (AML). Whether the different cohesin subunit mutations share clinical characteristics and prognostic significance is not known. We analyzed 790 cohesin-mutant patients from the Dana-Farber Cancer Institute (DFCI) and the Munich Leukemia Laboratory (MLL), 390 of which had available outcome data, and identified subunit-specific clinical, prognostic, and genetic characteristics suggestive of distinct ontogenies. We found that STAG2 mutations are acquired at MDS stage and are associated with secondary AML, adverse prognosis, and co-occurrence of secondary AML-type mutations. In contrast, mutations in RAD21, SMC1A and SMC3 share features with de novo AML with better prognosis, and co-occurrence with de novo AML-type lesions. The findings show the heterogeneous nature of cohesin complex mutations, and inform clinical and prognostic classification, as well as distinct biology of the cohesin complex.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Cohesins , Leukemia, Myeloid, Acute , Mutation , Myelodysplastic Syndromes , Humans , Chromosomal Proteins, Non-Histone/genetics , Cell Cycle Proteins/genetics , Prognosis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Male , Female , Middle Aged , Aged , Adult , Nuclear Proteins/genetics , Phosphoproteins/genetics , DNA-Binding Proteins/genetics , Aged, 80 and over , Chondroitin Sulfate Proteoglycans
15.
Sci Transl Med ; 16(728): eade2774, 2024 01 03.
Article in English | MEDLINE | ID: mdl-38170787

ABSTRACT

Splicing modulation is a promising treatment strategy pursued to date only in splicing factor-mutant cancers; however, its therapeutic potential is poorly understood outside of this context. Like splicing factors, genes encoding components of the cohesin complex are frequently mutated in cancer, including myelodysplastic syndromes (MDS) and secondary acute myeloid leukemia (AML), where they are associated with poor outcomes. Here, we showed that cohesin mutations are biomarkers of sensitivity to drugs targeting the splicing factor 3B subunit 1 (SF3B1) H3B-8800 and E-7107. We identified drug-induced alterations in splicing, and corresponding reduced gene expression, of a number of DNA repair genes, including BRCA1 and BRCA2, as the mechanism underlying this sensitivity in cell line models, primary patient samples and patient-derived xenograft (PDX) models of AML. We found that DNA damage repair genes are particularly sensitive to exon skipping induced by SF3B1 modulators due to their long length and large number of exons per transcript. Furthermore, we demonstrated that treatment of cohesin-mutant cells with SF3B1 modulators not only resulted in impaired DNA damage response and accumulation of DNA damage, but it sensitized cells to subsequent killing by poly(ADP-ribose) polymerase (PARP) inhibitors and chemotherapy and led to improved overall survival of PDX models of cohesin-mutant AML in vivo. Our findings expand the potential therapeutic benefits of SF3B1 splicing modulators to include cohesin-mutant MDS and AML.


Subject(s)
Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Humans , Cohesins , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , RNA Splicing , RNA Splicing Factors/genetics , Mutation/genetics , Transcription Factors/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , DNA Repair/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , DNA Damage
16.
Blood ; 118(5): 1264-73, 2011 Aug 04.
Article in English | MEDLINE | ID: mdl-21653327

ABSTRACT

The NOTCH signaling pathway is implicated in a broad range of developmental processes, including cell fate decisions. However, the molecular basis for its role at the different steps of stem cell lineage commitment is unclear. We recently identified the NOTCH signaling pathway as a positive regulator of megakaryocyte lineage specification during hematopoiesis, but the developmental pathways that allow hematopoietic stem cell differentiation into the erythro-megakaryocytic lineages remain controversial. Here, we investigated the role of downstream mediators of NOTCH during megakaryopoiesis and report crosstalk between the NOTCH and PI3K/AKT pathways. We demonstrate the inhibitory role of phosphatase with tensin homolog and Forkhead Box class O factors on megakaryopoiesis in vivo. Finally, our data annotate developmental mechanisms in the hematopoietic system that enable a decision to be made either at the hematopoietic stem cell or the committed progenitor level to commit to the megakaryocyte lineage, supporting the existence of 2 distinct developmental pathways.


Subject(s)
Cell Differentiation , Cell Lineage/physiology , Megakaryocytes/physiology , Oncogene Protein v-akt/metabolism , Receptors, Notch/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage/genetics , Cells, Cultured , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/physiology , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/physiology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/physiology , Receptor Cross-Talk/physiology , Receptors, Notch/genetics , Receptors, Notch/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Thrombopoiesis/genetics
17.
Curr Opin Oncol ; 24(4): 409-13, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22664824

ABSTRACT

PURPOSE OF REVIEW: Inflammatory myofibroblastic tumors (IMTs) are indolent mesenchymal neoplasms associated with a small risk of aggressive behavior and metastasis. Surgery is the mainstay of treatment and until recently there have been limited effective treatment options for unresectable disease. This review describes the identification of anaplastic lymphoma kinase (ALK) fusion genes in approximately 50% of IMTs and the role of ALK inhibition in the treatment of this disease. RECENT FINDINGS: A recent phase I dose-escalation trial of the selective MET/ALK inhibitor crizotinib showed a long-term partial response in a patient with IMT carrying an ALK translocation but not in a patient with ALK-negative disease. Emergence of resistance to crizotinib occurs approximately 5-8 months after initiation of therapy and has been shown to be driven by different mechanisms. Multiple second-generation ALK inhibitors are currently being investigated in the preclinical and clinical trial setting. SUMMARY: ALK-directed therapy has emerged as a highly effective treatment option for a subset of patients with IMT and pulmonary adenocarcinoma. A number of additional malignancies, including rhabdomyosarcoma, neuroblastoma, anaplastic large cell lymphoma, renal cell carcinoma, and inflammatory breast cancer, have been shown to activate ALK expression by means of ALK fusion proteins, ALK mutations, or increased ALK copy number. Development of more selective ALK inhibitors, which can overcome emergent crizotinib resistance mutations, as well as development of combination treatments with drugs targeting compensatory pathways, will be key to achieving therapeutic success in targeting this potent and prevalent oncogenic driver.


Subject(s)
Granuloma, Plasma Cell/enzymology , Granuloma, Plasma Cell/therapy , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Anaplastic Lymphoma Kinase , Gene Fusion , Granuloma, Plasma Cell/drug therapy , Granuloma, Plasma Cell/genetics , Humans , Receptor Protein-Tyrosine Kinases/biosynthesis
18.
Plants (Basel) ; 11(9)2022 May 06.
Article in English | MEDLINE | ID: mdl-35567257

ABSTRACT

The present article aimed to study the effects of four selected concentrations (1%, 2%, 5%, and 10%) of apple pomace powder (APP), obtained from juice production, on the nutritional value and selected physico-chemical, antioxidant, and sensory properties of wheat bread. We have found that the ash and total carbohydrate contents, total polyphenols content, and antioxidant activity of the supplemented bread loaves were markedly higher (p < 0.05) as compared to the control ones. On the other hand, values for protein and fat contents and loaf volume in APP-containing bread samples were statistically lower (p < 0.05). Finally, sensory evaluation revealed no significant differences in all tested attributes between the investigated groups of bread samples. The current results suggest that 10% APP addition appears to be an attractive ingredient applied to bread formulation to obtain a bakery product with high nutritional value and required qualitative and sensory properties. In such a manner, apple pomace as by-products from apple juice processing can be efficiently utilized in an eco-friendly way by the food industry to decrease unnecessary waste and environmental pollution.

19.
Nat Commun ; 13(1): 5495, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36127368

ABSTRACT

Conditional degron tags (CDTs) are a powerful tool for target validation that combines the kinetics and reversible action of pharmacological agents with the generalizability of genetic manipulation. However, successful design of a CDT fusion protein often requires a prolonged, ad hoc cycle of construct design, failure, and re-design. To address this limitation, we report here a system to rapidly compare the activity of five unique CDTs: AID/AID2, IKZF3d, dTAG, HaloTag, and SMASh. We demonstrate the utility of this system against 16 unique protein targets. We find that expression and degradation are highly dependent on the specific CDT, the construct design, and the target. None of the CDTs leads to efficient expression and/or degradation across all targets; however, our systematic approach enables the identification of at least one optimal CDT fusion for each target. To enable the adoption of CDT strategies more broadly, we have made these reagents, and a detailed protocol, available as a community resource.


Subject(s)
Proteolysis , Kinetics
20.
Nat Commun ; 12(1): 6241, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34716321

ABSTRACT

Precise control of gene expression during differentiation relies on the interplay of chromatin and nuclear structure. Despite an established contribution of nuclear membrane proteins to developmental gene regulation, little is known regarding the role of inner nuclear proteins. Here we demonstrate that loss of the nuclear scaffolding protein Matrin-3 (Matr3) in erythroid cells leads to morphological and gene expression changes characteristic of accelerated maturation, as well as broad alterations in chromatin organization similar to those accompanying differentiation. Matr3 protein interacts with CTCF and the cohesin complex, and its loss perturbs their occupancy at a subset of sites. Destabilization of CTCF and cohesin binding correlates with altered transcription and accelerated differentiation. This association is conserved in embryonic stem cells. Our findings indicate Matr3 negatively affects cell fate transitions and demonstrate that a critical inner nuclear protein impacts occupancy of architectural factors, culminating in broad effects on chromatin organization and cell differentiation.


Subject(s)
Chromatin/chemistry , Leukemia, Erythroblastic, Acute/pathology , Nuclear Matrix-Associated Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , CCCTC-Binding Factor , Cell Cycle Proteins/metabolism , Cell Differentiation/physiology , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Embryonic Stem Cells/physiology , Erythroid Cells/pathology , Leukemia, Erythroblastic, Acute/metabolism , Mice, Knockout , Nuclear Matrix-Associated Proteins/genetics , RNA-Binding Proteins/genetics , Cohesins
SELECTION OF CITATIONS
SEARCH DETAIL