Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33.341
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 35: 533-550, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28182501

ABSTRACT

Common gamma receptor-dependent cytokines and their JAK/STAT pathways play pivotal roles in T cell immunity. Abnormal activation of this system was pervasive in diverse T cell malignancies assessed by pSTAT3/pSTAT5 phosphorylation. Activating mutations were described in some but not all cases. JAK1 and STAT3 were required for proliferation and survival of these T cell lines whether or not JAKs or STATs were mutated. Activating JAK and STAT mutations were not sufficient to initiate leukemic cell proliferation but rather only augmented signals from upstream in the cytokine pathway. Activation required the full pathway, including cytokine receptors acting as scaffolds and docking sites for required downstream JAK/STAT proteins. JAK kinase inhibitors have depressed leukemic T cell line proliferation. The insight that JAK/STAT system activation is pervasive in T cell malignancies suggests novel therapeutic approaches that include antibodies to common gamma cytokines, inhibitors of cytokine-receptor interactions, and JAK kinase inhibitors that may revolutionize therapy for T cell malignancies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Enzyme Inhibitors/therapeutic use , Immunotherapy/methods , Janus Kinases/metabolism , Lymphoma, T-Cell/immunology , STAT Transcription Factors/metabolism , Animals , Antineoplastic Agents/therapeutic use , Carcinogenesis , Cytokines/immunology , Cytokines/metabolism , Humans , Interleukin Receptor Common gamma Subunit/metabolism , Lymphoma, T-Cell/therapy , Receptors, Cytokine/antagonists & inhibitors , Signal Transduction
2.
Cell ; 184(10): 2649-2664.e18, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33848463

ABSTRACT

Receptor tyrosine kinase (RTK)-mediated activation of downstream effector pathways such as the RAS GTPase/MAP kinase (MAPK) signaling cascade is thought to occur exclusively from lipid membrane compartments in mammalian cells. Here, we uncover a membraneless, protein granule-based subcellular structure that can organize RTK/RAS/MAPK signaling in cancer. Chimeric (fusion) oncoproteins involving certain RTKs including ALK and RET undergo de novo higher-order assembly into membraneless cytoplasmic protein granules that actively signal. These pathogenic biomolecular condensates locally concentrate the RAS activating complex GRB2/SOS1 and activate RAS in a lipid membrane-independent manner. RTK protein granule formation is critical for oncogenic RAS/MAPK signaling output in these cells. We identify a set of protein granule components and establish structural rules that define the formation of membraneless protein granules by RTK oncoproteins. Our findings reveal membraneless, higher-order cytoplasmic protein assembly as a distinct subcellular platform for organizing oncogenic RTK and RAS signaling.


Subject(s)
Biomolecular Condensates/metabolism , Cytoplasmic Granules/metabolism , Neoplasms/metabolism , Oncogene Proteins, Fusion/metabolism , ras Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Enzyme Activation , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , HEK293 Cells , Humans , SOS1 Protein/metabolism , Signal Transduction
3.
Cell ; 181(6): 1246-1262.e22, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32442405

ABSTRACT

There is considerable inter-individual variability in susceptibility to weight gain despite an equally obesogenic environment in large parts of the world. Whereas many studies have focused on identifying the genetic susceptibility to obesity, we performed a GWAS on metabolically healthy thin individuals (lowest 6th percentile of the population-wide BMI spectrum) in a uniquely phenotyped Estonian cohort. We discovered anaplastic lymphoma kinase (ALK) as a candidate thinness gene. In Drosophila, RNAi mediated knockdown of Alk led to decreased triglyceride levels. In mice, genetic deletion of Alk resulted in thin animals with marked resistance to diet- and leptin-mutation-induced obesity. Mechanistically, we found that ALK expression in hypothalamic neurons controls energy expenditure via sympathetic control of adipose tissue lipolysis. Our genetic and mechanistic experiments identify ALK as a thinness gene, which is involved in the resistance to weight gain.


Subject(s)
Anaplastic Lymphoma Kinase/genetics , Thinness/genetics , Adipose Tissue/metabolism , Adult , Animals , Cell Line , Cohort Studies , Drosophila/genetics , Estonia , Female , Humans , Leptin/genetics , Lipolysis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , RNA Interference/physiology , Young Adult
4.
Cell ; 180(5): 878-894.e19, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32059783

ABSTRACT

Pathogenic autoantibodies arise in many autoimmune diseases, but it is not understood how the cells making them evade immune checkpoints. Here, single-cell multi-omics analysis demonstrates a shared mechanism with lymphoid malignancy in the formation of public rheumatoid factor autoantibodies responsible for mixed cryoglobulinemic vasculitis. By combining single-cell DNA and RNA sequencing with serum antibody peptide sequencing and antibody synthesis, rare circulating B lymphocytes making pathogenic autoantibodies were found to comprise clonal trees accumulating mutations. Lymphoma driver mutations in genes regulating B cell proliferation and V(D)J mutation (CARD11, TNFAIP3, CCND3, ID3, BTG2, and KLHL6) were present in rogue B cells producing the pathogenic autoantibody. Antibody V(D)J mutations conferred pathogenicity by causing the antigen-bound autoantibodies to undergo phase transition to insoluble aggregates at lower temperatures. These results reveal a pre-neoplastic stage in human lymphomagenesis and a cascade of somatic mutations leading to an iconic pathogenic autoantibody.


Subject(s)
Autoantibodies/genetics , Autoimmune Diseases/genetics , B-Lymphocytes/immunology , Lymphoma/genetics , Animals , Autoantibodies/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , B-Lymphocytes/pathology , CARD Signaling Adaptor Proteins/genetics , Carrier Proteins/genetics , Clonal Evolution/genetics , Clonal Evolution/immunology , Cyclin D3/genetics , Guanylate Cyclase/genetics , Humans , Immediate-Early Proteins/genetics , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Inhibitor of Differentiation Proteins/genetics , Lymphoma/immunology , Lymphoma/pathology , Mice , Mutation/genetics , Mutation/immunology , Neoplasm Proteins/genetics , Sequence Analysis, DNA/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Suppressor Proteins/genetics , V(D)J Recombination/genetics
5.
Cell ; 182(2): 297-316.e27, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32619424

ABSTRACT

The most aggressive B cell lymphomas frequently manifest extranodal distribution and carry somatic mutations in the poorly characterized gene TBL1XR1. Here, we show that TBL1XR1 mutations skew the humoral immune response toward generating abnormal immature memory B cells (MB), while impairing plasma cell differentiation. At the molecular level, TBL1XR1 mutants co-opt SMRT/HDAC3 repressor complexes toward binding the MB cell transcription factor (TF) BACH2 at the expense of the germinal center (GC) TF BCL6, leading to pre-memory transcriptional reprogramming and cell-fate bias. Upon antigen recall, TBL1XR1 mutant MB cells fail to differentiate into plasma cells and instead preferentially reenter new GC reactions, providing evidence for a cyclic reentry lymphomagenesis mechanism. Ultimately, TBL1XR1 alterations lead to a striking extranodal immunoblastic lymphoma phenotype that mimics the human disease. Both human and murine lymphomas feature expanded MB-like cell populations, consistent with a MB-cell origin and delineating an unforeseen pathway for malignant transformation of the immune system.


Subject(s)
Immunologic Memory/physiology , Lymphoma, Large B-Cell, Diffuse/pathology , Nuclear Proteins/genetics , Precursor Cells, B-Lymphoid/immunology , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/genetics , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Chromatin/chemistry , Chromatin/metabolism , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Histone Deacetylases/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 2/chemistry , Nuclear Receptor Co-Repressor 2/metabolism , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Protein Binding , Proto-Oncogene Proteins c-bcl-6/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Transcription, Genetic
6.
Immunity ; 57(2): 379-399.e18, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38301653

ABSTRACT

Palatine tonsils are secondary lymphoid organs (SLOs) representing the first line of immunological defense against inhaled or ingested pathogens. We generated an atlas of the human tonsil composed of >556,000 cells profiled across five different data modalities, including single-cell transcriptome, epigenome, proteome, and immune repertoire sequencing, as well as spatial transcriptomics. This census identified 121 cell types and states, defined developmental trajectories, and enabled an understanding of the functional units of the tonsil. Exemplarily, we stratified myeloid slan-like subtypes, established a BCL6 enhancer as locally active in follicle-associated T and B cells, and identified SIX5 as putative transcriptional regulator of plasma cell maturation. Analyses of a validation cohort confirmed the presence, annotation, and markers of tonsillar cell types and provided evidence of age-related compositional shifts. We demonstrate the value of this resource by annotating cells from B cell-derived mantle cell lymphomas, linking transcriptional heterogeneity to normal B cell differentiation states of the human tonsil.


Subject(s)
B-Lymphocytes , Palatine Tonsil , Humans , Adult , B-Lymphocytes/metabolism
7.
Immunity ; 56(12): 2816-2835.e13, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38091953

ABSTRACT

Cancer cells can evade natural killer (NK) cell activity, thereby limiting anti-tumor immunity. To reveal genetic determinants of susceptibility to NK cell activity, we examined interacting NK cells and blood cancer cells using single-cell and genome-scale functional genomics screens. Interaction of NK and cancer cells induced distinct activation and type I interferon (IFN) states in both cell types depending on the cancer cell lineage and molecular phenotype, ranging from more sensitive myeloid to less sensitive B-lymphoid cancers. CRISPR screens in cancer cells uncovered genes regulating sensitivity and resistance to NK cell-mediated killing, including adhesion-related glycoproteins, protein fucosylation genes, and transcriptional regulators, in addition to confirming the importance of antigen presentation and death receptor signaling pathways. CRISPR screens with a single-cell transcriptomic readout provided insight into underlying mechanisms, including regulation of IFN-γ signaling in cancer cells and NK cell activation states. Our findings highlight the diversity of mechanisms influencing NK cell susceptibility across different cancers and provide a resource for NK cell-based therapies.


Subject(s)
Hematologic Neoplasms , Neoplasms , Humans , Killer Cells, Natural , Neoplasms/genetics , Antigen Presentation , Genomics , Cytotoxicity, Immunologic/genetics , Cell Line, Tumor
8.
Cell ; 171(2): 481-494.e15, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28985567

ABSTRACT

Diffuse large B cell lymphoma (DLBCL) is the most common form of blood cancer and is characterized by a striking degree of genetic and clinical heterogeneity. This heterogeneity poses a major barrier to understanding the genetic basis of the disease and its response to therapy. Here, we performed an integrative analysis of whole-exome sequencing and transcriptome sequencing in a cohort of 1,001 DLBCL patients to comprehensively define the landscape of 150 genetic drivers of the disease. We characterized the functional impact of these genes using an unbiased CRISPR screen of DLBCL cell lines to define oncogenes that promote cell growth. A prognostic model comprising these genetic alterations outperformed current established methods: cell of origin, the International Prognostic Index comprising clinical variables, and dual MYC and BCL2 expression. These results comprehensively define the genetic drivers and their functional roles in DLBCL to identify new therapeutic opportunities in the disease.


Subject(s)
CRISPR-Cas Systems , Gene Expression Profiling , Lymphoma, Large B-Cell, Diffuse/genetics , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cells, Cultured , Exome , Female , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Male , Rituximab/administration & dosage
9.
Cell ; 171(1): 103-119.e18, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28938112

ABSTRACT

It is now established that Bcl11b specifies T cell fate. Here, we show that in developing T cells, the Bcl11b enhancer repositioned from the lamina to the nuclear interior. Our search for factors that relocalized the Bcl11b enhancer identified a non-coding RNA named ThymoD (thymocyte differentiation factor). ThymoD-deficient mice displayed a block at the onset of T cell development and developed lymphoid malignancies. We found that ThymoD transcription promoted demethylation at CTCF bound sites and activated cohesin-dependent looping to reposition the Bcl11b enhancer from the lamina to the nuclear interior and to juxtapose the Bcl11b enhancer and promoter into a single-loop domain. These large-scale changes in nuclear architecture were associated with the deposition of activating epigenetic marks across the loop domain, plausibly facilitating phase separation. These data indicate how, during developmental progression and tumor suppression, non-coding transcription orchestrates chromatin folding and compartmentalization to direct with high precision enhancer-promoter communication.


Subject(s)
Enhancer Elements, Genetic , Promoter Regions, Genetic , RNA, Untranslated/genetics , Repressor Proteins/genetics , T-Lymphocytes/cytology , Tumor Suppressor Proteins/genetics , Animals , CCCTC-Binding Factor , Chromatin/metabolism , Leukemia/genetics , Locus Control Region , Lymphoma/genetics , Mice , Nuclear Lamina/metabolism , Repressor Proteins/metabolism , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Transcription, Genetic
10.
Genes Dev ; 38(5-6): 253-272, 2024 04 17.
Article in English | MEDLINE | ID: mdl-38565249

ABSTRACT

Oncogenic activation of MYC in cancers predominantly involves increased transcription rather than coding region mutations. However, MYC-dependent lymphomas frequently acquire point mutations in the MYC phosphodegron, including at threonine 58 (T58), where phosphorylation permits binding via the FBW7 ubiquitin ligase triggering MYC degradation. To understand how T58 phosphorylation functions in normal cell physiology, we introduced an alanine mutation at T58 (T58A) into the endogenous c-Myc locus in the mouse germline. While MYC-T58A mice develop normally, lymphomas and myeloid leukemias emerge in ∼60% of adult homozygous T58A mice. We found that primitive hematopoietic progenitor cells from MYC-T58A mice exhibit aberrant self-renewal normally associated with hematopoietic stem cells (HSCs) and up-regulate a subset of MYC target genes important in maintaining stem/progenitor cell balance. In lymphocytes, genomic occupancy by MYC-T58A was increased at all promoters compared with WT MYC, while genes differentially expressed in a T58A-dependent manner were significantly more proximal to MYC-bound enhancers. MYC-T58A lymphocyte progenitors exhibited metabolic alterations and decreased activation of inflammatory and apoptotic pathways. Our data demonstrate that a single point mutation stabilizing MYC is sufficient to skew target gene expression, producing a profound gain of function in multipotential hematopoietic progenitors associated with self-renewal and initiation of lymphomas and leukemias.


Subject(s)
F-Box-WD Repeat-Containing Protein 7 , Hematologic Neoplasms , Lymphoma , Proto-Oncogene Proteins c-myc , Animals , Mice , Germ Cells/metabolism , Hematopoietic Stem Cells/metabolism , Point Mutation , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , F-Box-WD Repeat-Containing Protein 7/metabolism
11.
Immunity ; 54(8): 1788-1806.e7, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34166622

ABSTRACT

Lymphoid stromal cells (LSCs) are essential organizers of immune responses. We analyzed tonsillar tissue by combining flow cytometry, in situ imaging, RNA sequencing, and functional assays, defining three distinct human LSC subsets. The integrin CD49a designated perivascular stromal cells exhibiting features of local committed LSC precursors and segregated cytokine and chemokine-producing fibroblastic reticular cells (FRCs) supporting B and T cell survival. The follicular dendritic cell transcriptional profile reflected active responses to B cell and non-B cell stimuli. We therefore examined the effect of B cell stimuli on LSCs in follicular lymphoma (FL). FL B cells interacted primarily with CD49a+ FRCs. Transcriptional analyses revealed LSC reprogramming in situ downstream of the cytokines tumor necrosis factor (TNF) and transforming growth factor ß (TGF-ß), including increased expression of the chemokines CCL19 and CCL21. Our findings define human LSC populations in healthy tissue and reveal bidirectional crosstalk between LSCs and malignant B cells that may present a targetable axis in lymphoma.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Lymphoma, Follicular/immunology , Lymphoma, Follicular/pathology , Palatine Tonsil/immunology , Stromal Cells/immunology , Cells, Cultured , Chemokine CCL19/metabolism , Chemokine CCL21/metabolism , Humans , Integrin alpha1/metabolism , Palatine Tonsil/cytology , Signal Transduction/immunology , Stromal Cells/cytology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
Immunity ; 54(8): 1807-1824.e14, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34380064

ABSTRACT

The transcription factor forkhead box O1 (FOXO1), which instructs the dark zone program to direct germinal center (GC) polarity, is typically inactivated by phosphatidylinositol 3-kinase (PI3K) signals. Here, we investigated how FOXO1 mutations targeting this regulatory axis in GC-derived B cell non-Hodgkin lymphomas (B-NHLs) contribute to lymphomagenesis. Examination of primary B-NHL tissues revealed that FOXO1 mutations and PI3K pathway activity were not directly correlated. Human B cell lines bearing FOXO1 mutations exhibited hyperactivation of PI3K and Stress-activated protein kinase (SAPK)/Jun amino-terminal kinase (JNK) signaling, and increased cell survival under stress conditions as a result of alterations in FOXO1 transcriptional affinities and activation of transcriptional programs characteristic of GC-positive selection. When modeled in mice, FOXO1 mutations conferred competitive advantage to B cells in response to key T-dependent immune signals, disrupting GC homeostasis. FOXO1 mutant transcriptional signatures were prevalent in human B-NHL and predicted poor clinical outcomes. Thus, rather than enforcing FOXO1 constitutive activity, FOXO1 mutations enable co-option of GC-positive selection programs during the pathogenesis of GC-derived lymphomas.


Subject(s)
B-Lymphocytes/cytology , Forkhead Box Protein O1/genetics , Germinal Center/immunology , Lymphoma, B-Cell/pathology , Animals , B-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cell Proliferation/genetics , Cell Survival/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Lymphoma, B-Cell/genetics , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics , Signal Transduction/immunology
13.
Immunity ; 54(1): 116-131.e10, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33271120

ABSTRACT

Tumors frequently subvert major histocompatibility complex class I (MHC-I) peptide presentation to evade CD8+ T cell immunosurveillance, though how this is accomplished is not always well defined. To identify the global regulatory networks controlling antigen presentation, we employed genome-wide screening in human diffuse large B cell lymphomas (DLBCLs). This approach revealed dozens of genes that positively and negatively modulate MHC-I cell surface expression. Validated genes clustered in multiple pathways including cytokine signaling, mRNA processing, endosomal trafficking, and protein metabolism. Genes can exhibit lymphoma subtype- or tumor-specific MHC-I regulation, and a majority of primary DLBCL tumors displayed genetic alterations in multiple regulators. We established SUGT1 as a major positive regulator of both MHC-I and MHC-II cell surface expression. Further, pharmacological inhibition of two negative regulators of antigen presentation, EZH2 and thymidylate synthase, enhanced DLBCL MHC-I presentation. These and other genes represent potential targets for manipulating MHC-I immunosurveillance in cancers, infectious diseases, and autoimmunity.


Subject(s)
B-Lymphocytes/physiology , Biomarkers, Tumor/genetics , HLA Antigens/genetics , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class I/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Carcinogenesis/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Lineage , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Neoplastic , Genetic Testing , Genome-Wide Association Study , HLA Antigens/metabolism , Humans , Immunologic Surveillance , Lymphoma, Large B-Cell, Diffuse/metabolism , Tumor Escape/genetics
14.
Immunity ; 53(5): 952-970.e11, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33098766

ABSTRACT

Precise targeting of activation-induced cytidine deaminase (AID) to immunoglobulin (Ig) loci promotes antibody class switch recombination (CSR) and somatic hypermutation (SHM), whereas AID targeting of non-Ig loci can generate oncogenic DNA lesions. Here, we examined the contribution of G-quadruplex (G4) nucleic acid structures to AID targeting in vivo. Mice bearing a mutation in Aicda (AIDG133V) that disrupts AID-G4 binding modeled the pathology of hyper-IgM syndrome patients with an orthologous mutation, lacked CSR and SHM, and had broad defects in genome-wide AIDG133V chromatin localization. Genome-wide analyses also revealed that wild-type AID localized to MHCII genes, and AID expression correlated with decreased MHCII expression in germinal center B cells and diffuse large B cell lymphoma. Our findings indicate a crucial role for G4 binding in AID targeting and suggest that AID activity may extend beyond Ig loci to regulate the expression of genes relevant to the physiology and pathology of activated B cells.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , G-Quadruplexes , Hyper-IgM Immunodeficiency Syndrome/etiology , Hyper-IgM Immunodeficiency Syndrome/metabolism , Mutation , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Computational Biology/methods , Disease Models, Animal , Disease Susceptibility , Enzyme Activation , Fluorescent Antibody Technique , Gene Expression Profiling , Genome-Wide Association Study , Germinal Center/immunology , Germinal Center/metabolism , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Hyper-IgM Immunodeficiency Syndrome/diagnosis , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Immunophenotyping , Lymphocyte Activation/genetics , Lymphoma, Large B-Cell, Diffuse/etiology , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Transgenic
15.
CA Cancer J Clin ; 72(1): 78-93, 2022 01.
Article in English | MEDLINE | ID: mdl-34613616

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy is a promising immunotherapeutic treatment concept that is changing the treatment approach to hematologic malignancies. The development of CAR T-cell therapy represents a prime example for the successful bench-to-bedside translation of advances in immunology and cellular therapy into clinical practice. The currently available CAR T-cell products have shown high response rates and long-term remissions in patients with relapsed/refractory acute lymphoblastic leukemia and relapsed/refractory lymphoma. However, CAR T-cell therapy can induce severe life-threatening toxicities such as cytokine release syndrome, neurotoxicity, or infection, which require rapid and aggressive medical treatment in the intensive care unit setting. In this review, the authors provide an overview of the state-of-the-art in the clinical management of severe life-threatening events in CAR T-cell recipients. Furthermore, key challenges that have to be overcome to maximize the safety of CAR T cells are discussed.


Subject(s)
Critical Care/methods , Cytokine Release Syndrome/therapy , Hematologic Neoplasms/therapy , Immunotherapy, Adoptive/adverse effects , Neurotoxicity Syndromes/therapy , Cytokine Release Syndrome/immunology , Hematologic Neoplasms/immunology , Humans , Immunotherapy, Adoptive/methods , Neurotoxicity Syndromes/immunology , Receptors, Chimeric Antigen/immunology , Treatment Outcome
16.
Mol Cell ; 81(19): 4059-4075.e11, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34437837

ABSTRACT

DDX3X is a ubiquitously expressed RNA helicase involved in multiple stages of RNA biogenesis. DDX3X is frequently mutated in Burkitt lymphoma, but the functional basis for this is unknown. Here, we show that loss-of-function DDX3X mutations are also enriched in MYC-translocated diffuse large B cell lymphoma and reveal functional cooperation between mutant DDX3X and MYC. DDX3X promotes the translation of mRNA encoding components of the core translational machinery, thereby driving global protein synthesis. Loss-of-function DDX3X mutations moderate MYC-driven global protein synthesis, thereby buffering MYC-induced proteotoxic stress during early lymphomagenesis. Established lymphoma cells restore full protein synthetic capacity by aberrant expression of DDX3Y, a Y chromosome homolog, the expression of which is normally restricted to the testis. These findings show that DDX3X loss of function can buffer MYC-driven proteotoxic stress and highlight the capacity of male B cell lymphomas to then compensate for this loss by ectopic DDX3Y expression.


Subject(s)
B-Lymphocytes/enzymology , DEAD-box RNA Helicases/metabolism , Lymphoma, B-Cell/enzymology , Minor Histocompatibility Antigens/metabolism , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins c-myc/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , B-Lymphocytes/pathology , Cell Line, Tumor , Child , Child, Preschool , DEAD-box RNA Helicases/genetics , Endoplasmic Reticulum Stress , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Loss of Function Mutation , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Male , Mice, Transgenic , Middle Aged , Minor Histocompatibility Antigens/genetics , Neoplasm Proteins/genetics , Protein Biosynthesis , Proteome , Proteostasis , Proto-Oncogene Proteins c-myc/genetics , Young Adult
17.
Immunity ; 50(5): 1305-1316.e6, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30979688

ABSTRACT

Epstein-Barr virus (EBV) causes infectious mononucleosis and is associated with epithelial-cell cancers and B cell lymphomas. An effective EBV vaccine is not available. We found that antibodies to the EBV glycoprotein gH/gL complex were the principal components in human plasma that neutralized infection of epithelial cells and that antibodies to gH/gL and gp42 contributed to B cell neutralization. Immunization of mice and nonhuman primates with nanoparticle vaccines that displayed components of the viral-fusion machinery EBV gH/gL or gH/gL/gp42 elicited antibodies that potently neutralized both epithelial-cell and B cell infection. Immune serum from nonhuman primates inhibited EBV-glycoprotein-mediated fusion of epithelial cells and B cells and targeted an epitope critical for virus-cell fusion. Therefore, unlike the leading EBV gp350 vaccine candidate, which only protects B cells from infection, these EBV nanoparticle vaccines elicit antibodies that inhibit the virus-fusion apparatus and provide cell-type-independent protection from virus infection.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Epithelial Cells/immunology , Epstein-Barr Virus Infections/prevention & control , Herpesvirus 4, Human/immunology , Membrane Glycoproteins/immunology , Viral Envelope Proteins/immunology , Animals , B-Lymphocytes/virology , CHO Cells , Cell Fusion , Cell Line, Tumor , Cricetulus , Epithelial Cells/virology , Epstein-Barr Virus Infections/immunology , Female , HEK293 Cells , HeLa Cells , Humans , Immune Sera/administration & dosage , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Vaccines, Virus-Like Particle/immunology , Viral Vaccines/immunology , Virus Attachment
18.
Immunity ; 51(3): 535-547.e9, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31519498

ABSTRACT

Inactivating mutations of the CREBBP and EP300 acetyltransferases are among the most common genetic alterations in diffuse large B cell lymphoma (DLBCL) and follicular lymphoma (FL). Here, we examined the relationship between these two enzymes in germinal center (GC) B cells, the normal counterpart of FL and DLBCL, and in lymphomagenesis by using conditional GC-directed deletion mouse models targeting Crebbp or Ep300. We found that CREBBP and EP300 modulate common as well as distinct transcriptional programs implicated in separate anatomic and functional GC compartments. Consistently, deletion of Ep300 but not Crebbp impaired the fitness of GC B cells in vivo. Combined loss of Crebbp and Ep300 completely abrogated GC formation, suggesting that these proteins partially compensate for each other through common transcriptional targets. This synthetic lethal interaction was retained in CREBBP-mutant DLBCL cells and could be pharmacologically targeted with selective small molecule inhibitors of CREBBP and EP300 function. These data provide proof-of-principle for the clinical development of EP300-specific inhibitors in FL and DLBCL.


Subject(s)
B-Lymphocytes/physiology , CREB-Binding Protein/genetics , E1A-Associated p300 Protein/genetics , Epigenesis, Genetic/genetics , Germinal Center/physiology , Lymphoma, Follicular/etiology , Lymphoma, Large B-Cell, Diffuse/genetics , Acetyltransferases/genetics , Animals , Cell Line , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Sequence Deletion/genetics , Transcription, Genetic/genetics
19.
Mol Cell ; 80(5): 845-861.e10, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33232656

ABSTRACT

Locus control region (LCR) functions define cellular identity and have critical roles in diseases such as cancer, although the hierarchy of structural components and associated factors that drive functionality are incompletely understood. Here we show that OCA-B, a B cell-specific coactivator essential for germinal center (GC) formation, forms a ternary complex with the lymphoid-enriched OCT2 and GC-specific MEF2B transcription factors and that this complex occupies and activates an LCR that regulates the BCL6 proto-oncogene and is uniquely required by normal and malignant GC B cells. Mechanistically, through OCA-B-MED1 interactions, this complex is required for Mediator association with the BCL6 promoter. Densely tiled CRISPRi screening indicates that only LCR segments heavily bound by this ternary complex are essential for its function. Our results demonstrate how an intimately linked complex of lineage- and stage-specific factors converges on specific and highly essential enhancer elements to drive the function of a cell-type-defining LCR.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Locus Control Region/immunology , Animals , B-Lymphocytes/cytology , Cell Line, Tumor , Germinal Center/cytology , HEK293 Cells , Humans , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/immunology , Mice , Mice, Knockout , Organic Cation Transporter 2/genetics , Organic Cation Transporter 2/immunology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , Trans-Activators/genetics , Trans-Activators/immunology
20.
CA Cancer J Clin ; 70(1): 47-70, 2020 01.
Article in English | MEDLINE | ID: mdl-31815293

ABSTRACT

Historical advances in the care of patients with non-Hodgkin lymphoma (NHL) have been restricted largely to patients with B-cell lymphoma. The peripheral T-cell lymphomas (PTCLs), which are rare and heterogeneous in nature, have yet to experience the same degree of improvement in outcome over the past 20 to 30 years. It is estimated that there are approximately 80,000 and 14,000 cases, respectively, of NHL and Hodgkin lymphoma per year in the United States. As a subgroup of NHL, the PTCLs account for 6% to 10% of all cases of NHL, making them exceedingly rare. In addition, the World Health Organization 2017 classification describes 29 distinct subtypes of PTCL. This intrinsic diversity, coupled with its rarity, has stymied progress in the disease. In addition, most subtypes carry an inferior prognosis compared with their B-cell counterparts, an outcome largely attributed to the fact that most treatment paradigms for patients with PTCL have been derived from B-cell neoplasms, a radically different disease. In fact, the first drug ever approved for patients with PTCL was approved only a decade ago. The plethora of recent drug approvals in PTCL, coupled with a deeper understanding of the molecular pathogenesis of the disease, has stimulated the field to pursue new avenues of research that are now largely predicated on the development of novel, targeted small molecules, which include a host of epigenetic modifiers and biologics. There is an expectation these advances may begin to favorably challenge the chemotherapy paradigms that have been used in the T-cell malignancies.


Subject(s)
Antineoplastic Agents/therapeutic use , Lymphoma, T-Cell, Peripheral/drug therapy , Neoplasm Staging , Humans , Lymphoma, T-Cell, Peripheral/pathology , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL