Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 184
1.
J Autoimmun ; 123: 102690, 2021 09.
Article En | MEDLINE | ID: mdl-34274825

Follicular CXCR5+ PD-1+ CD8 T cells (CD8 Tfc) arise in multiple models of systemic autoimmunity yet their functional contribution to disease remains in debate. Here we define the follicular localization and functional interactions of CD8 Tfc with B cells during autoimmune disease. The absence of functional T regulatory cells in autoimmunity allows for CD8 Tfc development that then expands with lymphoproliferation. CD8 Tfc are identifiable within the lymph nodes and spleen during systemic autoimmunity, but not during tissue-restricted autoimmune disease. Autoimmune CD8 Tfc cells are polyfunctional, producing helper cytokines IL-21, IL-4, and IFNγ while maintaining cytolytic proteins CD107a, granzyme B, and TNF. During autoimmune disease, IL-2-KO CD8 T cells infiltrate the B cell follicle and germinal center, including the dark zone, and in vitro induce activation-induced cytidine deaminase in naïve B cells via IL-4 secretion. CD8 Tfc represent a unique CD8 T cell population with a diverse effector cytokine repertoire that can contribute to pathogenic autoimmune B cell response.


Autoimmune Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Germinal Center/immunology , T Follicular Helper Cells/immunology , Animals , B-Lymphocytes/immunology , Cytidine Deaminase/biosynthesis , Cytokine-Induced Killer Cells , Female , Male , Mice , Mice, Inbred BALB C
2.
Sci Rep ; 10(1): 14164, 2020 08 25.
Article En | MEDLINE | ID: mdl-32843697

Activation-induced cytidine deaminase (AID) is one kind of the mutant enzymes, which target regulating the immunoglobulin (Ig) gene in Burkitt's lymphoma to initiate class switch recombination (CSR), resulting in c-Myc chromosomal translocation. However, it is not clear that whether AID induces c-Myc/IgH translocation in double-hit lymphoma (DHL) with c-Myc gene translocation. In this study, the AID in DHL tissues and classical diffuse large b-cell lymphoma (DLBCL) tissues were compared. The results suggested that AID is of important value in predicting DHL, stronger CSR of AID was observed in DHL patients, which exhibited AID overexpression and c-Myc gene translocation of DHL after CSR induction. It is concluded that AID directly induces CSR in DHL and may result in c-Myc gene translocation. Targeting AID may be a good treatment regimen for DHL.


Cytidine Deaminase/biosynthesis , Immunoglobulin Class Switching/genetics , Lymphoma, Large B-Cell, Diffuse/enzymology , Molecular Targeted Therapy , Neoplasm Proteins/biosynthesis , Adult , Aged , Aged, 80 and over , Cytidine Deaminase/genetics , Cytidine Deaminase/physiology , Enzyme Induction/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, bcl-2 , Genes, myc , Humans , Immunoglobulin Isotypes/biosynthesis , Immunoglobulin Isotypes/blood , Immunoglobulin Isotypes/genetics , Kaplan-Meier Estimate , Ki-67 Antigen/genetics , Lipopolysaccharides/pharmacology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/mortality , Male , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Prognosis , Proto-Oncogene Proteins c-bcl-6/genetics , Translocation, Genetic , Up-Regulation/drug effects
3.
Front Immunol ; 11: 491, 2020.
Article En | MEDLINE | ID: mdl-32265934

Estrogen contributes to females' strong antibody response to microbial vaccines and proneness to autoimmunity, particularly antibody-mediated systemic autoimmunity, in females. We have hypothesized that this is due to estrogen-mediated potentiation of class switch DNA recombination (CSR) and somatic hypermutation (SHM). As we have shown, estrogen boosts AID expression, which is critical for both CSR and SHM, through upregulation of HoxC4, which together with NF-κB critically mediates Aicda (AID gene) promoter activation. We contend here that additional regulation of Aicda expression by estrogen occurs through epigenetic mechanisms. As we have shown, histone deacetylase inhibitors (HDIs) short-chain fatty acid (SCFA) butyrate and propionate as well as the pharmacologic HDI valproic acid upregulate miRNAs that silence AID expression, thereby modulating specific antibody responses in C57BL/6 mice and autoantibody responses in lupus-prone MRL/Faslpr/lpr mice. Here, using constitutive knockout Esr1-/- mice and B cells as well as conditional knockout Aicdacre/creEsr1flox/flox mice and B cells, we showed that the HDI-mediated downregulation of Aicda expression as well as the maturation of antibody and autoantibody responses is reversed by estrogen and enhanced by deletion of ERα or E2 inhibition. Estrogen's reversion of HDI-mediated inhibition of Aicda and CSR in antibody and autoantibody responses occurred through downregulation of B cell miR-26a, which, as we showed, targets Aicda mRNA 3'UTR. miR-26a was significantly upregulated by HDIs. Accordingly, enforced expression of miR-26a reduced Aicda expression and CSR, while miR-26a-sponges (competitive inhibitors of miR-26a) increased Aicda expression and CSR. Thus, our findings show that estrogen reverses the HDI-mediated downregulation of AID expression and CSR through selective modulation of miR-26a. They also provide mechanistic insights into the immunomodulatory activity of this hormone and a proof-of-principle for using combined ER inhibitor-HDI as a potential therapeutic approach.


Autoantibodies/biosynthesis , B-Lymphocytes/drug effects , Butyrates/pharmacology , Cytidine Deaminase/biosynthesis , Estradiol/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Immunoglobulin Class Switching/drug effects , Isoantibodies/biosynthesis , MicroRNAs/biosynthesis , Propionates/pharmacology , Valproic Acid/pharmacology , 3' Untranslated Regions , Animals , Autoantibodies/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Base Sequence , Binding, Competitive , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , Down-Regulation/drug effects , Estrogen Receptor alpha/deficiency , Female , Gene Expression Regulation/drug effects , Humans , Immunoglobulin Class Switching/genetics , Isoantibodies/immunology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred MRL lpr , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Phosphoprotein Phosphatases/genetics , Phosphoric Monoester Hydrolases/genetics , Promoter Regions, Genetic , Proof of Concept Study , Recombinant Proteins/metabolism , Sequence Alignment , Sex Characteristics , Transduction, Genetic
4.
Gut ; 68(10): 1846-1857, 2019 10.
Article En | MEDLINE | ID: mdl-31154396

OBJECTIVE: APOBEC3B (A3B), a cytidine deaminase acting as a contributor to the APOBEC mutation pattern in many kinds of tumours, is upregulated in patients with hepatocellular carcinoma (HCC). However, APOBEC mutation patterns are absent in HCC. The mechanism of how A3B affects HCC progression remains elusive. DESIGN: A3B -promoter luciferase reporter and other techniques were applied to elucidate mechanisms of A3B upregulation in HCC. A3B overexpression and knockdown cell models, immunocompetent and immune-deficient mouse HCC model were conducted to investigate the influence of A3B on HCC progression. RNA-seq, flow cytometry and other techniques were conducted to analyse how A3B modulated the cytokine to enhance the recruitment of myeloid--derived suppressor cells (MDSCs) and tumour--associated macrophages (TAMs). RESULTS: A3B upregulation through non-classical nuclear factor-κB (NF-κB)signalling promotes HCC growth in immunocompetent mice, associated with an increase of MDSCs, TAMs and programmed cell death1 (PD1) exprssed CD8+ T cells. A CCR2 antagonist suppressed TAMs and MDSCs infiltration and delayed tumour growth in A3B and A3BE68Q/E255Q- expressing mouse tumours. Mechanistically, A3B upregulation in HCC depresses global H3K27me3 abundance via interaction with polycomb repressor complex 2 (PRC2) and reduces an occupancy of H3K27me3 on promoters of the chemokine CCL2 to recruit massive TAMs and MDSCs. CONCLUSION: Our observations uncover a deaminase-independent role of the A3B in modulating the HCC microenvironment and demonstrate a proof for the concept of targeting A3B in HCC immunotherapy.


Carcinoma, Hepatocellular/genetics , Cytidine Deaminase/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms, Experimental/genetics , Minor Histocompatibility Antigens/genetics , Tumor Microenvironment/physiology , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cytidine Deaminase/biosynthesis , DNA, Neoplasm/genetics , Disease Progression , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Minor Histocompatibility Antigens/biosynthesis , Promoter Regions, Genetic
5.
Blood ; 133(19): 2056-2068, 2019 05 09.
Article En | MEDLINE | ID: mdl-30814061

Activation-induced cytidine deaminase (AID) initiates somatic hypermutation and class switch recombination of the immunoglobulin genes. As a trade-off for its physiological function, AID also contributes to tumor development through its mutagenic activity. In chronic lymphocytic leukemia (CLL), AID is overexpressed in the proliferative fractions (PFs) of the malignant B lymphocytes, and its anomalous expression has been associated with a clinical poor outcome. Recent preclinical data suggested that ibrutinib and idelalisib, 2 clinically approved kinase inhibitors, increase AID expression and genomic instability in normal and neoplastic B cells. These results raise concerns about a potential mutagenic risk in patients receiving long-term therapy. To corroborate these findings in the clinical setting, we analyzed AID expression and PFs in a CLL cohort before and during ibrutinib treatment. We found that ibrutinib decreases the CLL PFs and, interestingly, also reduces AID expression, which correlates with dampened AKT and Janus Kinase 1 signaling. Moreover, although ibrutinib increases AID expression in a CLL cell line, it is unable to do so in primary CLL samples. Our results uncover a differential response to ibrutinib between cell lines and the CLL clone and imply that ibrutinib could differ from idelalisib in their potential to induce AID in treated patients. Possible reasons for the discrepancy between preclinical and clinical findings, and their effect on treatment safety, are discussed.


Antineoplastic Agents, Immunological/adverse effects , Cytidine Deaminase/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Pyrazoles/adverse effects , Pyrimidines/adverse effects , Adenine/analogs & derivatives , Aged , Cell Proliferation/drug effects , Cytidine Deaminase/biosynthesis , Down-Regulation , Female , Humans , Male , Middle Aged , Piperidines
6.
N Biotechnol ; 51: 67-79, 2019 Jul 25.
Article En | MEDLINE | ID: mdl-30822538

AID/APOBEC3 enzymes are cytidine deaminases that mutate antibody and retroviral genes and also mediate extensive tumor genome mutagenesis. The study of purified AID/APOBEC3 proteins is challenged by difficulties with their expression and purification arising from genotoxicity in expression hosts, extensive non-specific protein-protein/DNA/RNA interactions and haphazard oligomerization. To date, expression hosts for purification of AID/APOBEC3 enzymes include bacteria, insect and mammalian cells. Here the establishment and optimization of a yeast expression/secretion system for AID/APOBEC3s are reported, followed by comparison with the same enzymes expressed in bacterial and mammalian hosts. AID and APOBEC3G were expressed successfully in Pichia pastoris, each either with an N-terminal GST tag, C-terminal V5-His tag or as untagged native form. It was verified that the yeast-expressed enzymes exhibit identical biochemical properties to those reported using bacterial and mammalian expression, indicating high fidelity of protein folding. It was demonstrated that the system can be adapted for secretion of the enzymes into the media which was used directly in various enzyme assays. The system is also amenable to elimination of bulky fusion tags, providing native untagged enzymes. Thus, P. pastoris is an advantageous expression factory for AID/APOBEC3 enzymes, considering the cost, time, efficiency and quality of the obtained enzymes. The first report is also provided here of a functionally active, untagged, secreted AID, which may become a useful research reagent. A comprehensive comparison is made of the effect of fusion tags and expression hosts on the biochemical actions of AID and APOBEC3G.


APOBEC Deaminases/biosynthesis , APOBEC Deaminases/genetics , Cytidine Deaminase/biosynthesis , Cytidine Deaminase/genetics , Immunity , Neoplasms/enzymology , Pichia/genetics , APOBEC Deaminases/isolation & purification , Cytidine Deaminase/isolation & purification , Humans , Mutagens , Neoplasms/metabolism
7.
mBio ; 10(1)2019 02 05.
Article En | MEDLINE | ID: mdl-30723127

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.


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
8.
Histol Histopathol ; 34(4): 405-417, 2019 Apr.
Article En | MEDLINE | ID: mdl-30289149

APOBEC3 enzymes are part of the innate immune system and they are important in retroviral defense. The number of mutations in ovarian cancer increases with rising levels of APOBEC3B mRNA. We could confirm that APOBEC3B mRNA is upregulated in ovarian cancer cell lines and in ovarian cancer tissue. We evaluated APOBEC3B expression in histologically defined subtypes of ovarian cancer to identify its influence on overall survival (OS) and progression-free survival (PFS). Tissue microarrays from 219 patients with high-grade serous (HGSC), 61 with low-grade serous (LGSC), 62 with endometrioid (EC) and 55 with clear cell (CCC) ovarian carcinoma were stained using an antibody against APOBEC3B. Real-time quantitative PCR was performed to detect APOBEC3B mRNA levels in 274 cases of HGSC, in 11 cases of LGSC, in 47 cases of EC and in 29 cases of CCC. Tumor-infiltrating lymphocytes (TILs) have been evaluated in a previous project. APOBEC3B staining was cytoplasmic as well as nuclear and both were positively correlated (P<0.001). In HGSC a trend was detectable for positive cytoplasmic staining as favorable regarding OS (P=0.283) and PFS (P=0.137). High levels of APOBEC3B mRNA were associated with prolonged PFS in HGSC in univariate analyses (P=0.043) and multivariate analyses (HR 0.55; 95%CI 0.35-0.88; P=0.012). APOBEC3B cytoplasmic staining and APOBEC3B mRNA were positively correlated with TILs. APOBEC3B in HGSC is related to an active immune infiltrate. However, there is no evidence for APOBEC3B as a clinically relevant prognostic biomarker.


Cystadenocarcinoma, Serous/metabolism , Cytidine Deaminase/biosynthesis , Minor Histocompatibility Antigens/biosynthesis , Ovarian Neoplasms/metabolism , Aged , Biomarkers, Tumor/analysis , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/mortality , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/immunology , Middle Aged , Ovarian Neoplasms/immunology , Ovarian Neoplasms/mortality , RNA, Messenger/analysis
9.
Ann Hematol ; 98(2): 423-435, 2019 Feb.
Article En | MEDLINE | ID: mdl-30368590

Activation-induced cytidine deaminase (AID) is a mutator enzyme essential for somatic hypermutation (SHM) and class switch recombination (CSR) during effective adaptive immune responses. Its aberrant expression and activity have been detected in lymphomas, leukemias, and solid tumors. In chronic lymphocytic leukemia (CLL) increased expression of alternatively spliced AID variants has been documented. We used real-time RT-PCR to quantify the expression of AID and its alternatively spliced transcripts (AIDΔE4a, AIDΔE4, AIDivs3, and AIDΔE3E4) in 149 CLL patients and correlated this expression to prognostic markers including recurrent chromosomal aberrations, the presence of complex karyotype, mutation status of the immunoglobulin heavy chain variable gene, and recurrent mutations. We report a previously unappreciated association between higher AID transcript levels and trisomy of chromosome 12. Functional analysis of AID splice variants revealed loss of their activity with respect to SHM, CSR, and induction of double-strand DNA breaks. In silico modeling provided insight into the molecular interactions and structural dynamics of wild-type AID and a shortened AID variant closely resembling AIDΔE4, confirming its loss-of-function phenotype.


Alternative Splicing , Cytidine Deaminase , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Leukemia, Lymphocytic, Chronic, B-Cell , Models, Biological , Neoplasm Proteins , Trisomy , Aged , Animals , Chromosomes, Human, Pair 12/enzymology , Chromosomes, Human, Pair 12/genetics , Computer Simulation , Cytidine Deaminase/biosynthesis , Cytidine Deaminase/chemistry , Cytidine Deaminase/genetics , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Mice , Mice, Knockout , Middle Aged , Molecular Dynamics Simulation , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Trisomy/genetics , Trisomy/pathology
10.
Methods Mol Biol ; 1707: 1-14, 2018.
Article En | MEDLINE | ID: mdl-29388096

Activation-induced cytidine deaminase (AID) initiates somatic hypermutation (SHM) by converting deoxycytidines (dC) to deoxyuracils (dU) which then can induce other mutations, and plays a central role in introducing diversification of the antibody repertoire in B cells. Ectopic expression of AID in bacteria and non-B cells can also lead to frequent mutations in highly expressed genes. Taking advantage of this feature of AID, in recent years, systems coupling in vitro somatic hypermutation and mammalian cell surface display have been developed, with unique benefits in antibody discovery and optimization in vitro. Here, we provide a protocol for AID mediated in vitro protein evolution. A CHO cell clone bearing a single gene expression cassette has been constructed. The gene of an interested protein for in vitro evolution can be easily inserted into the cassette by dual recombinase-mediated cassette exchange (RMCE) and constantly expressed at high levels. Here, we matured an anti-TNFα antibody as an example. Firstly, we obtained a CHO cell clone highly displaying the antibody by dual RMCE. Then, the plasmid expressing AID is transfected into the CHO cells. After a few rounds of cell sorting-cell proliferation, mutant antibodies with improved features can be generated. This protocol can be applied for improving protein features based on displaying levels on cell surface and protein-protein interaction, and thus is able to enhance affinity, specificity, and stability besides others.


Antibodies, Monoclonal , Cytidine Deaminase , Directed Molecular Evolution/methods , Transfection , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , CHO Cells , Cricetinae , Cricetulus , Cytidine Deaminase/biosynthesis , Cytidine Deaminase/genetics , Humans
11.
Mem Inst Oswaldo Cruz ; 112(11): 785-789, 2017 Nov.
Article En | MEDLINE | ID: mdl-29091140

Cytidine deaminase (MtCDA), encoded by cdd gene (Rv3315c), is the only enzyme identified in nucleotide biosynthesis pathway of Mycobacterium tuberculosis that is able to recycle cytidine and deoxycytidine. An M. tuberculosis knockout strain for cdd gene was obtained by allelic replacement. Evaluation of mRNA expression validated cdd deletion and showed the absence of polar effect. MudPIT LC-MS/MS data indicated thymidine phosphorylase expression was decreased in knockout and complemented strains. The cdd disruption does not affect M. tuberculosis growth both in Mid- dlebrook 7H9 and in RAW 264.7 cells, which indicates that cdd is not important for macrophage invasion and virulence.


Cytidine Deaminase/genetics , Deoxycytidine/genetics , Macrophages/microbiology , Mycobacterium tuberculosis/pathogenicity , Cytidine Deaminase/biosynthesis , Deoxycytidine/biosynthesis , Gene Knockout Techniques , Humans , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/growth & development , Time Factors
12.
Mem. Inst. Oswaldo Cruz ; 112(11): 785-789, Nov. 2017. graf
Article En | LILACS | ID: biblio-1040564

Cytidine deaminase (MtCDA), encoded by cdd gene (Rv3315c), is the only enzyme identified in nucleotide biosynthesis pathway of Mycobacterium tuberculosis that is able to recycle cytidine and deoxycytidine. An M. tuberculosis knockout strain for cdd gene was obtained by allelic replacement. Evaluation of mRNA expression validated cdd deletion and showed the absence of polar effect. MudPIT LC-MS/MS data indicated thymidine phosphorylase expression was decreased in knockout and complemented strains. The cdd disruption does not affect M. tuberculosis growth both in Mid- dlebrook 7H9 and in RAW 264.7 cells, which indicates that cdd is not important for macrophage invasion and virulence.


Humans , Cytidine Deaminase/genetics , Deoxycytidine/genetics , Macrophages/microbiology , Mycobacterium tuberculosis/pathogenicity , Time Factors , Cytidine Deaminase/biosynthesis , Deoxycytidine/biosynthesis , Gene Knockout Techniques , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/enzymology
13.
Sci Rep ; 7(1): 1659, 2017 05 10.
Article En | MEDLINE | ID: mdl-28490810

B cell derived induced pluripotent stem cells (BiPSCs) were recently established from peripheral blood B cells by the simultaneous transfection of Yamanaka factors (Oct3/4, Sox2, Klf4, c-Myc) and C/EBPα using a Sendai virus vector. Here, using a different method, we established BiPSCs with immunoglobulin heavy chain (IgH) gene rearrangement from normal B cells purified from lymph nodes. The critical points of our method are pre-stimulation of B cells with IL-21 and CD40-ligand (CD40L), followed by consecutive transfection of highly concentrated Yamanaka factors using a retroviral vector. Following each transfection the cells were centrifuged onto a retronectin coated plate and the activated by IL-4, IL-2, and CD40L. Furthermore, we established BiPSCs (BiPSC-A) in which activation-induced cytidine deaminase (AID) could be induced using the doxycycline-controlled. Both the parental BiPSC and BiPSC-A showed the capability of differentiating into hematopoietic progenitor cells (HPCs) based on confirmation of CD34 expression and colony-formation from CD34-positive cells. The findings that BiPSC-A can differentiate into HPCs suggest that there is a possibility that induction of AID expression would result in chromosomal translocations in the process of differentiation from BiPSCs, and therefore that these BiPSCs could be useful in elucidating the tumor origin of abnormal B cells in myelomagenesis.


B-Lymphocytes/cytology , Cell Differentiation , Cytidine Deaminase/biosynthesis , Hematopoietic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Antigens, CD19/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Differentiation/drug effects , Colony-Forming Units Assay , Doxycycline/pharmacology , Enzyme Induction/drug effects , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Lymph Nodes/cytology , Mice, SCID , Models, Biological
14.
Sci Rep ; 7: 44089, 2017 03 09.
Article En | MEDLINE | ID: mdl-28276478

The key signature of cancer genomes is the accumulation of DNA mutations, the most abundant of which is the cytosine-to-thymine (C-to-T) transition that results from cytosine deamination. Analysis of The Cancer Genome Atlas (TCGA) database has demonstrated that this transition is caused mainly by upregulation of the cytosine deaminase APOBEC3B (A3B), but the mechanism has not been completely characterized. We found that B-Myb (encoded by MYBL2) binds the A3B promoter, causing transactivation, and this is responsible for the C-to-T transitions and DNA hypermutation in breast cancer cells. Analysis of TCGA database yielded similar results, supporting that MYBL2 and A3B are upregulated and putatively promote C-to-T transitions in multiple cancer types. Moreover, blockade of EGF receptor with afatinib attenuated B-Myb-A3B signaling, suggesting a clinically relevant means of suppressing mutagenesis. Our results suggest that B-Myb-A3B contributes to DNA damage and could be targeted by inhibiting EGF receptor.


Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Cytidine Deaminase/biosynthesis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Minor Histocompatibility Antigens/biosynthesis , Mutation , Neoplasm Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cytidine Deaminase/genetics , Female , Humans , MCF-7 Cells , Minor Histocompatibility Antigens/genetics , Neoplasm Proteins/genetics , Trans-Activators/genetics
15.
Gynecol Oncol ; 145(3): 577-583, 2017 06.
Article En | MEDLINE | ID: mdl-28215840

OBJECTIVE: Ovarian carcinomas that originate from fallopian epithelial cells are suggested to arise due to repeated exposure to ovulatory follicular fluid (FF). Mechanistic explanation(s) for how this occurs are unknown. Here, we sought to understand if FF exposure to fallopian epithelial cells could induce DNA damage and expression of a known family of DNA mutators, apolipoprotein B mRNA editing enzyme, catalytic polypeptide (APOBEC) cytidine deaminases. METHODS: Follicular fluid and matched patient plasma samples were obtained from donors. Fallopian epithelial cells (FT33-TAg, FT189, FT190, and FT194) were cultured with FF or plasma for 24h, and cell proliferation and DNA damage were assessed. Effects of FF on Apobec gene expression were determined by qRT-PCR and western blot analyses. Fallopian epithelial cells were transfected with an APOBEC3A expression vector and DNA damage was assessed. RESULTS: Follicular fluid exposure increased epithelial cell proliferation as measured by three independent methods, and DNA damage accumulation as assessed using three independent measures. This effect was specific to FF, as matched patient plasma did not have the same effects. Increased expression of Apobec3a was observed in fallopian epithelial cells following exposure to 5 of 8 patient FF samples, and transient overexpression of APOBEC3A was sufficient to induce double strand DNA breaks. CONCLUSIONS: Follicular fluid can induce cell proliferation and DNA damage accumulation in cultured fallopian epithelial cells. Increased expression of APOBEC3A, a known DNA mutator, may explain the high incidence of DNA damage after FF exposure. The role of Apobec3a in ovulation-induced inflammation warrants further investigation.


Cytidine Deaminase/biosynthesis , Epithelial Cells/enzymology , Fallopian Tubes/enzymology , Follicular Fluid/physiology , Adult , Cell Proliferation/physiology , Cytidine Deaminase/genetics , DNA Breaks, Double-Stranded , Enzyme Induction , Epithelial Cells/pathology , Fallopian Tubes/pathology , Female , Gene Expression , Humans , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Young Adult
16.
J Virol ; 91(6)2017 03 15.
Article En | MEDLINE | ID: mdl-28077648

The cytidine deaminase APOBEC3B (A3B) underlies the genetic heterogeneity of several human cancers, including cervical cancer, which is caused by human papillomavirus (HPV) infection. We previously identified a region within the A3B promoter that is activated by the viral protein HPV16 E6 in human keratinocytes. Here, we discovered three sites recognized by the TEAD family of transcription factors within this region of the A3B promoter. Reporter assays in HEK293 cells showed that exogenously expressed TEAD4 induced A3B promoter activation through binding to these sites. Normal immortalized human keratinocytes expressing E6 (NIKS-E6) displayed increased levels of TEAD1/4 protein compared to parental NIKS. A series of E6 mutants revealed that E6-mediated degradation of p53 was important for increasing TEAD4 levels. Knockdown of TEADs in NIKS-E6 significantly reduced A3B mRNA levels, whereas ectopic expression of TEAD4 in NIKS increased A3B mRNA levels. Finally, chromatin immunoprecipitation assays demonstrated increased levels of TEAD4 binding to the A3B promoter in NIKS-E6 compared to NIKS. Collectively, these results indicate that E6 induces upregulation of A3B through increased levels of TEADs, highlighting the importance of the TEAD-A3B axis in carcinogenesis.IMPORTANCE The expression of APOBEC3B (A3B), a cellular DNA cytidine deaminase, is upregulated in various human cancers and leaves characteristic, signature mutations in cancer genomes, suggesting that it plays a prominent role in carcinogenesis. Viral oncoproteins encoded by human papillomavirus (HPV) and polyomavirus have been reported to induce A3B expression, implying the involvement of A3B upregulation in virus-associated carcinogenesis. However, the molecular mechanisms causing A3B upregulation remain unclear. Here, we demonstrate that exogenous expression of the cellular transcription factor TEAD activates the A3B promoter. Further, the HPV oncoprotein E6 increases the levels of endogenous TEAD1/4 protein, thereby leading to A3B upregulation. Since increased levels of TEAD4 are frequently observed in many cancers, an understanding of the direct link between TEAD and A3B upregulation is of broad oncological interest.


Cytidine Deaminase/biosynthesis , DNA-Binding Proteins/metabolism , Host-Pathogen Interactions , Human papillomavirus 16/physiology , Minor Histocompatibility Antigens/biosynthesis , Muscle Proteins/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Up-Regulation , Cell Line , Chromatin Immunoprecipitation , Epithelial Cells/virology , Gene Expression , Gene Knockdown Techniques , Humans , Proteolysis , TEA Domain Transcription Factors , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
17.
Nucleic Acids Res ; 45(6): 3231-3241, 2017 04 07.
Article En | MEDLINE | ID: mdl-28100701

Foreign and self-cytoplasmic DNA are recognized by numerous DNA sensor molecules leading to the production of type I interferons. Such DNA agonists should be degraded otherwise cells would be chronically stressed. Most human APOBEC3 cytidine deaminases can initiate catabolism of cytoplasmic mitochondrial DNA. Using the human myeloid cell line THP-1 with an interferon inducible APOBEC3A gene, we show that cytoplasmic DNA triggers interferon α and ß production through the RNA polymerase III transcription/RIG-I pathway leading to massive upregulation of APOBEC3A. By catalyzing C→U editing in single stranded DNA fragments, the enzyme prevents them from re-annealing so attenuating the danger signal. The price to pay is chromosomal DNA damage in the form of CG→TA mutations and double stranded DNA breaks which, in the context of chronic inflammation, could drive cells down the path toward cancer.


Cytidine Deaminase/biosynthesis , DNA Breaks, Double-Stranded , DNA, Mitochondrial/metabolism , Cell Line, Tumor , Chromosomes, Human , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Cytosol/metabolism , DEAD Box Protein 58 , DNA, Mitochondrial/chemistry , Humans , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Interferon-beta/physiology , Proteins/genetics , Proteins/metabolism , RNA Polymerase III/metabolism , Receptors, Immunologic , Transcription, Genetic , Up-Regulation , Uracil/metabolism
18.
J Leukoc Biol ; 100(5): 1147-1153, 2016 11.
Article En | MEDLINE | ID: mdl-27365533

M1/M2 cytokine-dependent polarization of primary human MDMs has been shown to contain CCR5-dependent (R5) HIV-1 replication. In this study, a similar effect was achieved when monocytes were first polarized toward M1 or M2 and were infected 7 d after their differentiation into MDMs, regardless of whether the cytokines were removed 18 h after cell stimulation or were left in culture. Unlike polarized MDMs, no significant down-regulation of CD4 from the cell surface was observed in MDMs derived from M1/M2-polarized monocytes. A second stimulation of MDMs differentiated from M1/M2 monocytes with the opposite polarizing cytokines converted the virus replication profile according to the new stimuli. The expression of M1 and M2 markers (i.e., APOBEC3A and DC-SIGN, respectively) was induced by MDM stimulation with the opposite cytokines, although it also persisted in cells according to their first stimulatory condition. Thus, stimulation of monocytes with M1- and M2-inducing cytokines leads to a restriction of HIV-1 replication when these cells are infected several days later as differentiated MDMs. These observations imply that activation of circulating monocytes significantly influences their capacity to either support or restrict HIV-1 replication, once extravasated, and eventually to become infected as tissue macrophages.


HIV-1/physiology , Macrophages/virology , Monocytes/cytology , Virus Replication , Antigens, Differentiation/biosynthesis , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cell Differentiation , Cytidine Deaminase/biosynthesis , Cytidine Deaminase/genetics , Humans , Interferon-gamma/pharmacology , Interleukin-4/pharmacology , Lectins, C-Type/biosynthesis , Lectins, C-Type/genetics , Macrophages/classification , Proteins/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Recombinant Proteins/pharmacology , Time Factors , Tumor Necrosis Factor-alpha/pharmacology
19.
Neoplasia ; 18(2): 90-9, 2016 Feb.
Article En | MEDLINE | ID: mdl-26936395

In recent years, the notion that ovarian carcinoma results from ovulation-induced inflammation of the fallopian tube epithelial cells (FTECs) has gained evidence. However, the mechanistic pathway for this process has not been revealed yet. In the current study, we propose the mutator protein activation-induced cytidine deaminase (AID) as a link between ovulation-induced inflammation in FTECs and genotoxic damage leading to ovarian carcinogenesis. We show that AID, previously shown to be functional only in B lymphocytes, is expressed in FTECs under physiological conditions, and is induced in vitro upon ovulatory-like stimulation and in vivo in carcinoma-associated FTECs. We also report that AID activity results in epigenetic, genetic and genomic damage in FTECs. Overall, our data provides new insights into the etiology of ovarian carcinogenesis and may set the ground for innovative approaches aimed at prevention and early detection.


Carcinogenesis/genetics , Cytidine Deaminase/biosynthesis , Inflammation/genetics , Ovarian Neoplasms/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cytidine Deaminase/genetics , DNA Damage/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fallopian Tubes/metabolism , Fallopian Tubes/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Inflammation/complications , Inflammation/pathology , Ovarian Neoplasms/complications , Ovarian Neoplasms/pathology , Ovulation/genetics , Ovulation/metabolism
20.
Med Oncol ; 33(3): 26, 2016 Mar.
Article En | MEDLINE | ID: mdl-26880326

APOBEC3B belongs to the cytidine deaminase apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like (APOBEC3) family of enzymes and induces C to T transitions of target DNA by cytidine deamination. Recently, several mutations in various cancers have been linked to APOBEC3B, suggesting a crucial role for this protein in carcinogenesis and cancer development. However, the significance of APOBEC3B in esophageal squamous cell carcinoma (ESCC) remains uncertain. In addition, the APOBEC3B immunoreactivity in cancer tissues is uncertain. Recently, we have demonstrated that PIK3CA mutation and the methylation level of long interspersed nucleotide element 1 (LINE-1) (a surrogate marker of global DNA methylation level) are prognostic markers and have crucial role on malignancy in ESCC patients. This study aims to clarify the impact of APOBEC3B on the clinical, pathological, and molecular features of ESCC. We evaluated APOBEC3B expression in ESCC and investigated the relationships among the immunoreactivity of APOBEC3B, clinical and pathological features, and the molecular features of ESCC (PIK3CA mutation, p53 expression, and LINE-1 methylation level). The immunoreactivity and mRNA level of APOBEC3B were significantly higher in cancer tissues than in noncancerous esophageal mucosae (P = 0.050). APOBEC3B expression was significantly correlated with PIK3CA mutation (P = 0.013), particularly with C to T transitions of PIK3CA (P = 0.041). Moreover, a high expression of APOBEC3B was significantly associated with LINE-1 hypomethylation (P = 0.027). Given the crucial roles of PIK3CA mutation and LINE-1 methylation levels, our findings might provide new insights into the biological mechanisms of ESCC tumorigenesis and progression.


Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Cytidine Deaminase/biosynthesis , Cytidine Deaminase/genetics , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/genetics , Aged , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/mortality , Class I Phosphatidylinositol 3-Kinases , Esophageal Neoplasms/mortality , Esophageal Squamous Cell Carcinoma , Female , Humans , Male , Middle Aged , Minor Histocompatibility Antigens , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Point Mutation/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Survival Rate/trends
...