Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Biol Chem ; 289(36): 24936-42, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25074936

ABSTRACT

Advances in proteomics and large scale studies of potential mitochondrial proteins have led to the identification of many novel mitochondrial proteins in need of further characterization. Among these novel proteins are three mammalian rRNA methyltransferase family members RNMTL1, MRM1, and MRM2. MRM1 and MRM2 have bacterial and yeast homologs, whereas RNMTL1 appears to have evolved later in higher eukaryotes. We recently confirmed the localization of the three proteins to mitochondria, specifically in the vicinity of mtDNA nucleoids. In this study, we took advantage of the ability of 2'-O-ribose modification to block site-specific cleavage of RNA by DNAzymes to show that MRM1, MRM2, and RNMTL1 are responsible for modification of human large subunit rRNA at residues G(1145), U(1369), and G(1370), respectively.


Subject(s)
Methyltransferases/metabolism , Mitochondrial Proteins/metabolism , RNA, Ribosomal, 16S/metabolism , Ribosome Subunits, Large/metabolism , Base Sequence , Binding Sites/genetics , Blotting, Northern , HEK293 Cells , HeLa Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Methylation , Methyltransferases/genetics , Mitochondrial Proteins/genetics , Nuclear Proteins , RNA Interference , RNA, Ribosomal, 16S/genetics , Ribosome Subunits, Large/genetics
2.
J Biol Chem ; 288(43): 31386-99, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24036117

ABSTRACT

We have identified RNMTL1, MRM1, and MRM2 (FtsJ2) as members of the RNA methyltransferase family that may be responsible for the three known 2'-O-ribose modifications of the 16 S rRNA core of the large mitochondrial ribosome subunit. These proteins are confined to foci located in the vicinity of mtDNA nucleoids. They show distinct patterns of association with mtDNA nucleoids and/or mitochondrial ribosomes in cell fractionation studies. We focused on the role of the least studied protein in this set, RNMTL1, to show that this protein interacts with the large ribosomal subunit as well as with a series of non-ribosomal proteins that may be involved in coupling of the rate of rRNA transcription and ribosome assembly in mitochondria. siRNA-directed silencing of RNMTL1 resulted in a significant inhibition of translation on mitochondrial ribosomes. Our results are consistent with a role for RNMTL1 in methylation of G(1370) of human 16 S rRNA.


Subject(s)
DNA, Mitochondrial/metabolism , Methyltransferases/metabolism , RNA, Ribosomal, 16S/metabolism , RNA/metabolism , Ribosomes/metabolism , 3T3 Cells , Animals , DNA, Mitochondrial/genetics , Humans , Methyltransferases/genetics , Mice , Mitochondrial Proteins/biosynthesis , Protein Biosynthesis/physiology , RNA/genetics , RNA, Mitochondrial , RNA, Ribosomal, 16S/genetics , Ribosomes/genetics
3.
Nat Cancer ; 1(12): 1188-1203, 2021 12.
Article in English | MEDLINE | ID: mdl-33834176

ABSTRACT

Immune checkpoint blockade (ICB) has improved outcomes for patients with advanced cancer, but the determinants of response remain poorly understood. Here we report differential effects of mutations in the homologous recombination genes BRCA1 and BRCA2 on response to ICB in mouse and human tumors, and further show that truncating mutations in BRCA2 are associated with superior response compared to those in BRCA1. Mutations in BRCA1 and BRCA2 result in distinct mutational landscapes and differentially modulate the tumor-immune microenvironment, with gene expression programs related to both adaptive and innate immunity enriched in BRCA2-deficient tumors. Single-cell RNA sequencing further revealed distinct T cell, natural killer, macrophage, and dendritic cell populations enriched in BRCA2-deficient tumors. Taken together, our findings reveal the divergent effects of BRCA1 and BRCA2-deficiency on ICB outcome, and have significant implications for elucidating the genetic and microenvironmental determinants of response to immunotherapy.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Tumor Microenvironment , Animals , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Genes, BRCA2 , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Mice , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Tumor Microenvironment/genetics
4.
Science ; 364(6439): 485-491, 2019 05 03.
Article in English | MEDLINE | ID: mdl-31048490

ABSTRACT

Tumors with mismatch repair deficiency (MMR-d) are characterized by sequence alterations in microsatellites and can accumulate thousands of mutations. This high mutational burden renders tumors immunogenic and sensitive to programmed cell death-1 (PD-1) immune checkpoint inhibitors. Yet, despite their tumor immunogenicity, patients with MMR-deficient tumors experience highly variable responses, and roughly half are refractory to treatment. We present experimental and clinical evidence showing that the degree of microsatellite instability (MSI) and resultant mutational load, in part, underlies the variable response to PD-1 blockade immunotherapy in MMR-d human and mouse tumors. The extent of response is particularly associated with the accumulation of insertion-deletion (indel) mutational load. This study provides a rationale for the genome-wide characterization of MSI intensity and mutational load to better profile responses to anti-PD-1 immunotherapy across MMR-deficient human cancers.


Subject(s)
DNA Mismatch Repair/genetics , Immunotherapy/methods , Microsatellite Instability , Neoplasms/genetics , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antibodies/therapeutic use , Genetic Variation , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Mice , MutS Homolog 2 Protein/genetics , Mutation , Treatment Outcome
5.
Nat Med ; 25(5): 767-775, 2019 05.
Article in English | MEDLINE | ID: mdl-31011208

ABSTRACT

Anti-tumor immunity is driven by self versus non-self discrimination. Many immunotherapeutic approaches to cancer have taken advantage of tumor neoantigens derived from somatic mutations. Here, we demonstrate that gene fusions are a source of immunogenic neoantigens that can mediate responses to immunotherapy. We identified an exceptional responder with metastatic head and neck cancer who experienced a complete response to immune checkpoint inhibitor therapy, despite a low mutational load and minimal pre-treatment immune infiltration in the tumor. Using whole-genome sequencing and RNA sequencing, we identified a novel gene fusion and demonstrated that it produces a neoantigen that can specifically elicit a host cytotoxic T cell response. In a cohort of head and neck tumors with low mutation burden, minimal immune infiltration and prevalent gene fusions, we also identified gene fusion-derived neoantigens that generate cytotoxic T cell responses. Finally, analyzing additional datasets of fusion-positive cancers, including checkpoint-inhibitor-treated tumors, we found evidence of immune surveillance resulting in negative selective pressure against gene fusion-derived neoantigens. These findings highlight an important class of tumor-specific antigens and have implications for targeting gene fusion events in cancers that would otherwise be less poised for response to immunotherapy, including cancers with low mutational load and minimal immune infiltration.


Subject(s)
Antigens, Neoplasm/genetics , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/immunology , Gene Fusion , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/therapy , Humans , NFI Transcription Factors/genetics , NFI Transcription Factors/immunology , Neoplasms/genetics , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Oncogene Proteins/genetics , Oncogene Proteins/immunology , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/therapy , Whole Genome Sequencing
6.
J Natl Cancer Inst ; 110(12): 1386-1392, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29659925

ABSTRACT

Background: Tobacco smoking creates DNA damage, inducing mutations and potentially altering the tumor immune microenvironment. These types of genetic and immune microenvironment alterations are critical factors known to affect tumor response to immunotherapy. Here we analyze the association between the mutational signature of tobacco smoking, tumor mutational load, and metrics of immune activity in squamous cell carcinomas arising in the head and neck and lung. Methods: Using RNA and DNA sequencing data from The Cancer Genome Atlas head and neck (HNSC; n = 287) and lung (LUSC; n = 130) squamous cell carcinoma data sets and two independent gene expression data sets (HNSC, n = 136; LUSC, n = 75), we examined associations between the mutational smoking signature, mutation count, immune cell infiltration, cytolytic activity, and interferon-γ signaling. Results: An increasing mutational smoking signature was associated with statistically significantly increased overall mutational load in both HNSC (ρ = .33, P = 1.01 × 10-7) and LUSC (ρ = .49, P = 2.80 × 10-9). In HNSC, a higher mutational smoking signature was associated with lower levels of immune infiltration (ρ = -.37, P = 1.29 × 10-10), cytolytic activity (ρ = -.28, P = 4.07 × 10-6), and interferon-γ pathway signaling (ρ = .39, P = 3.20 × 10-11). In LUSC, these associations were reversed (ρ = .19, P = .03; ρ = .20, P = .02; and ρ = .18, P = .047, respectively). Differentially expressed genes between smoking-high and smoking-low tumors revealed broad tobacco-induced immunosuppression in HNSC, in contrast to a tumor-inflamed microenvironment in smokers with LUSC. Conclusions: In squamous cell carcinomas, the genetic smoking signature is associated with higher mutational load, but variable effects on tumor immunity can occur, depending on anatomic site. In HNSC, smoking is predominantly immunosuppressive; in LUSC, more pro-inflammatory. Both tumor mutation load and immune microenvironment affect clinical response to immunotherapy. Thus, the mutational smoking signature is likely to have relevance for immunotherapeutic investigation in smoking-associated cancers.


Subject(s)
Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/pathology , Tobacco Smoking/adverse effects , Tumor Microenvironment/immunology , Carcinoma, Squamous Cell/mortality , Computational Biology/methods , Databases, Genetic , Disease Susceptibility , Gene Expression Profiling , Humans , Mutation , Prognosis , Tumor Microenvironment/genetics
7.
Cell Rep ; 20(7): 1623-1640, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28813674

ABSTRACT

At the root of most fatal malignancies are aberrantly activated transcriptional networks that drive metastatic dissemination. Although individual metastasis-associated genes have been described, the complex regulatory networks presiding over the initiation and maintenance of metastatic tumors are still poorly understood. There is untapped value in identifying therapeutic targets that broadly govern coordinated transcriptional modules dictating metastatic progression. Here, we reverse engineered and interrogated a breast cancer-specific transcriptional interaction network (interactome) to define transcriptional control structures causally responsible for regulating genetic programs underlying breast cancer metastasis in individual patients. Our analyses confirmed established pro-metastatic transcription factors, and they uncovered TRIM25 as a key regulator of metastasis-related transcriptional programs. Further, in vivo analyses established TRIM25 as a potent regulator of metastatic disease and poor survival outcome. Our findings suggest that identifying and targeting keystone proteins, like TRIM25, can effectively collapse transcriptional hierarchies necessary for metastasis formation, thus representing an innovative cancer intervention strategy.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Transcription Factors/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Female , Gene Regulatory Networks , Genes, Reporter , Heterografts , Humans , Luciferases/genetics , Luciferases/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Mice , Mice, Nude , Neoplasm Proteins/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Signal Transduction , Survival Analysis , Systems Biology , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
8.
Nat Commun ; 8(1): 1197, 2017 10 30.
Article in English | MEDLINE | ID: mdl-29084941

ABSTRACT

Myoepithelial carcinoma (MECA) is an aggressive salivary gland cancer with largely unknown genetic features. Here we comprehensively analyze molecular alterations in 40 MECAs using integrated genomic analyses. We identify a low mutational load, and high prevalence (70%) of oncogenic gene fusions. Most fusions involve the PLAG1 oncogene, which is associated with PLAG1 overexpression. We find FGFR1-PLAG1 in seven (18%) cases, and the novel TGFBR3-PLAG1 fusion in six (15%) cases. TGFBR3-PLAG1 promotes a tumorigenic phenotype in vitro, and is absent in 723 other salivary gland tumors. Other novel PLAG1 fusions include ND4-PLAG1; a fusion between mitochondrial and nuclear DNA. We also identify higher number of copy number alterations as a risk factor for recurrence, independent of tumor stage at diagnosis. Our findings indicate that MECA is a fusion-driven disease, nominate TGFBR3-PLAG1 as a hallmark of MECA, and provide a framework for future diagnostic and therapeutic research in this lethal cancer.


Subject(s)
Genomics/methods , Myoepithelioma/genetics , Oncogene Fusion/genetics , Oncogene Proteins, Fusion/genetics , Salivary Gland Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , DNA-Binding Proteins/genetics , Female , HEK293 Cells , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Mutation , Receptor, Fibroblast Growth Factor, Type 1/genetics , Sequence Analysis, DNA/methods , Young Adult
9.
Methods Mol Biol ; 1351: 67-79, 2016.
Article in English | MEDLINE | ID: mdl-26530675

ABSTRACT

Isolation of mitochondria from cultured cells and animal tissues for analysis of nucleic acids and bona fide mitochondrial nucleic acid binding proteins and enzymes is complicated by contamination with cellular nucleic acids and their adherent proteins. Protocols presented here allow for quick isolation of mitochondria from a small number of cells and for preparation of highly purified mitochondria from a larger number of cells using nuclease treatment and high salt washing of mitochondria to reduce contamination. We further describe a method for the isolation of mitochondrial DNA-protein complexes known as nucleoids from these highly purified mitochondria using a combination of glycerol gradient sedimentation followed by isopycnic centrifugation in a non-ionic iodixanol gradient.


Subject(s)
Centrifugation, Density Gradient/methods , Centrifugation, Isopycnic/methods , DNA, Mitochondrial/analysis , DNA-Binding Proteins/analysis , RNA/analysis , Animals , Cell Line , Cell Nucleus/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/isolation & purification , DNA-Binding Proteins/isolation & purification , HeLa Cells , Humans , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Ribosomes/chemistry , RNA/genetics , RNA/isolation & purification , RNA, Mitochondrial , Triiodobenzoic Acids/chemistry
10.
JCI Insight ; 1(17): e89829, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27777979

ABSTRACT

Recent clinical trials have demonstrated a clear survival advantage in advanced head and neck squamous cell carcinoma (HNSCC) patients treated with immune checkpoint blockade. These emerging results reveal that HNSCC is one of the most promising frontiers for immunotherapy research. However, further progress in head and neck immuno-oncology will require a detailed understanding of the immune infiltrative landscape found in these tumors. We leveraged transcriptome data from 280 tumors profiled by The Cancer Genome Atlas (TCGA) to comprehensively characterize the immune landscape of HNSCC in order to develop a rationale for immunotherapeutic strategies in HNSCC and guide clinical investigation. We find that both HPV+ and HPV- HNSCC tumors are among the most highly immune-infiltrated cancer types. Strikingly, HNSCC had the highest median Treg/CD8+ T cell ratio and the highest levels of CD56dim NK cell infiltration, in our pan-cancer analysis of the most immune-infiltrated tumors. CD8+ T cell infiltration and CD56dim NK cell infiltration each correlated with superior survival in HNSCC. Tumors harboring genetic smoking signatures had lower immune infiltration and were associated with poorer survival, suggesting these patients may benefit from immune agonist therapy. These findings illuminate the immune landscape of HPV+ and HPV- HNSCC. Additionally, this landscape provides a potentially novel rationale for investigation of agents targeting modulators of Tregs (e.g., CTLA-4, GITR, ICOS, IDO, and VEGFA) and NK cells (e.g., KIR, TIGIT, and 4-1BB) as adjuncts to anti-PD-1 in the treatment of advanced HNSCC.


Subject(s)
Carcinoma, Squamous Cell/immunology , Head and Neck Neoplasms/immunology , Immunotherapy , CD8-Positive T-Lymphocytes/cytology , Head and Neck Neoplasms/genetics , Humans , Killer Cells, Natural/cytology , Papillomaviridae , Smoking , T-Lymphocytes, Regulatory/cytology , Transcriptome , Tumor Microenvironment
11.
Clin Cancer Res ; 22(18): 4623-33, 2016 Sep 15.
Article in English | MEDLINE | ID: mdl-27103403

ABSTRACT

PURPOSE: Salivary duct carcinoma (SDC) is an aggressive salivary malignancy, which is resistant to chemotherapy and has high mortality rates. We investigated the molecular landscape of SDC, focusing on genetic alterations and gene expression profiles. EXPERIMENTAL DESIGN: We performed whole-exome sequencing, RNA sequencing, and immunohistochemical analyses in 16 SDC tumors and examined selected alterations via targeted sequencing of 410 genes in a second cohort of 15 SDCs. RESULTS: SDCs harbored a higher mutational burden than many other salivary carcinomas (1.7 mutations/Mb). The most frequent genetic alterations were mutations in TP53 (55%), HRAS (23%), PIK3CA (23%), and amplification of ERBB2 (35%). Most (74%) tumors had alterations in either MAPK (BRAF/HRAS/NF1) genes or ERBB2 Potentially targetable alterations based on supportive clinical evidence were present in 61% of tumors. Androgen receptor (AR) was overexpressed in 75%; several potential resistance mechanisms to androgen deprivation therapy (ADT) were identified, including the AR-V7 splice variant (present in 50%, often at low ratios compared with full-length AR) and FOXA1 mutations (10%). Consensus clustering and pathway analyses in transcriptome data revealed striking similarities between SDC and molecular apocrine breast cancer. CONCLUSIONS: This study illuminates the landscape of genetic alterations and gene expression programs in SDC, identifying numerous molecular targets and potential determinants of response to AR antagonism. This has relevance for emerging clinical studies of ADT and other targeted therapies in SDC. The similarities between SDC and apocrine breast cancer indicate that clinical data in breast cancer may generate useful hypotheses for SDC. Clin Cancer Res; 22(18); 4623-33. ©2016 AACR.


Subject(s)
Apocrine Glands/pathology , Biomarkers, Tumor , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal/genetics , Salivary Gland Neoplasms/genetics , Aged , Alleles , Apocrine Glands/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Carcinoma, Ductal/metabolism , Carcinoma, Ductal/pathology , Carcinoma, Ductal/therapy , DNA Copy Number Variations , Female , Gene Expression , Gene Expression Profiling , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Mutation , Neoplasm Staging , Receptors, Androgen/metabolism , Recurrence , Salivary Gland Neoplasms/metabolism , Salivary Gland Neoplasms/pathology , Salivary Gland Neoplasms/therapy , Signal Transduction , Treatment Outcome , Tumor Burden , Exome Sequencing
12.
Clin Toxicol (Phila) ; 49(1): 34-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21114414

ABSTRACT

OBJECTIVE: To evaluate an algorithmic approach involving a multidisciplinary team for causality assessment of suspected herb-induced liver injury (HILI) cases and to compare the causality score using this multidisciplinary approach and Roussel Uclaf Causality Assessment Method (RUCAM). METHODS: A team consisting of hepatologist, clinical toxicologist, analytical toxicologist, and Chinese medicine (CM) pharmacist was formed to do causality assessment based on a protocol for suspected HILI cases. The likelihood of the diagnosis of individual case was first assessed systematically by a hepatologist and clinical toxicologist independently after collecting information about four aspects: (1) clinical course, (2) exclusion of alternative causes, (3) quality of the prescription and herbal product by examining the CM prescriptions and analysis of biological and herb samples, (4) scientific support on comprehensive literature review on English and Chinese medical database, and subsequently concluded in a consensus meeting held by the multidisciplinary team. The final causality score of each patient was compared with the likelihood of causality as assessed by RUCAM. RESULTS: Between 2005 and 2007, 48 consecutive patients with suspected HILI were enrolled and 21 patients were excluded due to the establishment of an alternative cause of liver impairment or the lack of any information on the herbs taken. Twenty-seven patients were recruited, among them 15 consumed Chinese herbal medicines, 10 used proprietary Chinese medicinal products, and 2 used both. The concordance between the causality assessment of the hepatologist and clinical toxicologist was moderate (weighted κ = 0.48, 95%CI 0.30-0.66). The causality assessment process concluded that the likelihood of HILI was "highly probable" in 5 cases and "probable" in 12, whereas there were 5 "highly probable" and 16 "probable" cases as assessed by RUCAM. The causality assessment by the multidisciplinary approach and RUCAM also showed moderate agreement (weighted κ= 0.51, 95%CI 0.22-0.81). CONCLUSION: A multidisciplinary approach using defined algorithms is a scientific approach in causality assessment for HILI. Further study is needed to assess its accuracy and applicability.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Drugs, Chinese Herbal/poisoning , Risk Assessment , Adult , Aged , Algorithms , Female , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL