Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 17.383
Filter
Add more filters

Uruguay Oncology Collection
Publication year range
1.
Nat Immunol ; 24(6): 991-1006, 2023 06.
Article in English | MEDLINE | ID: mdl-37095377

ABSTRACT

Germinal center (GC) B cells undergo proliferation at very high rates in a hypoxic microenvironment but the cellular processes driving this are incompletely understood. Here we show that the mitochondria of GC B cells are highly dynamic, with significantly upregulated transcription and translation rates associated with the activity of transcription factor A, mitochondrial (TFAM). TFAM, while also necessary for normal B cell development, is required for entry of activated GC precursor B cells into the germinal center reaction; deletion of Tfam significantly impairs GC formation, function and output. Loss of TFAM in B cells compromises the actin cytoskeleton and impairs cellular motility of GC B cells in response to chemokine signaling, leading to their spatial disorganization. We show that B cell lymphoma substantially increases mitochondrial translation and that deletion of Tfam in B cells is protective against the development of lymphoma in a c-Myc transgenic mouse model. Finally, we show that pharmacological inhibition of mitochondrial transcription and translation inhibits growth of GC-derived human lymphoma cells and induces similar defects in the actin cytoskeleton.


Subject(s)
Lymphoma, B-Cell , Lymphoma , Mice , Humans , Animals , B-Lymphocytes/pathology , Germinal Center/pathology , Transcription, Genetic , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Mice, Transgenic , Tumor Microenvironment
2.
Nat Immunol ; 24(2): 337-348, 2023 02.
Article in English | MEDLINE | ID: mdl-36577930

ABSTRACT

Our previous study using systems vaccinology identified an association between the sterol regulatory binding protein (SREBP) pathway and humoral immune response to vaccination in humans. To investigate the role of SREBP signaling in modulating immune responses, we generated mice with B cell- or CD11c+ antigen-presenting cell (APC)-specific deletion of SCAP, an essential regulator of SREBP signaling. Ablation of SCAP in CD11c+ APCs had no effect on immune responses. In contrast, SREBP signaling in B cells was critical for antibody responses, as well as the generation of germinal centers,memory B cells and bone marrow plasma cells. SREBP signaling was required for metabolic reprogramming in activated B cells. Upon mitogen stimulation, SCAP-deficient B cells could not proliferate and had decreased lipid rafts. Deletion of SCAP in germinal center B cells using AID-Cre decreased lipid raft content and cell cycle progression. These studies provide mechanistic insights coupling sterol metabolism with the quality and longevity of humoral immunity.


Subject(s)
Carrier Proteins , Lymphoma, B-Cell , Sterols , Animals , Humans , Mice , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sterols/metabolism , Lymphoma, B-Cell/metabolism
3.
Nat Immunol ; 23(10): 1424-1432, 2022 10.
Article in English | MEDLINE | ID: mdl-36138187

ABSTRACT

B cell progenitor acute lymphoblastic leukemia (B-ALL) treatment has been revolutionized by T cell-based immunotherapies-including chimeric antigen receptor T cell therapy (CAR-T) and the bispecific T cell engager therapeutic, blinatumomab-targeting surface glycoprotein CD19. Unfortunately, many patients with B-ALL will fail immunotherapy due to 'antigen escape'-the loss or absence of leukemic CD19 targeted by anti-leukemic T cells. In the present study, we utilized a genome-wide CRISPR-Cas9 screening approach to identify modulators of CD19 abundance on human B-ALL blasts. These studies identified a critical role for the transcriptional activator ZNF143 in CD19 promoter activation. Conversely, the RNA-binding protein, NUDT21, limited expression of CD19 by regulating CD19 messenger RNA polyadenylation and stability. NUDT21 deletion in B-ALL cells increased the expression of CD19 and the sensitivity to CD19-specific CAR-T and blinatumomab. In human B-ALL patients treated with CAR-T and blinatumomab, upregulation of NUDT21 mRNA coincided with CD19 loss at disease relapse. Together, these studies identify new CD19 modulators in human B-ALL.


Subject(s)
Burkitt Lymphoma , Lymphoma, B-Cell , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Receptors, Chimeric Antigen , Antigens, CD19/genetics , Antigens, CD19/metabolism , Cleavage And Polyadenylation Specificity Factor/metabolism , Humans , Immunotherapy, Adoptive/adverse effects , Membrane Glycoproteins/metabolism , Polyadenylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Chimeric Antigen/metabolism , Trans-Activators/metabolism
4.
Annu Rev Immunol ; 30: 565-610, 2012.
Article in English | MEDLINE | ID: mdl-22224767

ABSTRACT

The mechanisms that drive normal B cell differentiation and activation are frequently subverted by B cell lymphomas for their unlimited growth and survival. B cells are particularly prone to malignant transformation because the machinery used for antibody diversification can cause chromosomal translocations and oncogenic mutations. The advent of functional and structural genomics has greatly accelerated our understanding of oncogenic mechanisms in lymphomagenesis. The signaling pathways that normal B cells utilize to sense antigens are frequently derailed in B cell malignancies, leading to constitutive activation of prosurvival pathways. These malignancies co-opt transcriptional regulatory systems that characterize their normal B cell counterparts and frequently alter epigenetic regulators of chromatin structure and gene expression. These mechanistic insights are ushering in an era of targeted therapies for these cancers based on the principles of pathogenesis.


Subject(s)
Lymphoma, B-Cell/etiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epigenesis, Genetic , Humans , Immune Evasion , Lymphoma, B-Cell/drug therapy , MicroRNAs/genetics , MicroRNAs/metabolism , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Nat Immunol ; 22(2): 240-253, 2021 02.
Article in English | MEDLINE | ID: mdl-33432228

ABSTRACT

During the germinal center (GC) reaction, B cells undergo extensive redistribution of cohesin complex and three-dimensional reorganization of their genomes. Yet, the significance of cohesin and architectural programming in the humoral immune response is unknown. Herein we report that homozygous deletion of Smc3, encoding the cohesin ATPase subunit, abrogated GC formation, while, in marked contrast, Smc3 haploinsufficiency resulted in GC hyperplasia, skewing of GC polarity and impaired plasma cell (PC) differentiation. Genome-wide chromosomal conformation and transcriptional profiling revealed defects in GC B cell terminal differentiation programs controlled by the lymphoma epigenetic tumor suppressors Tet2 and Kmt2d and failure of Smc3-haploinsufficient GC B cells to switch from B cell- to PC-defining transcription factors. Smc3 haploinsufficiency preferentially impaired the connectivity of enhancer elements controlling various lymphoma tumor suppressor genes, and, accordingly, Smc3 haploinsufficiency accelerated lymphomagenesis in mice with constitutive Bcl6 expression. Collectively, our data indicate a dose-dependent function for cohesin in humoral immunity to facilitate the B cell to PC phenotypic switch while restricting malignant transformation.


Subject(s)
B-Lymphocytes/metabolism , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic/genetics , Chondroitin Sulfate Proteoglycans/genetics , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , Gene Dosage , Germinal Center/metabolism , Immunity, Humoral , Lymphoma, B-Cell/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Chondroitin Sulfate Proteoglycans/deficiency , Chondroitin Sulfate Proteoglycans/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Gene Deletion , Gene Expression Regulation, Neoplastic , Germinal Center/immunology , Germinal Center/pathology , Haploinsufficiency , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction , Cohesins
6.
Nat Immunol ; 19(9): 1013-1024, 2018 09.
Article in English | MEDLINE | ID: mdl-30104629

ABSTRACT

Most adult B cell lymphomas originate from germinal center (GC) B cells, but it is unclear to what extent B cells in overt lymphoma retain the functional dynamics of GC B cells or are blocked at a particular stage of the GC reaction. Here we used integrative single-cell analysis of phenotype, gene expression and variable-region sequence of the immunoglobulin heavy-chain locus to track the characteristic human GC B cell program in follicular lymphoma B cells. By modeling the cyclic continuum of GC B cell transitional states, we identified characteristic patterns of synchronously expressed gene clusters. GC-specific gene-expression synchrony was lost in single lymphoma B cells. However, distinct follicular lymphoma-specific cell states co-existed within single patient biopsies. Our data show that lymphoma B cells are not blocked in a GC B cell state but might adopt new dynamic modes of functional diversity, which opens the possibility of novel definitions of lymphoma identity.


Subject(s)
B-Lymphocyte Subsets/physiology , B-Lymphocytes/physiology , Germinal Center/physiology , Immunoglobulin Variable Region/genetics , Lymphoma, B-Cell/genetics , Adult , Cell Differentiation , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic , Germinal Center/pathology , Humans , Lymphoma, B-Cell/pathology , Male , Middle Aged , Single-Cell Analysis , Transcriptome/genetics
7.
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
8.
Cell ; 162(4): 697-8, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26276622

ABSTRACT

Nussenzweig and colleagues evaluate genomic instability and germinal center derived lymphomagenesis in mice infected with Plasmodium to recreate some of the hallmark characteristics of Burkitt lymphoma, a form of cancer more common in parts of Africa where malaria is endemic.


Subject(s)
Cytidine Deaminase/metabolism , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/genetics , Translocation, Genetic , Animals , Humans
9.
Cell ; 162(4): 727-37, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26276629

ABSTRACT

Chronic infection with Plasmodium falciparum was epidemiologically associated with endemic Burkitt's lymphoma, a mature B cell cancer characterized by chromosome translocation between the c-myc oncogene and Igh, over 50 years ago. Whether infection promotes B cell lymphoma, and if so by which mechanism, remains unknown. To investigate the relationship between parasitic disease and lymphomagenesis, we used Plasmodium chabaudi (Pc) to produce chronic malaria infection in mice. Pc induces prolonged expansion of germinal centers (GCs), unique compartments in which B cells undergo rapid clonal expansion and express activation-induced cytidine deaminase (AID), a DNA mutator. GC B cells elicited during Pc infection suffer widespread DNA damage, leading to chromosome translocations. Although infection does not change the overall rate, it modifies lymphomagenesis to favor mature B cell lymphomas that are AID dependent and show chromosome translocations. Thus, malaria infection favors mature B cell cancers by eliciting protracted AID expression in GC B cells. PAPERCLIP.


Subject(s)
Genomic Instability , Lymphoma, B-Cell/genetics , Malaria/complications , Malaria/genetics , Plasmodium chabaudi/physiology , Animals , B-Lymphocytes/pathology , Chronic Disease , Cytidine Deaminase/metabolism , DNA Replication , Genes, p53 , Germinal Center/parasitology , Malaria/parasitology , Malaria/pathology , Mice , Translocation, Genetic
10.
Mol Cell ; 82(3): 696-708.e4, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35090599

ABSTRACT

We have used a combination of chemical genetics, chromatin proteomics, and imaging to map the earliest chromatin transactions during vertebrate cell entry into mitosis. Chicken DT40 CDK1as cells undergo synchronous mitotic entry within 15 min following release from a 1NM-PP1-induced arrest in late G2. In addition to changes in chromatin association with nuclear pores and the nuclear envelope, earliest prophase is dominated by changes in the association of ribonucleoproteins with chromatin, particularly in the nucleolus, where pre-rRNA processing factors leave chromatin significantly before RNA polymerase I. Nuclear envelope barrier function is lost early in prophase, and cytoplasmic proteins begin to accumulate on the chromatin. As a result, outer kinetochore assembly appears complete by nuclear envelope breakdown (NEBD). Most interphase chromatin proteins remain associated with chromatin until NEBD, after which their levels drop sharply. An interactive proteomic map of chromatin transactions during mitotic entry is available as a resource at https://mitoChEP.bio.ed.ac.uk.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Chromosomes , DNA/metabolism , Lymphoma, B-Cell/metabolism , Nuclear Proteins/metabolism , Prophase , Proteome , Proteomics , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Chickens , Chromatin/genetics , DNA/genetics , Lamin Type B/genetics , Lamin Type B/metabolism , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Nuclear Proteins/genetics , Protein Binding , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Time Factors
12.
Nat Immunol ; 18(8): 889-898, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28604720

ABSTRACT

Engineered crystallizable fragment (Fc) regions of antibody domains, which assume a unique and unprecedented asymmetric structure within the homodimeric Fc polypeptide, enable completely selective binding to the complement component C1q and activation of complement via the classical pathway without any concomitant engagement of the Fcγ receptor (FcγR). We used the engineered Fc domains to demonstrate in vitro and in mouse models that for therapeutic antibodies, complement-dependent cell-mediated cytotoxicity (CDCC) and complement-dependent cell-mediated phagocytosis (CDCP) by immunological effector molecules mediated the clearance of target cells with kinetics and efficacy comparable to those of the FcγR-dependent effector functions that are much better studied, while they circumvented certain adverse reactions associated with FcγR engagement. Collectively, our data highlight the importance of CDCC and CDCP in monoclonal-antibody function and provide an experimental approach for delineating the effect of complement-dependent effector-cell engagement in various therapeutic settings.


Subject(s)
Complement C1q/immunology , Cytotoxicity, Immunologic/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Immunotherapy , Neoplasms/drug therapy , Phagocytosis/immunology , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/immunology , Cell Line, Tumor , Chromatography, Gel , Chromatography, Liquid , Complement C1q/metabolism , Crystallization , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/metabolism , In Vitro Techniques , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/immunology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Mass Spectrometry , Mice , Neoplasms/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, IgG/metabolism , Surface Plasmon Resonance , Tandem Mass Spectrometry
13.
Cell ; 157(7): 1644-1656, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24949974

ABSTRACT

Because apoptosis of infected cells can limit virus production and spread, some viruses have co-opted prosurvival genes from the host. This includes the Epstein-Barr virus (EBV) gene BHRF1, a homolog of human Bcl-2 proteins that block apoptosis and are associated with cancer. Computational design and experimental optimization were used to generate a novel protein called BINDI that binds BHRF1 with picomolar affinity. BINDI recognizes the hydrophobic cleft of BHRF1 in a manner similar to other Bcl-2 protein interactions but makes many additional contacts to achieve exceptional affinity and specificity. BINDI induces apoptosis in EBV-infected cancer lines, and when delivered with an antibody-targeted intracellular delivery carrier, BINDI suppressed tumor growth and extended survival in a xenograft disease model of EBV-positive human lymphoma. High-specificity-designed proteins that selectively kill target cells may provide an advantage over the toxic compounds used in current generation antibody-drug conjugates.


Subject(s)
Herpesvirus 4, Human/chemistry , Protein Engineering , Proteins/pharmacology , Viral Proteins/antagonists & inhibitors , Amino Acid Sequence , Animals , Apoptosis/drug effects , Computational Biology , Crystallography, X-Ray , Epstein-Barr Virus Infections/drug therapy , Herpesvirus 4, Human/physiology , Heterografts , Humans , Lymphoma, B-Cell/drug therapy , Mice , Models, Molecular , Molecular Sequence Data , Neoplasm Transplantation , Proteins/chemistry , Proteins/metabolism , Sequence Alignment , Viral Proteins/chemistry
14.
Cell ; 156(1-2): 317-331, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24439385

ABSTRACT

Ferroptosis is a form of nonapoptotic cell death for which key regulators remain unknown. We sought a common mediator for the lethality of 12 ferroptosis-inducing small molecules. We used targeted metabolomic profiling to discover that depletion of glutathione causes inactivation of glutathione peroxidases (GPXs) in response to one class of compounds and a chemoproteomics strategy to discover that GPX4 is directly inhibited by a second class of compounds. GPX4 overexpression and knockdown modulated the lethality of 12 ferroptosis inducers, but not of 11 compounds with other lethal mechanisms. In addition, two representative ferroptosis inducers prevented tumor growth in xenograft mouse tumor models. Sensitivity profiling in 177 cancer cell lines revealed that diffuse large B cell lymphomas and renal cell carcinomas are particularly susceptible to GPX4-regulated ferroptosis. Thus, GPX4 is an essential regulator of ferroptotic cancer cell death.


Subject(s)
Carbolines/pharmacology , Cell Death/drug effects , Glutathione Peroxidase/antagonists & inhibitors , Piperazines/pharmacology , Animals , Carcinoma, Renal Cell/drug therapy , Cell Line, Tumor , Gene Knockdown Techniques , Glutathione/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Heterografts , Humans , Lymphoma, B-Cell/drug therapy , Mice , Neoplasm Transplantation , Neoplasms/drug therapy , Phospholipid Hydroperoxide Glutathione Peroxidase
15.
Nature ; 615(7952): 507-516, 2023 03.
Article in English | MEDLINE | ID: mdl-36890224

ABSTRACT

Although chimeric antigen receptor (CAR) T cells have altered the treatment landscape for B cell malignancies, the risk of on-target, off-tumour toxicity has hampered their development for solid tumours because most target antigens are shared with normal cells1,2. Researchers have attempted to apply Boolean-logic gating to CAR T cells to prevent toxicity3-5; however, a truly safe and effective logic-gated CAR has remained elusive6. Here we describe an approach to CAR engineering in which we replace traditional CD3ζ domains with intracellular proximal T cell signalling molecules. We show that certain proximal signalling CARs, such as a ZAP-70 CAR, can activate T cells and eradicate tumours in vivo while bypassing upstream signalling proteins, including CD3ζ. The primary role of ZAP-70 is to phosphorylate LAT and SLP-76, which form a scaffold for signal propagation. We exploited the cooperative role of LAT and SLP-76 to engineer logic-gated intracellular network (LINK) CAR, a rapid and reversible Boolean-logic AND-gated CAR T cell platform that outperforms other systems in both efficacy and prevention of on-target, off-tumour toxicity. LINK CAR will expand the range of molecules that can be targeted with CAR T cells, and will enable these powerful therapeutic agents to be used for solid tumours and diverse diseases such as autoimmunity7 and fibrosis8. In addition, this work shows that the internal signalling machinery of cells can be repurposed into surface receptors, which could open new avenues for cellular engineering.


Subject(s)
Cell Engineering , Immunotherapy, Adoptive , Logic , Neoplasms , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen , Signal Transduction , T-Lymphocytes , Humans , Cell Engineering/methods , Immunotherapy, Adoptive/adverse effects , Leukemia, B-Cell , Lymphoma, B-Cell , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
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.
Mol Cell ; 81(10): 2094-2111.e9, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33878293

ABSTRACT

Even though SYK and ZAP70 kinases share high sequence homology and serve analogous functions, their expression in B and T cells is strictly segregated throughout evolution. Here, we identified aberrant ZAP70 expression as a common feature in a broad range of B cell malignancies. We validated SYK as the kinase that sets the thresholds for negative selection of autoreactive and premalignant clones. When aberrantly expressed in B cells, ZAP70 competes with SYK at the BCR signalosome and redirects SYK from negative selection to tonic PI3K signaling, thereby promoting B cell survival. In genetic mouse models for B-ALL and B-CLL, conditional expression of Zap70 accelerated disease onset, while genetic deletion impaired malignant transformation. Inducible activation of Zap70 during B cell development compromised negative selection of autoreactive B cells, resulting in pervasive autoantibody production. Strict segregation of the two kinases is critical for normal B cell selection and represents a central safeguard against the development of autoimmune disease and B cell malignancies.


Subject(s)
Autoimmunity , Neoplasms/enzymology , Neoplasms/prevention & control , Syk Kinase/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism , Animals , Antigens, CD19/metabolism , B-Lymphocytes , Calcium/metabolism , Cell Differentiation , Cell Transformation, Neoplastic , Enzyme Activation , Humans , Immune Tolerance , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/pathology , Mice , Models, Genetic , NFATC Transcription Factors/metabolism , Neoplasm Proteins , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Receptors, Antigen, B-Cell/metabolism , Signal Transduction
18.
Immunity ; 51(2): 324-336.e5, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31350178

ABSTRACT

High-affinity B cell selection in the germinal center (GC) is governed by signals delivered by follicular helper T (Tfh) cells to B cells. Selected B cells undergo clonal expansion and affinity maturation in the GC dark zone in direct proportion to the amount of antigen they capture and present to Tfh cells in the light zone. Here, we examined the mechanisms whereby Tfh cells program the number of GC B cell divisions. Gene expression analysis revealed that Tfh cells induce Myc expression in light-zone B cells in direct proportion to antigen capture. Conditional Myc haplo-insufficiency or overexpression combined with cell division tracking showed that MYC expression produces a metabolic reservoir in selected light-zone B cells that is proportional to the number of cell divisions in the dark zone. Thus, MYC constitutes the GC B cell division timer that when deregulated leads to emergence of B cell lymphoma.


Subject(s)
B-Lymphocytes/immunology , Genes, myc/genetics , Germinal Center/immunology , Lymphoma, B-Cell/genetics , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibody Affinity , Cell Differentiation , Cell Division , Cell Proliferation , Clonal Selection, Antigen-Mediated , Gene Expression Regulation , Humans , Mice
19.
Nature ; 609(7926): 369-374, 2022 09.
Article in English | MEDLINE | ID: mdl-36045296

ABSTRACT

Recently, chimeric antigen receptor (CAR)-T cell therapy has shown great promise in treating haematological malignancies1-7. However, CAR-T cell therapy currently has several limitations8-12. Here we successfully developed a two-in-one approach to generate non-viral, gene-specific targeted CAR-T cells through CRISPR-Cas9. Using the optimized protocol, we demonstrated feasibility in a preclinical study by inserting an anti-CD19 CAR cassette into the AAVS1 safe-harbour locus. Furthermore, an innovative type of anti-CD19 CAR-T cell with PD1 integration was developed and showed superior ability to eradicate tumour cells in xenograft models. In adoptive therapy for relapsed/refractory aggressive B cell non-Hodgkin lymphoma (ClinicalTrials.gov, NCT04213469 ), we observed a high rate (87.5%) of complete remission and durable responses without serious adverse events in eight patients. Notably, these enhanced CAR-T cells were effective even at a low infusion dose and with a low percentage of CAR+ cells. Single-cell analysis showed that the electroporation method resulted in a high percentage of memory T cells in infusion products, and PD1 interference enhanced anti-tumour immune functions, further validating the advantages of non-viral, PD1-integrated CAR-T cells. Collectively, our results demonstrate the high safety and efficacy of non-viral, gene-specific integrated CAR-T cells, thus providing an innovative technology for CAR-T cell therapy.


Subject(s)
Immunotherapy, Adoptive , Lymphoma, B-Cell , Receptors, Chimeric Antigen , Animals , Antigens, CD19/immunology , Electroporation , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/therapy , Memory T Cells/immunology , Programmed Cell Death 1 Receptor/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use , Recurrence , Single-Cell Analysis , Xenograft Model Antitumor Assays
20.
Nat Immunol ; 16(6): 653-62, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25867473

ABSTRACT

The methylcytosine dioxygenase TET1 ('ten-eleven translocation 1') is an important regulator of 5-hydroxymethylcytosine (5hmC) in embryonic stem cells. The diminished expression of TET proteins and loss of 5hmC in many tumors suggests a critical role for the maintenance of this epigenetic modification. Here we found that deletion of Tet1 promoted the development of B cell lymphoma in mice. TET1 was required for maintenance of the normal abundance and distribution of 5hmC, which prevented hypermethylation of DNA, and for regulation of the B cell lineage and of genes encoding molecules involved in chromosome maintenance and DNA repair. Whole-exome sequencing of TET1-deficient tumors revealed mutations frequently found in non-Hodgkin B cell lymphoma (B-NHL), in which TET1 was hypermethylated and transcriptionally silenced. Our findings provide in vivo evidence of a function for TET1 as a tumor suppressor of hematopoietic malignancy.


Subject(s)
B-Lymphocytes/physiology , Cytosine/analogs & derivatives , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/physiology , Lymphoma, B-Cell/genetics , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromosomal Instability , Cytosine/metabolism , DNA Methylation , DNA Repair , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Exome/genetics , Gene Expression Profiling , Humans , Mice , Mutation/genetics , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL