Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
2.
PLoS Genet ; 19(11): e1011043, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38033156

ABSTRACT

A prominent source of mutation in cancer is single-stranded DNA cytosine deamination by cellular APOBEC3 enzymes, which results in signature C-to-T and C-to-G mutations in TCA and TCT motifs. Although multiple enzymes have been implicated, reports conflict and it is unclear which protein(s) are responsible. Here we report the development of a selectable system to quantify genome mutation and demonstrate its utility by comparing the mutagenic activities of three leading candidates-APOBEC3A, APOBEC3B, and APOBEC3H. The human cell line, HAP1, is engineered to express the thymidine kinase (TK) gene of HSV-1, which confers sensitivity to ganciclovir. Expression of APOBEC3A and APOBEC3B, but not catalytic mutant controls or APOBEC3H, triggers increased frequencies of TK mutation and similar TC-biased cytosine mutation profiles in the selectable TK reporter gene. Whole genome sequences from independent clones enabled an analysis of thousands of single base substitution mutations and extraction of local sequence preferences with APOBEC3A preferring YTCW motifs 70% of the time and APOBEC3B 50% of the time (Y = C/T; W = A/T). Signature comparisons with breast tumor whole genome sequences indicate that most malignancies manifest intermediate percentages of APOBEC3 signature mutations in YTCW motifs, mostly between 50 and 70%, suggesting that both enzymes contribute in a combinatorial manner to the overall mutation landscape. Although the vast majority of APOBEC3A- and APOBEC3B-induced single base substitution mutations occur outside of predicted chromosomal DNA hairpin structures, whole genome sequence analyses and supporting biochemical studies also indicate that both enzymes are capable of deaminating the single-stranded loop regions of DNA hairpins at elevated rates. These studies combine to help resolve a long-standing etiologic debate on the source of APOBEC3 signature mutations in cancer and indicate that future diagnostic and therapeutic efforts should focus on both APOBEC3A and APOBEC3B.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/genetics , Mutation , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Cell Line , DNA/metabolism , Minor Histocompatibility Antigens/genetics , Cytosine/metabolism
3.
Nat Genet ; 55(10): 1721-1734, 2023 10.
Article in English | MEDLINE | ID: mdl-37735199

ABSTRACT

The single-stranded DNA cytosine-to-uracil deaminase APOBEC3B is an antiviral protein implicated in cancer. However, its substrates in cells are not fully delineated. Here APOBEC3B proteomics reveal interactions with a surprising number of R-loop factors. Biochemical experiments show APOBEC3B binding to R-loops in cells and in vitro. Genetic experiments demonstrate R-loop increases in cells lacking APOBEC3B and decreases in cells overexpressing APOBEC3B. Genome-wide analyses show major changes in the overall landscape of physiological and stimulus-induced R-loops with thousands of differentially altered regions, as well as binding of APOBEC3B to many of these sites. APOBEC3 mutagenesis impacts genes overexpressed in tumors and splice factor mutant tumors preferentially, and APOBEC3-attributed kataegis are enriched in RTCW motifs consistent with APOBEC3B deamination. Taken together with the fact that APOBEC3B binds single-stranded DNA and RNA and preferentially deaminates DNA, these results support a mechanism in which APOBEC3B regulates R-loops and contributes to R-loop mutagenesis in cancer.


Subject(s)
Neoplasms , R-Loop Structures , Humans , DNA, Single-Stranded/genetics , Genome-Wide Association Study , Mutagenesis , Neoplasms/genetics , Neoplasms/pathology , Cytidine Deaminase/genetics , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism
4.
Cancer Cell ; 41(1): 164-180.e8, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36563682

ABSTRACT

Therapy resistance is a major challenge in the treatment of cancer. Here, we performed CRISPR-Cas9 screens across a broad range of therapies used in acute myeloid leukemia to identify genomic determinants of drug response. Our screens uncover a selective dependency on RNA splicing factors whose loss preferentially enhances response to the BCL2 inhibitor venetoclax. Loss of the splicing factor RBM10 augments response to venetoclax in leukemia yet is completely dispensable for normal hematopoiesis. Combined RBM10 and BCL2 inhibition leads to mis-splicing and inactivation of the inhibitor of apoptosis XIAP and downregulation of BCL2A1, an anti-apoptotic protein implicated in venetoclax resistance. Inhibition of splicing kinase families CLKs (CDC-like kinases) and DYRKs (dual-specificity tyrosine-regulated kinases) leads to aberrant splicing of key splicing and apoptotic factors that synergize with venetoclax, and overcomes resistance to BCL2 inhibition. Our findings underscore the importance of splicing in modulating response to therapies and provide a strategy to improve venetoclax-based treatments.


Subject(s)
Leukemia, Myeloid, Acute , Proto-Oncogene Proteins c-bcl-2 , Humans , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Cell Line, Tumor , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , RNA Splicing/genetics , Leukemia, Myeloid, Acute/genetics , Protein-Tyrosine Kinases , Apoptosis/genetics , RNA-Binding Proteins/genetics
5.
Histopathology ; 82(4): 608-621, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36416305

ABSTRACT

AIMS: Primary head/neck mucosal melanomas (MMs) are rare and exhibit aggressive biologic behaviour and elevated mutational loads. The molecular mechanisms responsible for high genomic instability observed in head/neck MMs remain elusive. The DNA cytosine deaminase APOBEC3B (A3B) constitutes a major endogenous source of mutation in human cancer. A3B-related mutations are identified through C-to-T/-G base substitutions in 5'-TCA/T motifs. Herein, we present immunohistochemical and genomic data supportive of a role for A3B in head/neck MMs. METHODS AND RESULTS: A3B protein levels were assessed in oral (n = 13) and sinonasal (n = 13) melanomas, and oral melanocytic nevi (n = 13) by immunohistochemistry using a custom rabbit α-A3B mAb (5210-87-13). Heterogeneous, selective-to-diffuse, nuclear only, A3B immunopositivity was observed in 12 of 13 (92.3%) oral melanomas (H-score range = 9-72, median = 40) and 8 of 13 (62%) sinonasal melanomas (H-score range = 1-110, median = 24). Two cases negative for A3B showed prominent cytoplasmic staining consistent with A3G. A3B protein levels were significantly higher in oral and sinonasal MMs than intraoral melanocytic nevi (P < 0.0001 and P = 0.0022, respectively), which were A3B-negative (H-score range = 1-8, median = 4). A3B levels, however, did not differ significantly between oral and sinonasal tumours (P > 0.99). NGS performed in 10 sinonasal MMs revealed missense NRAS mutations in 50% of the studied cases and one each KIT and HRAS mutations. Publicly available whole-genome sequencing (WGS) data disclosed that the number of C-to-T mutations and APOBEC3 enrichment score were markedly elevated in head/neck MMs (n = 2). CONCLUSION: The above data strongly indicate a possible role for the mutagenic enzyme A3B in head/neck melanomagenesis, but not benign melanocytic neoplasms.


Subject(s)
Melanoma , Mouth Neoplasms , Nevus, Pigmented , Paranasal Sinus Neoplasms , Animals , Humans , Rabbits , Melanoma/pathology , Mutation , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Cytidine Deaminase/genetics
6.
J Med Imaging Radiat Oncol ; 66(3): 319-323, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34250746

ABSTRACT

INTRODUCTION: Prostate cancer diagnosis is shifting towards a minimally invasive approach, maintaining accuracy and efficacy while reducing morbidity. We aimed to assess if PSMA-Ga68 PET/CT can accurately grade and localise prostatic malignancy using objective methods, compared with pathology and MRI. METHODS: Retrospective analysis on 114 consecutive patients undergoing staging PSMA PET/CT scans over 12 months was carried out. The SUVmax and site of highest PSMA activity within the prostate gland were recorded. Pathology/biopsy review assessed maximum Gleason score (and location). MRI analysis assessed the highest PIRADS score and location. The grade, location and size of malignant tissue on biopsy, and PSA, were correlated with the SUVmax and the PIRADS score. RESULTS: SUVmax was significantly elevated in cases with PSA ≥10 (P = 0.003) and Gleason score ≥8 (P = 0.0002). SUVmax demonstrated equivalent sensitivity to MRI-PIRADS in predicting Gleason ≥8 disease, with higher specificity when tested under a high-specificity regime (SUVmax ≥10, PIRADS = 5, P = 0.002). Furthermore, the region of highest SUVmax was superior to MRI-PIRADS for localising the highest grade tumour region, correctly identifying 71% of highest grade regions compared to 54% with MRI (P = 0.015). CONCLUSION: PSMA PET/CT is as effective as MRI in identifying high-grade prostate malignancy. Our findings also support previous studies in showing a significant relationship between SUVmax and Gleason grade. These benefits, along with the known advantage in identifying distant metastases and the reduced cost, further support the argument that PSMA PET/CT should be offered as an initial investigation in the workup of prostate cancer.


Subject(s)
Positron Emission Tomography Computed Tomography , Prostatic Neoplasms , Humans , Magnetic Resonance Imaging , Male , Positron Emission Tomography Computed Tomography/methods , Prostate-Specific Antigen , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Retrospective Studies
7.
J Speech Lang Hear Res ; 64(1): 1-15, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33285082

ABSTRACT

Purpose Successful voice therapy requires the patient to learn new vocal behaviors, but little is currently known regarding how vocal motor skills are improved and retained. To quantitatively characterize the motor learning process in a clinically meaningful context, a virtual task was developed based on the Vocal Function Exercises. In the virtual task, subjects control a computational model of a ball floating on a column of airflow via modifications to mean airflow (L/s) and intensity (dB-C) to keep the ball within a target range representing a normative ratio (dB × s/L). Method One vocally healthy female and one female with nonphonotraumatic vocal hyperfunction practiced the task for 11 days and completed retention testing 1 and 6 months later. The mapping between the two execution variables (airflow and intensity) and one error measure (proximity to the normative ratio) was evaluated by quantifying distributional variability (tolerance cost and noise cost) and temporal variability (scaling index of detrended fluctuation analysis). Results Both subjects reduced their error over practice and retained their performance 6 months later. Tolerance cost and noise cost were positively correlated with decreases in error during early practice and late practice, respectively. After extended practice, temporal variability was modulated to align with the task's solution manifold. Conclusions These case studies illustrated, in a healthy control and a patient with nonphonotraumatic vocal hyperfunction, that the virtual floating ball task produces quantitative measures characterizing the learning process. Future work will further investigate the task's potential to enhance clinical assessment and treatments involving voice control. Supplemental Material https://doi.org/10.23641/asha.13322891.


Subject(s)
Voice Training , Voice , Exercise , Female , Humans , Learning , Motor Skills
8.
Mod Pathol ; 34(2): 280-290, 2021 02.
Article in English | MEDLINE | ID: mdl-32632179

ABSTRACT

The DNA cytosine deaminase APOBEC3B (A3B) is a newly recognized endogenous source of mutations in a range of human tumors, including head/neck cancer. A3B inflicts C-to-T and C-to-G base substitutions in 5'-TCA/T trinucleotide motifs, contributes to accelerated rates of tumor development, and affects clinical outcomes in a variety of cancer types. High-risk human papillomavirus (HPV) infection causes A3B overexpression, and HPV-positive cervical and head/neck cancers are among tumor types with the highest degree of APOBEC signature mutations. A3B overexpression in HPV-positive tumor types is caused by the viral E6/E7 oncoproteins and may be an early off-to-on switch in tumorigenesis. In comparison, less is known about the molecular mechanisms responsible for A3B overexpression in HPV-negative head/neck cancers. Here, we utilize an immunohistochemical approach to determine whether A3B is turned from off-to-on or if it undergoes a more gradual transition to overexpression in HPV-negative head/neck cancers. As positive controls, almost all HPV-positive oral epithelial dysplasias and oropharyngeal cancers showed high levels of nuclear A3B staining regardless of diagnosis. As negative controls, A3B levels were low in phenotypically normal epithelium adjacent to cancer and oral epithelial hyperplasias. Interestingly, HPV-negative and low-grade oral epithelial dysplasias showed intermediate A3B levels, while high-grade oral dysplasias showed high A3B levels similar to oral squamous cell carcinomas. A3B levels were highest in grade 2 and grade 3 oral squamous cell carcinomas. In addition, a strong positive association was found between nuclear A3B and Ki67 scores suggesting a linkage to the cell cycle. Overall, these results support a model in which gradual activation of A3B expression occurs during HPV-negative tumor development and suggest that A3B overexpression may provide a marker for advanced grade oral dysplasia and cancer.


Subject(s)
Cytidine Deaminase/metabolism , Minor Histocompatibility Antigens/metabolism , Mouth Neoplasms/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Mouth Mucosa/pathology , Mouth Neoplasms/virology , Papillomavirus Infections/complications , Precancerous Conditions/metabolism , Precancerous Conditions/virology , Squamous Cell Carcinoma of Head and Neck/virology
9.
J Exp Med ; 217(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-32870257

ABSTRACT

The APOBEC3 family of antiviral DNA cytosine deaminases is implicated as the second largest source of mutation in cancer. This mutational process may be a causal driver or inconsequential passenger to the overall tumor phenotype. We show that human APOBEC3A expression in murine colon and liver tissues increases tumorigenesis. All other APOBEC3 family members, including APOBEC3B, fail to promote liver tumor formation. Tumor DNA sequences from APOBEC3A-expressing animals display hallmark APOBEC signature mutations in TCA/T motifs. Bioinformatic comparisons of the observed APOBEC3A mutation signature in murine tumors, previously reported APOBEC3A and APOBEC3B mutation signatures in yeast, and reanalyzed APOBEC mutation signatures in human tumor datasets support cause-and-effect relationships for APOBEC3A-catalyzed deamination and mutagenesis in driving multiple human cancers.


Subject(s)
Biocatalysis , Carcinogenesis/genetics , Cytidine Deaminase/genetics , Mutation/genetics , Proteins/genetics , Adenomatous Polyposis Coli/pathology , Animals , Base Sequence , Carcinogenesis/pathology , DNA Transposable Elements/genetics , Humans , Hydrolases/genetics , Intestinal Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Regeneration , Loss of Heterozygosity/genetics , Mice, Transgenic , Polyps/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Elife ; 92020 09 28.
Article in English | MEDLINE | ID: mdl-32985974

ABSTRACT

APOBEC3B (A3B)-catalyzed DNA cytosine deamination contributes to the overall mutational landscape in breast cancer. Molecular mechanisms responsible for A3B upregulation in cancer are poorly understood. Here we show that a single E2F cis-element mediates repression in normal cells and that expression is activated by its mutational disruption in a reporter construct or the endogenous A3B gene. The same E2F site is required for A3B induction by polyomavirus T antigen indicating a shared molecular mechanism. Proteomic and biochemical experiments demonstrate the binding of wildtype but not mutant E2F promoters by repressive PRC1.6/E2F6 and DREAM/E2F4 complexes. Knockdown and overexpression studies confirm the involvement of these repressive complexes in regulating A3B expression. Altogether, these studies demonstrate that A3B expression is suppressed in normal cells by repressive E2F complexes and that viral or mutational disruption of this regulatory network triggers overexpression in breast cancer and provides fuel for tumor evolution.


Subject(s)
Cytidine Deaminase/genetics , E2F Transcription Factors/genetics , Minor Histocompatibility Antigens/genetics , Signal Transduction , Cytidine Deaminase/metabolism , E2F Transcription Factors/metabolism , HEK293 Cells , Humans , MCF-7 Cells , Minor Histocompatibility Antigens/metabolism , Protein Binding
11.
Clin Cancer Res ; 26(13): 3397-3407, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32060098

ABSTRACT

PURPOSE: Clear cell ovarian carcinoma (CCOC) is an aggressive disease that often demonstrates resistance to standard chemotherapies. Approximately 25% of patients with CCOC show a strong APOBEC mutation signature. Here, we determine which APOBEC3 enzymes are expressed in CCOC, establish clinical correlates, and identify a new biomarker for detection and intervention. EXPERIMENTAL DESIGNS: APOBEC3 expression was analyzed by IHC and qRT-PCR in a pilot set of CCOC specimens (n = 9 tumors). The IHC analysis of APOBEC3B was extended to a larger cohort to identify clinical correlates (n = 48). Dose-response experiments with platinum-based drugs in CCOC cell lines and carboplatin treatment of patient-derived xenografts (PDXs) were done to address mechanistic linkages. RESULTS: One DNA deaminase, APOBEC3B, is overexpressed in a formidable subset of CCOC tumors and is low or absent in normal ovarian and fallopian tube epithelial tissues. High APOBEC3B expression associates with improved progression-free survival (P = 0.026) and moderately with overall survival (P = 0.057). Cell-based studies link APOBEC3B activity and subsequent uracil processing to sensitivity to cisplatin and carboplatin. PDX studies extend this mechanistic relationship to CCOC tissues. CONCLUSIONS: These studies demonstrate that APOBEC3B is overexpressed in a subset of CCOC and, contrary to initial expectations, associated with improved (not worse) clinical outcomes. A likely molecular explanation is that APOBEC3B-induced DNA damage sensitizes cells to additional genotoxic stress by cisplatin. Thus, APOBEC3B is a molecular determinant and a candidate predictive biomarker of the therapeutic response to platinum-based chemotherapy. These findings may have broader translational relevance, as APOBEC3B is overexpressed in many different cancer types.


Subject(s)
Cytidine Deaminase/metabolism , Minor Histocompatibility Antigens/metabolism , Ovarian Neoplasms/metabolism , Platinum/pharmacology , Animals , Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cytidine Deaminase/genetics , Disease Models, Animal , Drug Resistance, Neoplasm , Female , Gene Expression , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Mice , Minor Histocompatibility Antigens/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Prognosis , Synthetic Lethal Mutations/drug effects , Synthetic Lethal Mutations/genetics , Xenograft Model Antitumor Assays
12.
Cell Rep ; 29(5): 1057-1065.e4, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31665623

ABSTRACT

HIV-1 Vif hijacks a cellular ubiquitin ligase complex to degrade antiviral APOBEC3 enzymes and PP2A phosphatase regulators (PPP2R5A-E). APOBEC3 counteraction is essential for viral pathogenesis. However, Vif also functions through an unknown mechanism to induce G2 cell cycle arrest. Here, deep mutagenesis is used to define the Vif surface required for PPP2R5 degradation and isolate a panel of separation-of-function mutants (PPP2R5 degradation-deficient and APOBEC3G degradation-proficient). Functional studies with Vif and PPP2R5 mutants were combined to demonstrate that PPP2R5 is, in fact, the target Vif degrades to induce G2 arrest. Pharmacologic and genetic approaches show that direct modulation of PP2A function or depletion of specific PPP2R5 proteins causes an indistinguishable arrest phenotype. Vif function in the cell cycle checkpoint is present in common HIV-1 subtypes worldwide and likely advantageous for viral pathogenesis.


Subject(s)
Cell Cycle Checkpoints , Protein Phosphatase 2/metabolism , Proteolysis , vif Gene Products, Human Immunodeficiency Virus/metabolism , Cell Line , G2 Phase Cell Cycle Checkpoints , High-Throughput Nucleotide Sequencing , Humans , Phosphorylation , Protein Binding , Reproducibility of Results , Static Electricity , Substrate Specificity
13.
Proc Natl Acad Sci U S A ; 116(44): 22158-22163, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611371

ABSTRACT

Human cells express up to 9 active DNA cytosine deaminases with functions in adaptive and innate immunity. Many cancers manifest an APOBEC mutation signature and APOBEC3B (A3B) is likely the main enzyme responsible. Although significant numbers of APOBEC signature mutations accumulate in tumor genomes, the majority of APOBEC-catalyzed uracil lesions are probably counteracted in an error-free manner by the uracil base excision repair pathway. Here, we show that A3B-expressing cells can be selectively killed by inhibiting uracil DNA glycosylase 2 (UNG) and that this synthetic lethal phenotype requires functional mismatch repair (MMR) proteins and p53. UNG knockout human 293 and MCF10A cells elicit an A3B-dependent death. This synthetic lethal phenotype is dependent on A3B catalytic activity and reversible by UNG complementation. A3B expression in UNG-null cells causes a buildup of genomic uracil, and the ensuing lethality requires processing of uracil lesions (likely U/G mispairs) by MSH2 and MLH1 (likely noncanonical MMR). Cancer cells expressing high levels of endogenous A3B and functional p53 can also be killed by expressing an UNG inhibitor. Taken together, UNG-initiated base excision repair is a major mechanism counteracting genomic mutagenesis by A3B, and blocking UNG is a potential strategy for inducing the selective death of tumors.


Subject(s)
Cell Death , Cytidine Deaminase/genetics , DNA Glycosylases/genetics , APOBEC Deaminases , Cell Line, Tumor , DNA Glycosylases/antagonists & inhibitors , DNA Mismatch Repair , DNA Repair , Gene Knockout Techniques , Humans , Models, Molecular , Ubiquitination
14.
J Virol ; 93(23)2019 12 01.
Article in English | MEDLINE | ID: mdl-31534038

ABSTRACT

An integral part of the antiviral innate immune response is the APOBEC3 family of single-stranded DNA cytosine deaminases, which inhibits virus replication through deamination-dependent and -independent activities. Viruses have evolved mechanisms to counteract these enzymes, such as HIV-1 Vif-mediated formation of a ubiquitin ligase to degrade virus-restrictive APOBEC3 enzymes. A new example is Epstein-Barr virus (EBV) ribonucleotide reductase (RNR)-mediated inhibition of cellular APOBEC3B (A3B). The large subunit of the viral RNR, BORF2, causes A3B relocalization from the nucleus to cytoplasmic bodies and thereby protects viral DNA during lytic replication. Here, we use coimmunoprecipitation and immunofluorescence microscopy approaches to ask whether this mechanism is shared with the closely related gammaherpesvirus Kaposi's sarcoma-associated herpesvirus (KSHV) and the more distantly related alphaherpesvirus herpes simplex virus 1 (HSV-1). The large RNR subunit of KSHV, open reading frame 61 (ORF61), coprecipitated multiple APOBEC3s, including A3B and APOBEC3A (A3A). KSHV ORF61 also caused relocalization of these two enzymes to perinuclear bodies (A3B) and to oblong cytoplasmic structures (A3A). The large RNR subunit of HSV-1, ICP6, also coprecipitated A3B and A3A and was sufficient to promote the relocalization of these enzymes from nuclear to cytoplasmic compartments. HSV-1 infection caused similar relocalization phenotypes that required ICP6. However, unlike the infectivity defects previously reported for BORF2-null EBV, ICP6 mutant HSV-1 showed normal growth rates and plaque phenotypes. Combined, these results indicate that both gamma- and alphaherpesviruses use a conserved RNR-dependent mechanism to relocalize A3B and A3A and furthermore suggest that HSV-1 possesses at least one additional mechanism to neutralize these antiviral enzymes.IMPORTANCE The APOBEC3 family of DNA cytosine deaminases constitutes a vital innate immune defense against a range of different viruses. A novel counterrestriction mechanism has recently been uncovered for the gammaherpesvirus EBV, in which a subunit of the viral protein known to produce DNA building blocks (ribonucleotide reductase) causes A3B to relocalize from the nucleus to the cytosol. Here, we extend these observations with A3B to include a closely related gammaherpesvirus, KSHV, and a more distantly related alphaherpesvirus, HSV-1. These different viral ribonucleotide reductases also caused relocalization of A3A, which is 92% identical to A3B. These studies are important because they suggest a conserved mechanism of APOBEC3 evasion by large double-stranded DNA herpesviruses. Strategies to block this host-pathogen interaction may be effective for treating infections caused by these herpesviruses.


Subject(s)
Cytidine Deaminase/metabolism , Ribonucleotide Reductases/metabolism , Viral Proteins/metabolism , APOBEC Deaminases , Cell Line , Cytosine Deaminase/metabolism , HEK293 Cells , Herpes Simplex , Herpesviridae Infections , Herpesvirus 1, Human/metabolism , Herpesvirus 4, Human/metabolism , Herpesvirus 8, Human/metabolism , Host-Pathogen Interactions , Humans , Immunity, Innate , Minor Histocompatibility Antigens/metabolism , Proteins/metabolism , Virus Replication
15.
J Gen Virol ; 100(7): 1140-1152, 2019 07.
Article in English | MEDLINE | ID: mdl-31145054

ABSTRACT

HIV-1 replication in CD4-positive T lymphocytes requires counteraction of multiple different innate antiviral mechanisms. Macrophage cells are also thought to provide a reservoir for HIV-1 replication but less is known in this cell type about virus restriction and counteraction mechanisms. Many studies have combined to demonstrate roles for APOBEC3D, APOBEC3F, APOBEC3G and APOBEC3H in HIV-1 restriction and mutation in CD4-positive T lymphocytes, whereas the APOBEC enzymes involved in HIV-1 restriction in macrophages have yet to be delineated fully. We show that multiple APOBEC3 genes including APOBEC3G are expressed in myeloid cell lines such as THP-1. Vif-deficient HIV-1 produced from THP-1 is less infectious than Vif-proficient virus, and proviral DNA resulting from such Vif-deficient infections shows strong G to A mutation biases in the dinucleotide motif preferred by APOBEC3G. Moreover, Vif mutant viruses with selective sensitivity to APOBEC3G show Vif null-like infectivity levels and similarly strong APOBEC3G-biased mutation spectra. Importantly, APOBEC3G-null THP-1 cells yield Vif-deficient particles with significantly improved infectivities and proviral DNA with background levels of G to A hypermutation. These studies combine to indicate that APOBEC3G is the main HIV-1 restricting APOBEC3 family member in THP-1 cells.


Subject(s)
APOBEC-3G Deaminase/metabolism , HIV Infections/enzymology , HIV-1/physiology , APOBEC-3G Deaminase/genetics , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , Host-Pathogen Interactions , Humans , Mutation , Myeloid Cells , THP-1 Cells , Virus Replication , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/metabolism
16.
Zoonoses Public Health ; 66(5): 546-557, 2019 08.
Article in English | MEDLINE | ID: mdl-30848076

ABSTRACT

Rotavirus C (RVC) causes enteric disease in multiple species, including humans, swine, bovines, and canines. To date, the evolutionary relationships of RVC populations circulating in different host species are poorly understood, owing to the low availability of genetic sequence data. To address this gap, we sequenced 45 RVC complete genomes from swine samples collected in the United States and Mexico. A phylogenetic analysis of each genome segment indicates that RVC populations have been evolving independently in human, swine, canine, and bovine hosts for at least the last century, with inter-species transmission events occurring deep in the phylogenetic tree, and none in the last 100 years. Bovine and canine RVC populations clustered together nine of the 11 gene segments, indicating a shared common ancestor centuries ago. The evolutionary relationships of RVC in humans and swine were more complex, due to the extensive genetic diversity and multiple RVC clades identified in pigs, which were not structured geographically. Topological differences between trees inferred for different genome segments occurred frequently, including at nodes deep in the tree, indicating that RVC's evolutionary history includes multiple reassortment events that occurred a long time ago. Overall, we find that RVC is evolving within host-defined lineages, but the evolutionary history of RVC is more complex than previously recognized due to the high genetic diversity of RVC in swine, with a common ancestor dating back centuries. Pigs may act as a reservoir host for RVC, and a source of the lineages identified in other species, including humans, but additional sequencing is needed to understand the full diversity of this understudied pathogen across multiple host species.


Subject(s)
Biological Evolution , Cattle Diseases/virology , Rotavirus Infections/veterinary , Rotavirus/genetics , Swine Diseases/virology , Animals , Cattle , Cattle Diseases/epidemiology , Humans , Mexico/epidemiology , Rotavirus Infections/epidemiology , Rotavirus Infections/virology , Selection, Genetic , Swine , Swine Diseases/epidemiology , United States/epidemiology , Viral Proteins/genetics
17.
mBio ; 10(1)2019 02 05.
Article in English | MEDLINE | ID: mdl-30723127

ABSTRACT

APOBEC3B is a single-stranded DNA cytosine deaminase with beneficial innate antiviral functions. However, misregulated APOBEC3B can also be detrimental by inflicting APOBEC signature C-to-T and C-to-G mutations in genomic DNA of multiple cancer types. Polyomavirus and papillomavirus oncoproteins induce APOBEC3B overexpression, perhaps to their own benefit, but little is known about the cellular mechanisms hijacked by these viruses to do so. Here we investigate the molecular mechanism of APOBEC3B upregulation by the polyomavirus large T antigen. First, we demonstrate that the upregulated APOBEC3B enzyme is strongly nuclear and partially localized to virus replication centers. Second, truncated T antigen (truncT) is sufficient for APOBEC3B upregulation, and the RB-interacting motif (LXCXE), but not the p53-binding domain, is required. Third, genetic knockdown of RB1 alone or in combination with RBL1 and/or RBL2 is insufficient to suppress truncT-mediated induction of APOBEC3B Fourth, CDK4/6 inhibition by palbociclib is also insufficient to suppress truncT-mediated induction of APOBEC3B Last, global gene expression analyses in a wide range of human cancers show significant associations between expression of APOBEC3B and other genes known to be regulated by the RB/E2F axis. These experiments combine to implicate the RB/E2F axis in promoting APOBEC3B transcription, yet they also suggest that the polyomavirus RB-binding motif has at least one additional function in addition to RB inactivation for triggering APOBEC3B upregulation in virus-infected cells.IMPORTANCE The APOBEC3B DNA cytosine deaminase is overexpressed in many different cancers and correlates with elevated frequencies of C-to-T and C-to-G mutations in 5'-TC motifs, oncogene activation, acquired drug resistance, and poor clinical outcomes. The mechanisms responsible for APOBEC3B overexpression are not fully understood. Here, we show that the polyomavirus truncated T antigen (truncT) triggers APOBEC3B overexpression through its RB-interacting motif, LXCXE, which in turn likely modulates the binding of E2F family transcription factors to promote APOBEC3B expression. This work strengthens the mechanistic linkage between active cell cycling, APOBEC3B overexpression, and cancer mutagenesis. Although this mutational mechanism damages cellular genomes, viruses may leverage it to promote evolution, immune escape, and pathogenesis. The cellular portion of the mechanism may also be relevant to nonviral cancers, where genetic mechanisms often activate the RB/E2F axis and APOBEC3B mutagenesis contributes to tumor evolution.


Subject(s)
Antigens, Viral, Tumor/metabolism , Cytidine Deaminase/biosynthesis , Host-Pathogen Interactions , Minor Histocompatibility Antigens/biosynthesis , Polyomavirus/growth & development , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Antigens, Viral, Tumor/genetics , Binding Sites , Cells, Cultured , E2F Transcription Factors/metabolism , Gene Expression Profiling , Humans , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neoplasms/pathology , Retinoblastoma Binding Proteins/metabolism
18.
Nat Microbiol ; 4(1): 78-88, 2019 01.
Article in English | MEDLINE | ID: mdl-30420783

ABSTRACT

The apolipoprotein B messenger RNA editing enzyme, catalytic polypeptide-like (APOBEC) family of single-stranded DNA (ssDNA) cytosine deaminases provides innate immunity against virus and transposon replication1-4. A well-studied mechanism is APOBEC3G restriction of human immunodeficiency virus type 1, which is counteracted by a virus-encoded degradation mechanism1-4. Accordingly, most work has focused on retroviruses with obligate ssDNA replication intermediates and it is unclear whether large double-stranded DNA (dsDNA) viruses may be similarly susceptible to restriction. Here, we show that the large dsDNA herpesvirus Epstein-Barr virus (EBV), which is the causative agent of infectious mononucleosis and multiple cancers5, utilizes a two-pronged approach to counteract restriction by APOBEC3B. Proteomics studies and immunoprecipitation experiments showed that the ribonucleotide reductase large subunit of EBV, BORF26,7, binds APOBEC3B. Mutagenesis mapped the interaction to the APOBEC3B catalytic domain, and biochemical studies demonstrated that BORF2 stoichiometrically inhibits APOBEC3B DNA cytosine deaminase activity. BORF2 also caused a dramatic relocalization of nuclear APOBEC3B to perinuclear bodies. On lytic reactivation, BORF2-null viruses were susceptible to APOBEC3B-mediated deamination as evidenced by lower viral titres, lower infectivity and hypermutation. The Kaposi's sarcoma-associated herpesvirus homologue, ORF61, also bound APOBEC3B and mediated relocalization. These data support a model where the genomic integrity of human γ-herpesviruses is maintained by active neutralization of the antiviral enzyme APOBEC3B.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Herpesvirus 4, Human/metabolism , Herpesvirus 8, Human/metabolism , Ribonucleotide Reductases/metabolism , Viral Proteins/metabolism , CRISPR-Cas Systems , Catalytic Domain/genetics , Cell Line , Genome, Viral/genetics , HEK293 Cells , Herpesvirus 4, Human/growth & development , Humans , Minor Histocompatibility Antigens , RNA Interference , RNA, Small Interfering/genetics , Ribonucleotide Reductases/genetics , Viral Proteins/genetics
19.
JNCI Cancer Spectr ; 2(1)2018 Jan.
Article in English | MEDLINE | ID: mdl-29888758

ABSTRACT

BACKGROUND: Multiple endogenous and exogenous sources of DNA damage contribute to the overall mutation burden in cancer, with distinct and overlapping combinations contributing to each cancer type. Many mutation sources result in characteristic mutation signatures, which can be deduced from tumor genomic DNA sequences. Examples include spontaneous hydrolytic deamination of methyl-cytosine bases in CG motifs (AGEING signature) and C-to-T and C-to-G mutations in 5'-TC(A/T) motifs (APOBEC signature). METHODS: The deconstructSigs R package was used to analyze single base substitution mutation signatures in over 1000 cancer cell lines. Two additional approaches were used to analyze the APOBEC mutation signature. RESULTS: Most cell lines show evidence for multiple mutation signatures. For instance, the AGEING signature, which is the largest source of mutation in most primary tumors, predominates in the majority of cancer cell lines. The APOBEC mutation signature is enriched in cancer cell lines from breast, lung, head/neck, bladder, and cervical cancers, where this signature also comprises a large fraction of all mutations. CONCLUSIONS: The single base substitution mutation signatures of cancer cell lines often reflect those of the original tumors from which they are derived. Cancer cell lines with enrichments for distinct mutation signatures such as APOBEC have the potential to become model systems for fundamental research on the underlying mechanisms and for advancing clinical strategies to exploit these processes.

20.
JPEN J Parenter Enteral Nutr ; 42(8): 1349-1357, 2018 11.
Article in English | MEDLINE | ID: mdl-29701877

ABSTRACT

BACKGROUND: The Augmented Versus Routine Approach to Giving Energy Trial (TARGET) is the largest blinded enteral nutrition (EN) intervention trial evaluating energy delivery to be conducted in the critically ill. To determine the external validity of TARGET results, nutrition practices in intensive care units (ICUs) in Australia and New Zealand (ANZ) are described and compared with international practices. METHODS: This was a retrospective analysis of prospectively collected data for the International Nutrition Surveys, 2007-2013. Data are presented as mean (SD). RESULTS: A total of 17,154 patients (ANZ: n = 2776 vs international n = 14,378) from 923 ICUs (146 and 777, respectively) were included. EN was the most common route of feeding (ANZ: 85%, n = 2365 patients vs international: 84%, n = 12,034; P = .258), and EN concentration was also similar (<1.25 kcal/mL ANZ: 70%, n = 12,396 vs international: 65%, n = 56,891 administrations; P < .001). Protein delivery was substantially below the estimated prescriptions but similar between the regions (0.6 [0.4] g/kg/day vs 0.6 [0.4] g/kg/day; P = .849). Patients in ANZ received slightly more energy (1133 [572] vs 948[536] kcal/day; P < .001), possibly because more energy was prescribed (1947 [348] vs 1747 [376] kcal/day; P < .001), nutrition protocols were more commonly used (98% vs 75%; P < .001) and included recommendations for therapies such as prokinetic agents (87% vs 51%, n = 399; P < .001) and small bowel feeding (62% vs 40%; P < .001) when compared with international ICUs. CONCLUSIONS: Key elements of nutrition practice are similar in ANZ and international ICUs. These data can be used to determine the external validity and relevance of TARGET results.


Subject(s)
Critical Care/methods , Critical Illness/therapy , Enteral Nutrition/methods , Intensive Care Units , Parenteral Nutrition/methods , Aged , Aged, 80 and over , Australia , Cross-Cultural Comparison , Dietary Proteins/administration & dosage , Energy Intake , Female , Gastrointestinal Agents/therapeutic use , Humans , Intestine, Small , Male , Middle Aged , New Zealand , Nutrition Surveys , Nutrition Therapy , Nutritional Status , Nutritional Support/methods , Prospective Studies , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...