Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 179
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(18): 3903-3920.e21, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37557169

ABSTRACT

Immune-checkpoint blockade has revolutionized cancer treatment, but some cancers, such as acute myeloid leukemia (AML), do not respond or develop resistance. A potential mode of resistance is immune evasion of T cell immunity involving aberrant major histocompatibility complex class I (MHC-I) antigen presentation (AP). To map such mechanisms of resistance, we identified key MHC-I regulators using specific peptide-MHC-I-guided CRISPR-Cas9 screens in AML. The top-ranked negative regulators were surface protein sushi domain containing 6 (SUSD6), transmembrane protein 127 (TMEM127), and the E3 ubiquitin ligase WWP2. SUSD6 is abundantly expressed in AML and multiple solid cancers, and its ablation enhanced MHC-I AP and reduced tumor growth in a CD8+ T cell-dependent manner. Mechanistically, SUSD6 forms a trimolecular complex with TMEM127 and MHC-I, which recruits WWP2 for MHC-I ubiquitination and lysosomal degradation. Together with the SUSD6/TMEM127/WWP2 gene signature, which negatively correlates with cancer survival, our findings define a membrane-associated MHC-I inhibitory axis as a potential therapeutic target for both leukemia and solid cancers.


Subject(s)
Histocompatibility Antigens Class I , Neoplasms , Tumor Escape , Humans , Antigen Presentation , CD8-Positive T-Lymphocytes , Histocompatibility Antigens Class I/metabolism , HLA Antigens , Neoplasms/immunology , Ubiquitin-Protein Ligases/genetics
2.
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
3.
Cell ; 170(6): 1079-1095.e20, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28823558

ABSTRACT

Loss-of-function mutations in TET2 occur frequently in patients with clonal hematopoiesis, myelodysplastic syndrome (MDS), and acute myeloid leukemia (AML) and are associated with a DNA hypermethylation phenotype. To determine the role of TET2 deficiency in leukemia stem cell maintenance, we generated a reversible transgenic RNAi mouse to model restoration of endogenous Tet2 expression. Tet2 restoration reverses aberrant hematopoietic stem and progenitor cell (HSPC) self-renewal in vitro and in vivo. Treatment with vitamin C, a co-factor of Fe2+ and α-KG-dependent dioxygenases, mimics TET2 restoration by enhancing 5-hydroxymethylcytosine formation in Tet2-deficient mouse HSPCs and suppresses human leukemic colony formation and leukemia progression of primary human leukemia PDXs. Vitamin C also drives DNA hypomethylation and expression of a TET2-dependent gene signature in human leukemia cell lines. Furthermore, TET-mediated DNA oxidation induced by vitamin C treatment in leukemia cells enhances their sensitivity to PARP inhibition and could provide a safe and effective combination strategy to selectively target TET deficiency in cancer. PAPERCLIP.


Subject(s)
Ascorbic Acid/pharmacology , DNA-Binding Proteins/metabolism , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Proto-Oncogene Proteins/metabolism , Vitamins/pharmacology , Animals , Ascorbic Acid/administration & dosage , Cell Death , Cell Line, Tumor , DNA Methylation , DNA-Binding Proteins/genetics , Dioxygenases , Gene Knockdown Techniques , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Myelodysplastic Syndromes/genetics , Neoplasm Transplantation , Poly (ADP-Ribose) Polymerase-1/genetics , Proto-Oncogene Proteins/genetics , Transcription, Genetic , Transplantation, Heterologous , Vitamins/administration & dosage
4.
Nat Immunol ; 20(10): 1393-1403, 2019 10.
Article in English | MEDLINE | ID: mdl-31477919

ABSTRACT

In B lymphopoiesis, activation of the pre-B cell antigen receptor (pre-BCR) is associated with both cell cycle exit and Igk recombination. Yet how the pre-BCR mediates these functions remains unclear. Here, we demonstrate that the pre-BCR initiates a feed-forward amplification loop mediated by the transcription factor interferon regulatory factor 4 and the chemokine receptor C-X-C motif chemokine receptor 4 (CXCR4). CXCR4 ligation by C-X-C motif chemokine ligand 12 activates the mitogen-activated protein kinase extracellular-signal-regulated kinase, which then directs the development of small pre- and immature B cells, including orchestrating cell cycle exit, pre-BCR repression, Igk recombination and BCR expression. In contrast, pre-BCR expression and escape from interleukin-7 have only modest effects on B cell developmental transcriptional and epigenetic programs. These data show a direct and central role for CXCR4 in orchestrating late B cell lymphopoiesis. Furthermore, in the context of previous findings, our data provide a three-receptor system sufficient to recapitulate the essential features of B lymphopoiesis in vitro.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin kappa-Chains/genetics , Precursor Cells, B-Lymphoid/physiology , Receptors, Antigen, B-Cell/metabolism , Receptors, CXCR4/metabolism , Animals , Cell Cycle Checkpoints , Cells, Cultured , Chemokine CXCL12/metabolism , Female , Interferon Regulatory Factors/genetics , Lymphopoiesis , Male , Mice , Receptors, Antigen, B-Cell/genetics , Receptors, CXCR4/genetics , Recombination, Genetic
5.
Nat Immunol ; 20(9): 1196-1207, 2019 09.
Article in English | MEDLINE | ID: mdl-31406379

ABSTRACT

The response to systemic infection and injury requires the rapid adaptation of hematopoietic stem cells (HSCs), which proliferate and divert their differentiation toward the myeloid lineage. Significant interest has emerged in understanding the signals that trigger the emergency hematopoietic program. However, the mechanisms that halt this response of HSCs, which is critical to restore homeostasis, remain unknown. Here we reveal that the E3 ubiquitin ligase Speckle-type BTB-POZ protein (SPOP) restrains the inflammatory activation of HSCs. In the absence of Spop, systemic inflammation proceeded in an unresolved manner, and the sustained response in the HSCs resulted in a lethal phenotype reminiscent of hyper-inflammatory syndrome or sepsis. Our proteomic studies decipher that SPOP restricted inflammation by ubiquitinating the innate signal transducer myeloid differentiation primary response protein 88 (MYD88). These findings unearth an HSC-intrinsic post-translational mechanism that is essential for reestablishing homeostasis after emergency hematopoiesis.


Subject(s)
Inflammation/immunology , Leukocytosis/immunology , Myeloid Differentiation Factor 88/metabolism , Neutrophils/immunology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Animals , Cell Line , Female , HEK293 Cells , Hematopoiesis/immunology , Humans , Male , Mice , Neutrophils/cytology , Ubiquitin-Protein Ligase Complexes , Ubiquitin-Protein Ligases/metabolism
6.
Mol Cell ; 83(8): 1216-1236.e12, 2023 04 20.
Article in English | MEDLINE | ID: mdl-36944333

ABSTRACT

Highly coordinated changes in gene expression underlie T cell activation and exhaustion. However, the mechanisms by which such programs are regulated and how these may be targeted for therapeutic benefit remain poorly understood. Here, we comprehensively profile the genomic occupancy of mSWI/SNF chromatin remodeling complexes throughout acute and chronic T cell stimulation, finding that stepwise changes in localization over transcription factor binding sites direct site-specific chromatin accessibility and gene activation leading to distinct phenotypes. Notably, perturbation of mSWI/SNF complexes using genetic and clinically relevant chemical strategies enhances the persistence of T cells with attenuated exhaustion hallmarks and increased memory features in vitro and in vivo. Finally, pharmacologic mSWI/SNF inhibition improves CAR-T expansion and results in improved anti-tumor control in vivo. These findings reveal the central role of mSWI/SNF complexes in the coordination of T cell activation and exhaustion and nominate small-molecule-based strategies for the improvement of current immunotherapy protocols.


Subject(s)
Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Transcription Factors/metabolism , Chromatin/genetics , Transcriptional Activation
7.
Immunity ; 54(6): 1304-1319.e9, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34048708

ABSTRACT

Despite mounting evidence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) engagement with immune cells, most express little, if any, of the canonical receptor of SARS-CoV-2, angiotensin-converting enzyme 2 (ACE2). Here, using a myeloid cell receptor-focused ectopic expression screen, we identified several C-type lectins (DC-SIGN, L-SIGN, LSECtin, ASGR1, and CLEC10A) and Tweety family member 2 (TTYH2) as glycan-dependent binding partners of the SARS-CoV-2 spike. Except for TTYH2, these molecules primarily interacted with spike via regions outside of the receptor-binding domain. Single-cell RNA sequencing analysis of pulmonary cells from individuals with coronavirus disease 2019 (COVID-19) indicated predominant expression of these molecules on myeloid cells. Although these receptors do not support active replication of SARS-CoV-2, their engagement with the virus induced robust proinflammatory responses in myeloid cells that correlated with COVID-19 severity. We also generated a bispecific anti-spike nanobody that not only blocked ACE2-mediated infection but also the myeloid receptor-mediated proinflammatory responses. Our findings suggest that SARS-CoV-2-myeloid receptor interactions promote immune hyperactivation, which represents potential targets for COVID-19 therapy.


Subject(s)
COVID-19/metabolism , COVID-19/virology , Host-Pathogen Interactions , Lectins, C-Type/metabolism , Membrane Proteins/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasm Proteins/metabolism , SARS-CoV-2/physiology , Angiotensin-Converting Enzyme 2/metabolism , Binding Sites , COVID-19/genetics , Cell Line , Cytokines , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Inflammation Mediators/metabolism , Lectins, C-Type/chemistry , Membrane Proteins/chemistry , Models, Molecular , Neoplasm Proteins/chemistry , Protein Binding , Protein Conformation , Single-Domain Antibodies/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , Structure-Activity Relationship
8.
Cell ; 158(3): 593-606, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25083870

ABSTRACT

Notch signaling is a key developmental pathway that is subject to frequent genetic and epigenetic perturbations in many different human tumors. Here we investigate whether long noncoding RNA (lncRNA) genes, in addition to mRNAs, are key downstream targets of oncogenic Notch1 in human T cell acute lymphoblastic leukemia (T-ALL). By integrating transcriptome profiles with chromatin state maps, we have uncovered many previously unreported T-ALL-specific lncRNA genes, a fraction of which are directly controlled by the Notch1/Rpbjκ activator complex. Finally we have shown that one specific Notch-regulated lncRNA, LUNAR1, is required for efficient T-ALL growth in vitro and in vivo due to its ability to enhance IGF1R mRNA expression and sustain IGF1 signaling. These results confirm that lncRNAs are important downstream targets of the Notch signaling pathway, and additionally they are key regulators of the oncogenic state in T-ALL.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA, Long Noncoding/analysis , Receptor, Notch1/metabolism , Genome-Wide Association Study , Humans , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Long Noncoding/genetics , Signal Transduction , Thymus Gland/pathology
9.
Nat Immunol ; 17(9): 1016-24, 2016 09.
Article in English | MEDLINE | ID: mdl-27478938

ABSTRACT

The past decade brought a revolution in understanding of the structure, topology and disease-inducing lesions of RNA and DNA, fueled by unprecedented progress in next-generation sequencing. This technological revolution has also affected understanding of the epigenome and has provided unique opportunities for the analysis of DNA and histone modifications, as well as the first map of the non-protein-coding genome and three-dimensional (3D) chromosomal interactions. Overall, these advances have facilitated studies that combine genetic, transcriptomics and epigenomics data to address a wide range of issues ranging from understanding the role of the epigenome in development to targeting the transcription of noncoding genes in human cancer. Here we describe recent insights into epigenetic dysregulation characteristic of the malignant differentiation of blood stem cells based on studies of alterations that affect epigenetic complexes, enhancers, chromatin, long noncoding RNAs (lncRNAs), RNA splicing, nuclear topology and the 3D conformation of chromatin.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic , Hematologic Neoplasms/genetics , Hematopoiesis/genetics , RNA, Long Noncoding/genetics , Animals , Chromatin/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Molecular Conformation , Transcriptome
10.
Nat Immunol ; 17(11): 1312-1321, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27668798

ABSTRACT

Hematopoietic stem cells (HSCs) are dormant in the bone marrow and can be activated in response to diverse stresses to replenish all blood cell types. We identified the ubiquitin ligase Huwe1 as a crucial regulator of HSC function via its post-translational control of the oncoprotein N-myc (encoded by Mycn). We found Huwe1 to be essential for HSC self-renewal, quiescence and lymphoid-fate specification in mice. Through the use of a fluorescent fusion allele (MycnM), we observed that N-myc expression was restricted to the most immature, multipotent stem and progenitor populations. N-myc expression was upregulated in response to stress or following loss of Huwe1, which led to increased proliferation and stem-cell exhaustion. Mycn depletion reversed most of these phenotypes in vivo, which suggested that the attenuation of N-myc by Huwe1 is essential for reestablishing homeostasis following stress.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Lymphocytes/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Cell Cycle/genetics , Cell Line , Cell Self Renewal/genetics , Cluster Analysis , Gene Expression Profiling , Genes, myc , Lymphocytes/cytology , Mice , Mice, Knockout , Mice, Transgenic , Protein Stability , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Stress, Physiological , Transcription, Genetic , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism
11.
Cell ; 153(7): 1552-66, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23791182

ABSTRACT

Sequencing efforts led to the identification of somatic mutations that could affect the self-renewal and differentiation of cancer-initiating cells. One such recurrent mutation targets the binding pocket of the ubiquitin ligase Fbxw7. Missense FBXW7 mutations are prevalent in various tumors, including T cell acute lymphoblastic leukemia (T-ALL). To study the effects of such lesions, we generated animals carrying regulatable Fbxw7 mutant alleles. Here, we show that these mutations specifically bolster cancer-initiating cell activity in collaboration with Notch1 oncogenes but spare normal hematopoietic stem cell function. We were also able to show that FBXW7 mutations specifically affect the ubiquitylation and half-life of c-Myc protein, a key T-ALL oncogene. Using animals carrying c-Myc fusion alleles, we connected Fbxw7 function to c-Myc abundance and correlated c-Myc expression to leukemia-initiating activity. Finally, we demonstrated that small-molecule-mediated suppression of MYC activity leads to T-ALL remission, suggesting an effective therapeutic strategy.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Cycle Proteins/genetics , Disease Models, Animal , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Knockout , Mutation, Missense , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Receptor, Notch1/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
12.
Nature ; 601(7893): 428-433, 2022 01.
Article in English | MEDLINE | ID: mdl-34937946

ABSTRACT

Although deregulation of transfer RNA (tRNA) biogenesis promotes the translation of pro-tumorigenic mRNAs in cancers1,2, the mechanisms and consequences of tRNA deregulation in tumorigenesis are poorly understood. Here we use a CRISPR-Cas9 screen to focus on genes that have been implicated in tRNA biogenesis, and identify a mechanism by which altered valine tRNA biogenesis enhances mitochondrial bioenergetics in T cell acute lymphoblastic leukaemia (T-ALL). Expression of valine aminoacyl tRNA synthetase is transcriptionally upregulated by NOTCH1, a key oncogene in T-ALL, underlining a role for oncogenic transcriptional programs in coordinating tRNA supply and demand. Limiting valine bioavailability through restriction of dietary valine intake disrupted this balance in mice, resulting in decreased leukaemic burden and increased survival in vivo. Mechanistically, valine restriction reduced translation rates of mRNAs that encode subunits of mitochondrial complex I, leading to defective assembly of complex I and impaired oxidative phosphorylation. Finally, a genome-wide CRISPR-Cas9 loss-of-function screen in differential valine conditions identified several genes, including SLC7A5 and BCL2, whose genetic ablation or pharmacological inhibition synergized with valine restriction to reduce T-ALL growth. Our findings identify tRNA deregulation as a critical adaptation in the pathogenesis of T-ALL and provide a molecular basis for the use of dietary approaches to target tRNA biogenesis in blood malignancies.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Valine-tRNA Ligase , Valine , Animals , Biological Availability , CRISPR-Cas Systems , Diet , Electron Transport Complex I/genetics , Large Neutral Amino Acid-Transporter 1 , Mice , Mitochondria/metabolism , Oxidative Phosphorylation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-bcl-2 , RNA, Transfer/genetics , Valine/metabolism , Valine-tRNA Ligase/metabolism
13.
Nature ; 610(7933): 737-743, 2022 10.
Article in English | MEDLINE | ID: mdl-36071167

ABSTRACT

The mutualistic relationship of gut-resident microbiota and the host immune system promotes homeostasis that ensures maintenance of the microbial community and of a largely non-aggressive immune cell compartment1,2. The consequences of disturbing this balance include proximal inflammatory conditions, such as Crohn's disease, and systemic illnesses. This equilibrium is achieved in part through the induction of both effector and suppressor arms of the adaptive immune system. Helicobacter species induce T regulatory (Treg) and T follicular helper (TFH) cells under homeostatic conditions, but induce inflammatory T helper 17 (TH17) cells when induced Treg (iTreg) cells are compromised3,4. How Helicobacter and other gut bacteria direct T cells to adopt distinct functions remains poorly understood. Here we investigated the cells and molecular components required for iTreg cell differentiation. We found that antigen presentation by cells expressing RORγt, rather than by classical dendritic cells, was required and sufficient for induction of Treg cells. These RORγt+ cells-probably type 3 innate lymphoid cells and/or Janus cells5-require the antigen-presentation machinery, the chemokine receptor CCR7 and the TGFß activator αv integrin. In the absence of any of these factors, there was expansion of pathogenic TH17 cells instead of iTreg cells, induced by CCR7-independent antigen-presenting cells. Thus, intestinal commensal microbes and their products target multiple antigen-presenting cells with pre-determined features suited to directing appropriate T cell differentiation programmes, rather than a common antigen-presenting cell that they endow with appropriate functions.


Subject(s)
Cell Differentiation , Gastrointestinal Microbiome , Nuclear Receptor Subfamily 1, Group F, Member 3 , T-Lymphocytes, Regulatory , Dendritic Cells/immunology , Gastrointestinal Microbiome/immunology , Homeostasis , Immunity, Innate , Integrin alphaV/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, CCR7/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/metabolism , Antigen Presentation/immunology , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology
14.
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
15.
Trends Immunol ; 45(3): 177-187, 2024 03.
Article in English | MEDLINE | ID: mdl-38433029

ABSTRACT

The MHC-I antigen presentation (AP) pathway is key to shaping mammalian CD8+ T cell immunity, with its aberrant expression closely linked to low tumor immunogenicity and immunotherapy resistance. While significant attention has been given to genetic mutations and downregulation of positive regulators that are essential for MHC-I AP, there is a growing interest in understanding how tumors actively evade MHC-I expression and/or AP through the induction of MHC-I inhibitory pathways. This emerging field of study may offer more viable therapeutic targets for future cancer immunotherapy. Here, we explore potential mechanisms by which cancer cells evade MHC-I AP and function and propose therapeutic strategies that might target these MHC-I inhibitors to restore impaired T cell immunity within the tumor microenvironment (TME).


Subject(s)
Histocompatibility Antigens Class I , Neoplasms , Animals , Humans , Histocompatibility Antigens Class I/metabolism , CD8-Positive T-Lymphocytes , Immunotherapy , Antigens, Neoplasm , Mammals , Tumor Microenvironment
16.
N Engl J Med ; 386(8): 735-743, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35196427

ABSTRACT

BACKGROUND: Covalent (irreversible) Bruton's tyrosine kinase (BTK) inhibitors have transformed the treatment of multiple B-cell cancers, especially chronic lymphocytic leukemia (CLL). However, resistance can arise through multiple mechanisms, including acquired mutations in BTK at residue C481, the binding site of covalent BTK inhibitors. Noncovalent (reversible) BTK inhibitors overcome this mechanism and other sources of resistance, but the mechanisms of resistance to these therapies are currently not well understood. METHODS: We performed genomic analyses of pretreatment specimens as well as specimens obtained at the time of disease progression from patients with CLL who had been treated with the noncovalent BTK inhibitor pirtobrutinib. Structural modeling, BTK-binding assays, and cell-based assays were conducted to study mutations that confer resistance to noncovalent BTK inhibitors. RESULTS: Among 55 treated patients, we identified 9 patients with relapsed or refractory CLL and acquired mechanisms of genetic resistance to pirtobrutinib. We found mutations (V416L, A428D, M437R, T474I, and L528W) that were clustered in the kinase domain of BTK and that conferred resistance to both noncovalent BTK inhibitors and certain covalent BTK inhibitors. Mutations in BTK or phospholipase C gamma 2 (PLCγ2), a signaling molecule and downstream substrate of BTK, were found in all 9 patients. Transcriptional activation reflecting B-cell-receptor signaling persisted despite continued therapy with noncovalent BTK inhibitors. CONCLUSIONS: Resistance to noncovalent BTK inhibitors arose through on-target BTK mutations and downstream PLCγ2 mutations that allowed escape from BTK inhibition. A proportion of these mutations also conferred resistance across clinically approved covalent BTK inhibitors. These data suggested new mechanisms of genomic escape from established covalent and novel noncovalent BTK inhibitors. (Funded by the American Society of Hematology and others.).


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Drug Resistance, Neoplasm , Leukemia, Lymphocytic, Chronic, B-Cell , Mutation , Phospholipase C gamma , Protein Kinase Inhibitors , Humans , Middle Aged , Adenine/analogs & derivatives , Adenine/pharmacology , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/genetics , Agammaglobulinaemia Tyrosine Kinase/ultrastructure , Drug Resistance, Neoplasm/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Phospholipase C gamma/genetics , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptors, Antigen, B-Cell/metabolism , Sequence Analysis, RNA , Signal Transduction/drug effects
18.
Nat Immunol ; 14(3): 262-70, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23377202

ABSTRACT

The physiological basis and mechanistic requirements for a large number of functional immunoreceptor tyrosine-based activation motifs (ITAMs; high ITAM multiplicity) in the complex of the T cell antigen receptor (TCR) and the invariant signaling protein CD3 remain obscure. Here we found that whereas a low multiplicity of TCR-CD3 ITAMs was sufficient to engage canonical TCR-induced signaling events that led to cytokine secretion, a high multiplicity of TCR-CD3 ITAMs was required for TCR-driven proliferation. This was dependent on the formation of compact immunological synapses, interaction of the adaptor Vav1 with phosphorylated CD3 ITAMs to mediate the recruitment and activation of the oncogenic transcription factor Notch1 and, ultimately, proliferation induced by the cell-cycle regulator c-Myc. Analogous mechanistic events were also needed to drive proliferation in response to weak peptide agonists. Thus, the TCR-driven pathways that initiate cytokine secretion and proliferation are separable and are coordinated by the multiplicity of phosphorylated ITAMs in TCR-CD3.


Subject(s)
CD3 Complex/immunology , Cytokines/biosynthesis , Immunoreceptor Tyrosine-Based Activation Motif/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , CD3 Complex/metabolism , Cell Line , Cell Proliferation , HEK293 Cells , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-vav/metabolism , Receptor, Notch1/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/metabolism
19.
Trends Immunol ; 43(6): 466-477, 2022 06.
Article in English | MEDLINE | ID: mdl-35490133

ABSTRACT

tRNAs are central players in decoding the genetic code linking codons in mRNAs with cognate amino acids during protein synthesis. Recent discoveries have placed tRNAs as key regulators of gene expression during hematopoiesis, especially in hematopoietic stem cell (HSC) maintenance and immune development. These functions have been shown to be influenced by dynamic changes in tRNA expression, post-transcriptional base modifications, tRNA-interacting proteins, and tRNA fragmentation; these events underlie the complexity of tRNA-mediated regulatory events in hematopoiesis. In this review, we discuss these recent findings and highlight how deregulation of tRNA biogenesis can contribute to hematological malignancies.


Subject(s)
Hematologic Neoplasms , RNA, Transfer , Codon , Genetic Code , Hematologic Neoplasms/genetics , Hematopoiesis/genetics , Humans , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Transfer/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL