Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 147
Filter
Add more filters

Publication year range
1.
Blood ; 142(1): 44-61, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37023372

ABSTRACT

In chronic lymphocytic leukemia (CLL), epigenetic alterations are considered to centrally shape the transcriptional signatures that drive disease evolution and underlie its biological and clinical subsets. Characterizations of epigenetic regulators, particularly histone-modifying enzymes, are very rudimentary in CLL. In efforts to establish effectors of the CLL-associated oncogene T-cell leukemia 1A (TCL1A), we identified here the lysine-specific histone demethylase KDM1A to interact with the TCL1A protein in B cells in conjunction with an increased catalytic activity of KDM1A. We demonstrate that KDM1A is upregulated in malignant B cells. Elevated KDM1A and associated gene expression signatures correlated with aggressive disease features and adverse clinical outcomes in a large prospective CLL trial cohort. Genetic Kdm1a knockdown in Eµ-TCL1A mice reduced leukemic burden and prolonged animal survival, accompanied by upregulated p53 and proapoptotic pathways. Genetic KDM1A depletion also affected milieu components (T, stromal, and monocytic cells), resulting in significant reductions in their capacity to support CLL-cell survival and proliferation. Integrated analyses of differential global transcriptomes (RNA sequencing) and H3K4me3 marks (chromatin immunoprecipitation sequencing) in Eµ-TCL1A vs iKdm1aKD;Eµ-TCL1A mice (confirmed in human CLL) implicate KDM1A as an oncogenic transcriptional repressor in CLL which alters histone methylation patterns with pronounced effects on defined cell death and motility pathways. Finally, pharmacologic KDM1A inhibition altered H3K4/9 target methylation and revealed marked anti-B-cell leukemic synergisms. Overall, we established the pathogenic role and effector networks of KDM1A in CLL via tumor-cell intrinsic mechanisms and its impacts in cells of the microenvironment. Our data also provide rationales to further investigate therapeutic KDM1A targeting in CLL.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Humans , Mice , Animals , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Histones/metabolism , Lysine , Prospective Studies , Histone Demethylases/genetics , Histone Demethylases/metabolism , Tumor Microenvironment
2.
Mod Pathol ; 37(5): 100466, 2024 May.
Article in English | MEDLINE | ID: mdl-38460674

ABSTRACT

This manuscript represents a review of lymphoblastic leukemia/lymphoma (acute lymphoblastic leukemia/lymphoblastic lymphoma), acute leukemias of ambiguous lineage, mixed-phenotype acute leukemias, myeloid/lymphoid neoplasms with eosinophilia and defining gene rearrangements, histiocytic and dendritic neoplasms, and genetic tumor syndromes of the 5th edition of the World Health Organization Classification of Tumors of the Hematopoietic and Lymphoid Tissues. The diagnostic, clinicopathologic, cytogenetic, and molecular genetic features are discussed. The differences in comparison to the 4th revised edition of the World Health Organization classification of hematolymphoid neoplasms are highlighted.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , World Health Organization , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/classification , Eosinophilia/pathology , Eosinophilia/genetics , Histiocytic Disorders, Malignant/genetics , Histiocytic Disorders, Malignant/pathology , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Hematologic Neoplasms/classification , Phenotype
3.
Immunity ; 43(5): 870-83, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26522984

ABSTRACT

Pan-NOTCH inhibitors are poorly tolerated in clinical trials because NOTCH signals are crucial for intestinal homeostasis. These inhibitors might also promote cancer because NOTCH can act as a tumor suppressor. We previously reported that the PIAS-like coactivator ZMIZ1 is frequently co-expressed with activated NOTCH1 in T cell acute lymphoblastic leukemia (T-ALL). Here, we show that similar to Notch1, Zmiz1 was important for T cell development and controlled the expression of certain Notch target genes, such as Myc. However, unlike Notch, Zmiz1 had no major role in intestinal homeostasis or myeloid suppression. Deletion of Zmiz1 impaired the initiation and maintenance of Notch-induced T-ALL. Zmiz1 directly interacted with Notch1 via a tetratricopeptide repeat domain at a special class of Notch-regulatory sites. In contrast to the Notch cofactor Maml, which is nonselective, Zmiz1 was selective. Thus, targeting the NOTCH1-ZMIZ1 interaction might combat leukemic growth while avoiding the intolerable toxicities of NOTCH inhibitors.


Subject(s)
Leukemia/metabolism , Protein Inhibitors of Activated STAT/metabolism , Receptor, Notch1/metabolism , T-Lymphocytes/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Cell Line, Tumor , Humans , Jurkat Cells , Leukemia/pathology , Mice , Mice, Inbred C57BL , Signal Transduction/physiology , T-Lymphocytes/pathology
4.
Nature ; 553(7687): 222-227, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323298

ABSTRACT

Chromosomal translocations that generate in-frame oncogenic gene fusions are notable examples of the success of targeted cancer therapies. We have previously described gene fusions of FGFR3-TACC3 (F3-T3) in 3% of human glioblastoma cases. Subsequent studies have reported similar frequencies of F3-T3 in many other cancers, indicating that F3-T3 is a commonly occuring fusion across all tumour types. F3-T3 fusions are potent oncogenes that confer sensitivity to FGFR inhibitors, but the downstream oncogenic signalling pathways remain unknown. Here we show that human tumours with F3-T3 fusions cluster within transcriptional subgroups that are characterized by the activation of mitochondrial functions. F3-T3 activates oxidative phosphorylation and mitochondrial biogenesis and induces sensitivity to inhibitors of oxidative metabolism. Phosphorylation of the phosphopeptide PIN4 is an intermediate step in the signalling pathway of the activation of mitochondrial metabolism. The F3-T3-PIN4 axis triggers the biogenesis of peroxisomes and the synthesis of new proteins. The anabolic response converges on the PGC1α coactivator through the production of intracellular reactive oxygen species, which enables mitochondrial respiration and tumour growth. These data illustrate the oncogenic circuit engaged by F3-T3 and show that F3-T3-positive tumours rely on mitochondrial respiration, highlighting this pathway as a therapeutic opportunity for the treatment of tumours with F3-T3 fusions. We also provide insights into the genetic alterations that initiate the chain of metabolic responses that drive mitochondrial metabolism in cancer.


Subject(s)
Cell Respiration , Microtubule-Associated Proteins/genetics , Mitochondria/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Oncogene Proteins, Fusion/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Cell Respiration/drug effects , Cell Transformation, Neoplastic/drug effects , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Mice , Mitochondria/drug effects , Mitochondria/genetics , NIMA-Interacting Peptidylprolyl Isomerase/chemistry , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Organelle Biogenesis , Oxidative Phosphorylation/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisomes/drug effects , Peroxisomes/metabolism , Phosphorylation , Protein Biosynthesis , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Transcription, Genetic , Xenograft Model Antitumor Assays
5.
Lab Invest ; 103(11): 100246, 2023 11.
Article in English | MEDLINE | ID: mdl-37659445

ABSTRACT

Digital pathology workflows can improve pathology operations by allowing reliable and fast retrieval of digital images, digitally reviewing pathology slides, enabling remote work and telepathology, use of computer-aided tools, and sharing of digital images for research and educational purposes. The need for quality systems is a prerequisite for successful clinical-grade digital pathology adoption and patient safety. In this article, we describe the development of a structured digital pathology laboratory quality management system (QMS) for clinical digital pathology operations at Memorial Sloan Kettering Cancer Center (MSK). This digital pathology-specific QMS development stemmed from the gaps that were identified when MSK integrated digital pathology into its clinical practice. The digital scan team in conjunction with the Department of Pathology and Laboratory Medicine quality team developed a QMS tailored to the scanning operation to support departmental and institutional needs. As a first step, systemic mapping of the digital pathology operations identified the prescan, scan, and postscan processes; instrumentation; and staffing involved in the digital pathology operation. Next, gaps identified in quality control and quality assurance measures led to the development of standard operating procedures and training material for the different roles and workflows in the process. All digital pathology-related documents were subject to regulatory review and approval by departmental leadership. The quality essentials were developed into an extensive Digital Pathology Quality Essentials framework to specifically address the needs of the growing clinical use of digital pathology technologies. Using the unique digital experience gained at MSK, we present our recommendations for QMS for large-scale digital pathology operations in clinical settings.


Subject(s)
Neoplasms , Pathology, Clinical , Telepathology , Humans , Laboratories , Neoplasms/diagnosis , Neoplasms/surgery , Pathology, Clinical/methods , Telepathology/methods , Total Quality Management
6.
Mod Pathol ; 36(7): 100170, 2023 07.
Article in English | MEDLINE | ID: mdl-36997001

ABSTRACT

High-grade B-cell lymphomas with 11q aberrations (HGBL-11q) represent a World Health Organization-defined group of lymphomas that harbor recurrent chromosome 11q aberrations involving proximal gains and telomeric losses. Although a limited number of HGBL-11q cases evaluated thus far appear to show a similar course and prognosis as Burkitt lymphoma (BL), many molecular differences have been appreciated, most notably the absence of MYC rearrangement. Despite biological differences between BL and HGBL-11q, histomorphologic and immunophenotypic distinction remains challenging. Here, we provide a comparative whole proteomic profile of BL- and HGBL-11q-derived cell lines, identifying numerous shared and differentially expressed proteins. Transcriptome profiling performed on paraffin-embedded tissue samples from primary BL and HGBL-11q lymphomas was additionally performed to provide further molecular characterization. Overlap of proteomic and transcriptomic data sets identified several potential novel biomarkers of HGBL-11q, including diminished lymphoid enhancer-binding factor 1 expression, which was validated by immunohistochemistry staining in a cohort of 23 cases. Altogether, these findings provide a comprehensive multimodal and comparative molecular profiling of BL and HGBL-11q and suggest the use of enhancer-binding factor 1 as an immunohistochemistry target to distinguish between these aggressive lymphomas.


Subject(s)
Burkitt Lymphoma , Lymphoma, B-Cell , Lymphoma, Large B-Cell, Diffuse , Proteogenomics , Humans , Burkitt Lymphoma/genetics , Burkitt Lymphoma/pathology , Lymphoid Enhancer-Binding Factor 1 , Proteomics , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Chromosome Aberrations , Biomarkers , Lymphoma, Large B-Cell, Diffuse/pathology
7.
EMBO J ; 36(21): 3232-3249, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29030483

ABSTRACT

Notch signaling is an evolutionarily conserved signal transduction pathway that is essential for metazoan development. Upon ligand binding, the Notch intracellular domain (NOTCH ICD) translocates into the nucleus and forms a complex with the transcription factor RBPJ (also known as CBF1 or CSL) to activate expression of Notch target genes. In the absence of a Notch signal, RBPJ acts as a transcriptional repressor. Using a proteomic approach, we identified L3MBTL3 (also known as MBT1) as a novel RBPJ interactor. L3MBTL3 competes with NOTCH ICD for binding to RBPJ In the absence of NOTCH ICD, RBPJ recruits L3MBTL3 and the histone demethylase KDM1A (also known as LSD1) to the enhancers of Notch target genes, leading to H3K4me2 demethylation and to transcriptional repression. Importantly, in vivo analyses of the homologs of RBPJ and L3MBTL3 in Drosophila melanogaster and Caenorhabditis elegans demonstrate that the functional link between RBPJ and L3MBTL3 is evolutionarily conserved, thus identifying L3MBTL3 as a universal modulator of Notch signaling in metazoans.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Histone Demethylases/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Neuroglia/metabolism , Receptors, Notch/genetics , Animals , Biological Evolution , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cell Line, Tumor , Conserved Sequence , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Neuroglia/cytology , Protein Binding , Protein Domains , Receptors, Notch/metabolism , Transcription, Genetic , Two-Hybrid System Techniques
8.
Blood ; 131(21): 2320-2330, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29666117

ABSTRACT

T-cell lymphomas are a heterogeneous group of rare malignancies with overlapping clinical, immunologic, and histologic features. Recent advances in our understanding of T-cell differentiation based on gene expression profiling, next-generation sequencing, and transgenic mouse modeling studies have better elucidated the pathogenetic mechanisms underlying the diverse biology of T-cell lymphomas. These studies show that although genetic alterations in epigenetic modifiers are implicated in all subtypes of T-cell lymphomas, specific subtypes demonstrate enrichment for particular recurrent alterations targeting specific genes. In this regard, RHOA and TET2 alterations are prevalent in nodal T-cell lymphomas, particularly angioimmunoblastic T-cell lymphomas, peripheral T-cell lymphomas (PTCLs) not otherwise specified, and nodal PTCLs with T-follicular helper phenotype. JAK-STAT signaling pathways are mutationally activated in many extranodal T-cell lymphomas, such as natural killer/T-cell and hepatosplenic T-cell lymphomas. The functional significance of many of these genetic alterations is becoming better understood. Altogether these advances will continue to refine diagnostic criteria, improve prognostication, and identify novel therapeutic targets, resulting in improved outcomes for patient with T-cell lymphomas.


Subject(s)
Lymphoma, T-Cell/etiology , Animals , Biomarkers , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Susceptibility , Energy Metabolism , Gene Expression Regulation, Neoplastic , Humans , Lymphoma, T-Cell/diagnosis , Lymphoma, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
10.
Blood ; 132(20): 2115-2124, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30181172

ABSTRACT

Castleman disease (CD) describes a group of heterogeneous hematologic disorders with characteristic histopathological features. CD can present with unicentric or multicentric (MCD) regions of lymph node enlargement. Some cases of MCD are caused by human herpesvirus-8 (HHV-8), whereas others are HHV-8-negative/idiopathic (iMCD). Treatment of iMCD is challenging, and outcomes can be poor because no uniform treatment guidelines exist, few systematic studies have been conducted, and no agreed upon response criteria have been described. The purpose of this paper is to establish consensus, evidence-based treatment guidelines based on the severity of iMCD to improve outcomes. An international Working Group of 42 experts from 10 countries was convened by the Castleman Disease Collaborative Network to establish consensus guidelines for the management of iMCD based on published literature, review of treatment effectiveness for 344 cases, and expert opinion. The anti-interleukin-6 monoclonal antibody siltuximab (or tocilizumab, if siltuximab is not available) with or without corticosteroids is the preferred first-line therapy for iMCD. In the most severe cases, adjuvant combination chemotherapy is recommended. Additional agents are recommended, tailored by disease severity, as second- and third-line therapies for treatment failures. Response criteria were formulated to facilitate the evaluation of treatment failure or success. These guidelines should help treating physicians to stratify patients based on disease severity in order to select the best available therapeutic option. An international registry for patients with CD (ACCELERATE, #NCT02817997) was established in October 2016 to collect patient outcomes to increase the evidence base for selection of therapies in the future.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Castleman Disease/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Castleman Disease/pathology , Castleman Disease/therapy , Clinical Trials as Topic , Critical Illness/therapy , Disease Management , Evidence-Based Medicine , Humans , Practice Guidelines as Topic
11.
Semin Diagn Pathol ; 37(1): 72-78, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31917110

ABSTRACT

Mature T-cell and NK-cell leukemias represent a clinically heterogeneous group of diseases, ranging from indolent expansions of large granular lymphocytes, to aggressive diseases that are associated with a fulminant clinical course. Recent advances in genomic methodologies have massively increased the understanding of the pathogenesis of this group of diseases. While the entities are genetically heterogeneous, JAK-STAT pathway activation appears to be important across these disorders. The identification of constitutively activated pathways and the emergence of novel targeted pharmaceutical agents raise the expectation that more effective therapies will be identified for these disorders in the coming years.


Subject(s)
Leukemia, T-Cell , Humans
12.
Proc Natl Acad Sci U S A ; 114(25): 6581-6586, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28607076

ABSTRACT

Identification of biomarkers and therapeutic targets is a critical goal of precision medicine. N-glycoproteins are a particularly attractive class of proteins that constitute potential cancer biomarkers and therapeutic targets for small molecules, antibodies, and cellular therapies. Using mass spectrometry (MS), we generated a compendium of 1,091 N-glycoproteins (from 40 human primary lymphomas and cell lines). Hierarchical clustering revealed distinct subtype signatures that included several subtype-specific biomarkers. Orthogonal immunological studies in 671 primary lymphoma tissue biopsies and 32 lymphoma-derived cell lines corroborated MS data. In anaplastic lymphoma kinase-positive (ALK+) anaplastic large cell lymphoma (ALCL), integration of N-glycoproteomics and transcriptome sequencing revealed an ALK-regulated cytokine/receptor signaling network, including vulnerabilities corroborated by a genome-wide clustered regularly interspaced short palindromic screen. Functional targeting of IL-31 receptor ß, an ALCL-enriched and ALK-regulated N-glycoprotein in this network, abrogated ALK+ALCL growth in vitro and in vivo. Our results highlight the utility of functional proteogenomic approaches for discovery of cancer biomarkers and therapeutic targets.


Subject(s)
Biomarkers, Tumor/genetics , Lymphoma/genetics , Transcriptome/genetics , Cell Line, Tumor , Humans , Proteogenomics/methods , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics
13.
Oncologist ; 24(10): 1287-1290, 2019 10.
Article in English | MEDLINE | ID: mdl-31366725

ABSTRACT

Drawing on discussions at a workshop hosted by the National Cancer Policy Forum, current challenges in pathology are reviewed and practical steps to facilitate high­quality cancer diagnosis and care through improved patient access to expertise in oncologic pathology are highlighted.


Subject(s)
Medical Oncology/methods , Neoplasms/diagnosis , Neoplasms/therapy , Quality of Health Care/standards , Humans
15.
Blood ; 129(12): 1646-1657, 2017 Mar 23.
Article in English | MEDLINE | ID: mdl-28087540

ABSTRACT

Human herpesvirus-8 (HHV-8)-negative, idiopathic multicentric Castleman disease (iMCD) is a rare and life-threatening disorder involving systemic inflammatory symptoms, polyclonal lymphoproliferation, cytopenias, and multiple organ system dysfunction caused by a cytokine storm often including interleukin-6. iMCD accounts for one third to one half of all cases of MCD and can occur in individuals of any age. Accurate diagnosis is challenging, because no standard diagnostic criteria or diagnostic biomarkers currently exist, and there is significant overlap with malignant, autoimmune, and infectious disorders. An international working group comprising 34 pediatric and adult pathology and clinical experts in iMCD and related disorders from 8 countries, including 2 physicians that are also iMCD patients, was convened to establish iMCD diagnostic criteria. The working group reviewed data from 244 cases, met twice, and refined criteria over 15 months (June 2015 to September 2016). The proposed consensus criteria require both Major Criteria (characteristic lymph node histopathology and multicentric lymphadenopathy), at least 2 of 11 Minor Criteria with at least 1 laboratory abnormality, and exclusion of infectious, malignant, and autoimmune disorders that can mimic iMCD. Characteristic histopathologic features may include a constellation of regressed or hyperplastic germinal centers, follicular dendritic cell prominence, hypervascularization, and polytypic plasmacytosis. Laboratory and clinical Minor Criteria include elevated C-reactive protein or erythrocyte sedimentation rate, anemia, thrombocytopenia or thrombocytosis, hypoalbuminemia, renal dysfunction or proteinuria, polyclonal hypergammaglobulinemia, constitutional symptoms, hepatosplenomegaly, effusions or edema, eruptive cherry hemangiomatosis or violaceous papules, and lymphocytic interstitial pneumonitis. iMCD consensus diagnostic criteria will facilitate consistent diagnosis, appropriate treatment, and collaborative research.


Subject(s)
Castleman Disease/diagnosis , Castleman Disease/etiology , Herpesvirus 8, Human , Consensus , Diagnosis, Differential , Humans , Internationality , Practice Guidelines as Topic
16.
Immunol Rev ; 263(1): 224-39, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25510280

ABSTRACT

Ubiquitination is a post-translational modification process that regulates several critical cellular processes. Ubiquitination is orchestrated by the ubiquitin proteasome system (UPS), which constitutes a cascade of enzymes that transfer ubiquitin onto protein substrates. The UPS catalyzes the destruction of many critical protein substrates involved in cancer pathogenesis. This review article focuses on components of the UPS that have been demonstrated to be deregulated by a variety of mechanisms in hematologic malignancies. These include E3 ubiquitin ligases and deubiquitinating enzymes. The prospects of specific targeting of key enzymes in this pathway that are critical to the pathogenesis of particular hematologic neoplasia are also discussed.


Subject(s)
Hematologic Neoplasms/enzymology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Carcinogenesis , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/immunology , Humans , Molecular Targeted Therapy , Ubiquitination
17.
Curr Opin Pulm Med ; 24(3): 281-286, 2018 05.
Article in English | MEDLINE | ID: mdl-29470255

ABSTRACT

PURPOSE OF REVIEW: Langerhans cell histiocytosis (LCH) is a neoplasm of dendritic cells with a wide clinical spectrum. Localized pulmonary LCH occurs in young adults with a history of smoking and can either resolve spontaneously or lead to progressive decline in pulmonary function. Young children can also present with localized disease - frequently bone or skin - or with multifocal or multisystem disease. Clinical outcomes in these patients also vary widely, ranging from spontaneous resolution to multiorgan failure and death. This review describes recent developments in our understanding of the underlying pathogenesis of LCH and how these discoveries and other research are affecting how the disease is classified, treated and monitored. RECENT FINDINGS: Somatic mutations resulting in activation of the mitogen-activated protein kinase (MAPK) pathway were recently identified as a key pathogenetic mechanism in both pediatric and pulmonary LCH. SUMMARY: Knowledge of underlying pathogenetic mechanisms of LCH transforming how this disease and other histocytic/dendritic disorders are classified, treated and monitored.


Subject(s)
Carcinogenesis/genetics , Histiocytosis, Langerhans-Cell/drug therapy , Histiocytosis, Langerhans-Cell/genetics , Humans , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/genetics , Mutation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics
18.
Mol Cell ; 38(6): 853-63, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20541448

ABSTRACT

MLL1 fusion proteins activate HoxA9 gene expression and cause aggressive leukemias that respond poorly to treatment, but how they recognize and stably bind to HoxA9 is not clearly understood. In a systematic analysis of MLL1 domain recruitment activity, we identified an essential MLL1 recruitment domain that includes the CXXC domain and PHD fingers and is controlled by direct interactions with the PAF elongation complex and H3K4Me2/3. MLL1 fusion proteins lack the PHD fingers and require prebinding of a wild-type MLL1 complex and CXXC domain recognition of DNA for stable HoxA9 association. Together, these results suggest that specific recruitment of MLL1 requires multiple interactions and is a precondition for stable recruitment of MLL1 fusion proteins to HoxA9 in leukemogenesis. Since wild-type MLL1 and oncogenic MLL1 fusion proteins have overlapping yet distinct recruitment mechanisms, this creates a window of opportunity that could be exploited for the development of targeted therapies.


Subject(s)
Homeodomain Proteins/metabolism , Leukemia/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Animals , Cell Line , Genetic Loci , Histone-Lysine N-Methyltransferase , Homeodomain Proteins/genetics , Humans , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/metabolism , Point Mutation , Protein Structure, Tertiary , Protein Transport , Transcription Factors
19.
J Biol Chem ; 291(6): 3030-42, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26601958

ABSTRACT

UBE2W ubiquitinates N termini of proteins rather than internal lysine residues, showing a preference for substrates with intrinsically disordered N termini. The in vivo functions of this intriguing E2, however, remain unknown. We generated Ube2w germ line KO mice that proved to be susceptible to early postnatal lethality without obvious developmental abnormalities. Although the basis of early death is uncertain, several organ systems manifest changes in Ube2w KO mice. Newborn Ube2w KO mice often show altered epidermal maturation with reduced expression of differentiation markers. Mirroring higher UBE2W expression levels in testis and thymus, Ube2w KO mice showed a disproportionate decrease in weight of these two organs (~50%), suggesting a functional role for UBE2W in the immune and male reproductive systems. Indeed, Ube2w KO mice displayed sustained neutrophilia accompanied by increased G-CSF signaling and testicular vacuolation associated with decreased fertility. Proteomic analysis of a vulnerable organ, presymptomatic testis, showed a preferential accumulation of disordered proteins in the absence of UBE2W, consistent with the view that UBE2W preferentially targets disordered polypeptides. These mice further allowed us to establish that UBE2W is ubiquitously expressed as a single isoform localized to the cytoplasm and that the absence of UBE2W does not alter cell viability in response to various stressors. Our results establish that UBE2W is an important, albeit not essential, protein for early postnatal survival and normal functioning of multiple organ systems.


Subject(s)
Epidermis , Skin Abnormalities , Ubiquitin-Conjugating Enzymes , Animals , Epidermis/abnormalities , Epidermis/enzymology , Epidermis/immunology , Leukocyte Disorders/congenital , Leukocyte Disorders/enzymology , Leukocyte Disorders/genetics , Leukocyte Disorders/immunology , Male , Mice , Mice, Knockout , Skin Abnormalities/enzymology , Skin Abnormalities/genetics , Skin Abnormalities/immunology , Testis/enzymology , Testis/immunology , Thymus Gland/enzymology , Thymus Gland/immunology , Ubiquitin-Conjugating Enzymes/deficiency , Ubiquitin-Conjugating Enzymes/immunology
20.
Lab Invest ; 97(8): 962-970, 2017 08.
Article in English | MEDLINE | ID: mdl-28414323

ABSTRACT

PKM2 (pyruvate kinase M2), a critical regulator of glycolysis, is phosphorylated by numerous growth factor receptors and oncogenic tyrosine kinases including NPM-ALK which is expressed in a subset of aggressive T-cell non-Hodgkin lymphomas known as anaplastic large cell lymphoma, ALK-positive. Our previous work demonstrated that phosphorylation of Y105-PKM2 by NPM-ALK regulates a major metabolic shift to promote lymphomagenesis. In addition to its role in metabolism, recent studies have shown that PKM2 promotes oncogenesis by phosphorylating nuclear STAT3 (signal transducer and activator of transcription 3) and regulating transcription of genes involved in cell survival and proliferation. We hypothesized that identification of novel PKM2 interactors could provide additional insights into its expanding functional role in cancer. To this end, immunocomplexes of FLAG-tagged PKM2 were isolated from NPM-ALK-positive ALCL (anaplastic large cell lymphoma) cells and subjected to liquid chromatography tandem mass spectrometry (LC-MS/MS) which led to the identification of polypyrimidine tract-binding protein (PTBP1) as a novel interactor of PKM2. The interaction between PTBP1 and PKM2 was restricted to the nucleus and was dependent on NPM-ALK mediated Y105 phosphorylation of PKM2. Stable shRNA-mediated silencing of PTBP1 resulted in a marked decrease in pY105-PKM2 and pY705-STAT3 which led to decreased ALCL cell proliferation and colony formation. Overall, our data demonstrate that PTBP1 interacts with PKM2 and promotes ALCL oncogenesis by facilitating PKM2-dependent activation of STAT3 within the nucleus.


Subject(s)
Carcinogenesis/metabolism , Carrier Proteins/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Lymphoma, Large-Cell, Anaplastic/metabolism , Membrane Proteins/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , STAT3 Transcription Factor/metabolism , Thyroid Hormones/metabolism , Cell Line, Tumor , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cell Proliferation , Cytoplasm/chemistry , Cytoplasm/metabolism , Humans , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Thyroid Hormone-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL