Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.817
Filtrar
1.
Nat Commun ; 15(1): 8603, 2024 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-39366934

RESUMO

Metastatic urothelial carcinoma (mUC) is a lethal cancer, with limited therapeutic options. Large-scale studies in early settings provided critical insights into the genomic and transcriptomic characteristics of non-metastatic UC. The genomic landscape of mUC remains however unclear. Using Whole Exome (WES) and mRNA sequencing (RNA-seq) performed on metastatic biopsies from 111 patients, we show that driver genomic alterations from mUC were comparable to primary UC (TCGA data). APOBEC, platin, and HRD mutational signatures are the most prevalent in mUC, identified in 56%, 14%, and 9% of mUC samples, respectively. Molecular subtyping using consensus transcriptomic classification in mUC shows enrichment in neuroendocrine subtype. Paired samples analysis reveals subtype heterogeneity and temporal evolution. We identify potential therapeutic targets in 73% of mUC patients, of which FGFR3 (26%), ERBB2 (7%), TSC1 (7%), and PIK3CA (13%) are the most common. NECTIN4 and TACSTD2 are highly expressed regardless of molecular subtypes, FGFR3 alterations and sites of metastases.


Assuntos
Sequenciamento do Exoma , Mutação , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos , Transcriptoma , Humanos , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Masculino , Feminino , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia , Nectinas/genética , Nectinas/metabolismo , Idoso , Proteína 1 do Complexo Esclerose Tuberosa/genética , Proteína 1 do Complexo Esclerose Tuberosa/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Genômica , Pessoa de Meia-Idade , Desaminases APOBEC/genética , Desaminases APOBEC/metabolismo , Urotélio/patologia , Urotélio/metabolismo , Regulação Neoplásica da Expressão Gênica , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Metástase Neoplásica/genética , Idoso de 80 Anos ou mais , Carcinoma de Células de Transição/genética , Carcinoma de Células de Transição/patologia , Neoplasias Urológicas/genética , Neoplasias Urológicas/patologia , Perfilação da Expressão Gênica/métodos
2.
Methods Enzymol ; 705: 311-345, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39389668

RESUMO

In recent years, the connection between APOBEC3 cytosine deaminases and cancer mutagenesis has become ever more apparent. This growing awareness and lack of inhibitory drugs has created a distinct need for biochemical tools that can be used to identify and characterize potential inhibitors of this family of enzymes. In response to this challenge, we have developed a Real-time APOBEC3-mediated DNA Deamination (RADD) assay. The RADD assay provides a rapid, real-time fluorescence readout of APOBEC3 DNA deamination and serves as a crucial addition to the existing APOBEC3 biochemical and cellular toolkit. This method improves upon contemporary DNA deamination assays by offering a more rapid and quantifiable readout as well as providing a platform that is readily adaptable to a high-throughput format for inhibitor discovery. In this chapter we provide a detailed guide for the usage of the RADD assay for the characterization of APOBEC3 enzymes and potential inhibitors.


Assuntos
DNA , Transferência Ressonante de Energia de Fluorescência , Humanos , Transferência Ressonante de Energia de Fluorescência/métodos , DNA/metabolismo , Desaminação , Citidina Desaminase/metabolismo , Citidina Desaminase/genética , Ensaios Enzimáticos/métodos , Inibidores Enzimáticos/farmacologia , Desaminases APOBEC/metabolismo
3.
J Med Virol ; 96(10): e70002, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39400339

RESUMO

High-risk human papillomavirus (HPV) infections are responsible for cervical cancer. However, little is known about the differences between HPV types and risk categories regarding their genetic diversity and particularly APOBEC3-induced mutations - which contribute to the innate immune response to HPV. Using a capture-based next-generation sequencing, 156 HPV whole genome sequences covering 43 HPV types were generated from paired cervical and anal swabs of 30 Togolese female sex workers (FSWs) sampled in 2017. Genetic diversity and APOBEC3-induced mutations were assessed at the viral whole genome and gene levels. Thirty-four pairwise sequence comparisons covering 24 HPV types in cervical and anal swabs revealed identical infections in the two anatomical sites. Differences in genetic diversity among HPV types was observed between patients. The E6 gene was significantly less conserved in low-risk HPVs (lrHPVs) compared to high-risk HPVs (hrHPVs) (p = 0.009). APOBEC3-induced mutations were found to be more common in lrHPVs than in hrHPVs (p = 0.005), supported by our data and by using large HPV sequence collections from the GenBank database. Focusing on the most common lrHPVs 6 and 11 and hrHPVs 16 and 18, APOBEC3-induced mutations were predominantly found in the E4 and E6 genes in lrHPVs, but were almost absent in these genes in hrHPVs. The variable APOBEC3 mutational signatures could contribute to the different oncogenic potentials between HPVs. Further studies are needed to conclusively determine whether APOBEC3 editing levels are associated to the carcinogenic potential of HPVs at the type and sublineage scales.


Assuntos
Desaminases APOBEC , Variação Genética , Genoma Viral , Mutação , Infecções por Papillomavirus , Sequenciamento Completo do Genoma , Humanos , Feminino , Infecções por Papillomavirus/virologia , Infecções por Papillomavirus/genética , Desaminases APOBEC/genética , Genoma Viral/genética , Adulto , Papillomaviridae/genética , Papillomaviridae/classificação , Profissionais do Sexo , Colo do Útero/virologia , Adulto Jovem , Canal Anal/virologia , Sequenciamento de Nucleotídeos em Larga Escala , Citidina Desaminase/genética
4.
Nat Commun ; 15(1): 8140, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39289397

RESUMO

Continuous directed evolution of base editors (BEs) has been successful in bacteria cells, but not yet in mammalian cells. Here, we report the development of a Continuous Directed Evolution system in Mammalian cells (CDEM). CDEM enables the BE evolution in a full-length manner with Cas9 nickase. We harness CDEM to evolve the deaminases of cytosine base editor BE3 and adenine base editors, ABEmax and ABE8e. The evolved cytidine deaminase variants on BE4 architecture show not only narrowed editing windows, but also higher editing purity and low off-target activity without a trade-off in on-targeting activity. The evolved ABEmax and ABE8e variants exhibit narrowed or shifted editing windows to different extents, and lower off-target effects. The results illustrate that CDEM is a simple but powerful approach to continuously evolve BEs without size restriction in the mammalian environment, which is advantageous over continuous directed evolution system in bacteria cells.


Assuntos
Adenina , Sistemas CRISPR-Cas , Citidina Desaminase , Citidina , Evolução Molecular Direcionada , Edição de Genes , Edição de Genes/métodos , Humanos , Adenina/metabolismo , Evolução Molecular Direcionada/métodos , Citidina/metabolismo , Citidina/genética , Citidina Desaminase/metabolismo , Citidina Desaminase/genética , Células HEK293 , Proteína 9 Associada à CRISPR/metabolismo , Proteína 9 Associada à CRISPR/genética , Animais
5.
J Cancer Res Clin Oncol ; 150(9): 426, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39299959

RESUMO

BACKGROUND: Double-hit lymphoma (DHL) with c-MYC gene translocation is highly aggressive and has a poor prognosis. In DHL cells, activation-induced cytidine deaminase (AID) promotes antibody class switch recombination (CSR), ultimately leading to c-MYC gene translocation caused by Myc/IgH DNA double-strand breaks. However, currently there is still no method to suppress the expression of AID. METHODS: In this study, we compared the clinical significance of AID expression in DHL, Additionally, two human double-hit lymphoma cell lines were used to analyze the effect of imatinib mesylate on c-MYC in vitro, and the therapeutic effect was also evaluated in xenograft mouse models. RESULTS: Imatinib mesylate downregulated the AID and c-MYC proteins in patients with chronic myelogenous leukemia associated with DHL. In addition, imatinib mesylate reduced AID and c-MYC expression in SU-DHL-4 and OCI-Ly18 DHL cells. Imatinib mesylate exerted significant inhibitory effects on the proliferation and metastasis of SU-DHL-4 and OCI-Ly18 cells. Finally, imatinib mesylate reduced not only tumor burden in DHL mouse models, but also AID and c-MYC expression in vivo. CONCLUSION: These findings reveal that imatinib mesylate effectively reduces the carcinogenic function of c-MYC in DHL, providing novel strategies for developing therapies targeting c-MYC-driven DHL.


Assuntos
Citidina Desaminase , Mesilato de Imatinib , Proteínas Proto-Oncogênicas c-myc , Ensaios Antitumorais Modelo de Xenoenxerto , Mesilato de Imatinib/farmacologia , Animais , Humanos , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Linhagem Celular Tumoral , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Feminino , Antineoplásicos/farmacologia , Translocação Genética , Masculino , Proliferação de Células/efeitos dos fármacos , Linfoma/tratamento farmacológico , Linfoma/patologia , Linfoma/genética , Linfoma/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos
6.
Nucleic Acids Res ; 52(18): 11188-11202, 2024 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-39271120

RESUMO

Cytidine base editors (CBEs) hold significant potential in genetic disease treatment and in breeding superior traits into animals. However, their large protein sizes limit their delivery by adeno-associated virus (AAV), given its packing capacity of <4.7 kb. To overcome this, we employed a web-based fast generic discovery (WFG) strategy, identifying several small ssDNA deaminases (Sdds) and constructing multiple Sdd-CBE 1.0 versions. SflSdd-CBE 1.0 demonstrated high C-to-T editing efficiency, comparable to AncBE4max, while SviSdd-CBE 1.0 exhibited moderate C-to-T editing efficiency with a narrow editing window (C3 to C5). Utilizing AlphaFold2, we devised a one-step miniaturization strategy, reducing the size of Sdds while preserving their efficiency. Notably, we administered AAV8 expressing PCSK9 targeted sgRNA and SflSdd-CBEs (nSaCas9) 2.0 into mice, leading to gene-editing events (with editing efficiency up to 15%) and reduced serum cholesterol levels, underscoring the potential of Sdds in gene therapy. These findings offer new single-stranded editing tools for the treatment of rare genetic diseases.


Assuntos
Citidina Desaminase , Dependovirus , Edição de Genes , Animais , Edição de Genes/métodos , Dependovirus/genética , Camundongos , Humanos , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , DNA de Cadeia Simples/metabolismo , DNA de Cadeia Simples/genética , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/metabolismo , Células HEK293 , Terapia Genética/métodos , Sistemas CRISPR-Cas , Camundongos Endogâmicos C57BL , RNA Guia de Sistemas CRISPR-Cas/genética
7.
Theranostics ; 14(15): 6036-6052, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39346550

RESUMO

Background: Clinically, the persistence of HBV cccDNA is the major obstacle in anti-HBV therapy. However, the underlying mechanism of HBV cccDNA is poorly understood. The transcriptional factor STAT3 is able to activate HBV replication in liver. Approach & Results: RNA-Seq analysis demonstrated that cucurbitacin I targeting STAT3 was associated with virus replication in liver. HBV-infected human liver chimeric mouse model and HBV hydrodynamic injection mouse model were established. Then, we validated that cucurbitacin I effectively limited the stability of HBV cccDNA and HBV replication in cells, in which cucurbitacin I enhanced the sensitivity of pegylated interferon α (PEG-IFN α) against HBV via combination in vitro and in vivo. Mechanistically, we identified that cucurbitacin I increased the levels of APOBEC3B to control HBV cccDNA by inhibiting p-STAT3 in cells, resulting in the inhibition of HBV replication. Moreover, RNA-Seq data showed that E3 ubiquitin ligase DTX4 might be involved in the events. Then, we observed that HBV particles could upregulate DTX4 by increasing the levels of p-STAT3 in vitro and in vivo. The p-STAT3-elevated DTX4/male-specific lethal 2 (MSL2) independently and synergistically enhanced the stability of HBV cccDNA by facilitating the ubiquitination degradation of APOBEC3B in cells, leading to the HBV replication. Conclusions: p-STAT3-elevated DTX4 confers the stability of HBV cccDNA and HBV replication by facilitating the ubiquitination degradation of APOBEC3B. Cucurbitacin Ⅰ effectively enhances the sensitivity of PEG-IFN α in anti-HBV therapy by inhibiting the p-STAT3/DTX4/MSL2/APOBEC3B signalling. Our finding provides new insights into the mechanism of HBV cccDNA. The p-STAT3 and DTX4/MSL2 might serve as the therapeutical targets of HBV cccDNA.


Assuntos
Citidina Desaminase , Vírus da Hepatite B , Fígado , Antígenos de Histocompatibilidade Menor , Fator de Transcrição STAT3 , Ubiquitina-Proteína Ligases , Replicação Viral , Fator de Transcrição STAT3/metabolismo , Humanos , Animais , Vírus da Hepatite B/efeitos dos fármacos , Camundongos , Replicação Viral/efeitos dos fármacos , Antígenos de Histocompatibilidade Menor/metabolismo , Antígenos de Histocompatibilidade Menor/genética , Fígado/virologia , Fígado/metabolismo , Citidina Desaminase/metabolismo , Citidina Desaminase/genética , Masculino , Ubiquitina-Proteína Ligases/metabolismo , DNA Circular/metabolismo , DNA Circular/genética , Ubiquitinação/efeitos dos fármacos , DNA Viral/metabolismo , DNA Viral/genética , Hepatite B/metabolismo , Hepatite B/virologia , Hepatite B/tratamento farmacológico , Antivirais/farmacologia , Triterpenos/farmacologia , Triterpenos/metabolismo , Células Hep G2 , Modelos Animais de Doenças , Interferon-alfa/metabolismo , Interferon-alfa/farmacologia
8.
Commun Biol ; 7(1): 1065, 2024 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-39215188

RESUMO

Cytidine deaminase (CDA) converts cytidine and deoxycytidine into uridine and deoxyuridine as part of the pyrimidine salvage pathway. Elevated levels of CDA are found in pancreatic tumors and associated with chemoresistance. Recent evidence suggests that CDA has additional functions in cancer cell biology. In this work, we uncover a novel role of CDA in pancreatic cancer cell metabolism. CDA silencing impairs mitochondrial metabolite production, respiration, and ATP production in pancreatic cancer cells, leading to a so-called Pasteur effect metabolic shift towards glycolysis. Conversely, we find that CDA expression promotes mitochondrial biogenesis and oxidative phosphorylation, independently of CDA deaminase activity. Furthermore, we observe that patient primary cells overexpressing CDA are more sensitive to mitochondria-targeting drugs. Collectively, this work shows that CDA plays a non-canonical role in pancreatic cancer biology by promoting mitochondrial function, which could be translated into novel therapeutic vulnerabilities.


Assuntos
Citidina Desaminase , Mitocôndrias , Biogênese de Organelas , Neoplasias Pancreáticas , Humanos , Citidina Desaminase/metabolismo , Citidina Desaminase/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Mitocôndrias/metabolismo , Linhagem Celular Tumoral , Fosforilação Oxidativa , Glicólise
9.
Immunol Lett ; 269: 106909, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39128629

RESUMO

Activation-induced cytidine deaminase (AID) is responsible for the initiation of somatic hypermutation (SHM) and class-switch recombination (CSR), which result in antibody affinity maturation and isotype switching, thus producing pathogen-specific antibodies. Chromatin dynamics and accessibility play a significant role in determining AID expression and its targeting. Chromatin remodelers contribute to the accessibility of the chromatin structure, thereby influencing the targeting of AID to Ig genes. Epigenetic modifications, including DNA methylation, histone modifications, and miRNA expression, profoundly impact the regulation of AID and chromatin remodelers targeting Ig genes. Additionally, epigenetic modifications lead to chromatin rearrangement and thereby can change AID expression levels and its preferential targeting to Ig genes. This interplay is symbolized as the ACE phenomenon encapsulates three interconnected aspects: AID, Chromatin remodelers, and Epigenetic modifications. This review emphasizes the importance of understanding the intricate relationship between these aspects to unlock the therapeutic potential of these molecular processes and molecules.


Assuntos
Diversidade de Anticorpos , Montagem e Desmontagem da Cromatina , Citidina Desaminase , Epigênese Genética , Citidina Desaminase/metabolismo , Citidina Desaminase/genética , Humanos , Animais , Diversidade de Anticorpos/genética , Switching de Imunoglobulina/genética , Metilação de DNA , Hipermutação Somática de Imunoglobulina , Cromatina/metabolismo , Cromatina/genética , Regulação da Expressão Gênica
10.
PLoS Pathog ; 20(8): e1012505, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39208378

RESUMO

Replication of the complex retrovirus mouse mammary tumor virus (MMTV) is antagonized by murine Apobec3 (mA3), a member of the Apobec family of cytidine deaminases. We have shown that MMTV-encoded Rem protein inhibits proviral mutagenesis by the Apobec enzyme, activation-induced cytidine deaminase (AID) during viral replication in BALB/c mice. To further study the role of Rem in vivo, we have infected C57BL/6 (B6) mice with a superantigen-independent lymphomagenic strain of MMTV (TBLV-WT) or a mutant strain that is defective in Rem and its cleavage product Rem-CT (TBLV-SD). Compared to BALB/c, B6 mice were more susceptible to TBLV infection and tumorigenesis. Furthermore, unlike MMTV, TBLV induced T-cell tumors in B6 µMT mice, which lack membrane-bound IgM and conventional B-2 cells. At limiting viral doses, loss of Rem expression in TBLV-SD-infected B6 mice accelerated tumorigenesis compared to TBLV-WT in either wild-type B6 or AID-knockout mice. Unlike BALB/c results, high-throughput sequencing indicated that proviral G-to-A or C-to-T mutations were unchanged regardless of Rem expression in B6 tumors. However, knockout of both AID and mA3 reduced G-to-A mutations. Ex vivo stimulation showed higher levels of mA3 relative to AID in B6 compared to BALB/c splenocytes, and effects of agonists differed in the two strains. RNA-Seq revealed increased transcripts related to growth factor and cytokine signaling in TBLV-SD-induced tumors relative to TBLV-WT-induced tumors, consistent with another Rem function. Thus, Rem-mediated effects on tumorigenesis in B6 mice are independent of Apobec-mediated proviral hypermutation.


Assuntos
Citidina Desaminase , Vírus do Tumor Mamário do Camundongo , Infecções por Retroviridae , Animais , Feminino , Camundongos , Desaminases APOBEC/genética , Desaminases APOBEC/metabolismo , Carcinogênese/genética , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Vírus do Tumor Mamário do Camundongo/genética , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Infecções por Retroviridae/genética , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/virologia , Infecções Tumorais por Vírus/imunologia , Replicação Viral
11.
Sci Rep ; 14(1): 18206, 2024 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-39107509

RESUMO

The combination of cisplatin and pemetrexed remains the gold standard chemotherapy for malignant pleural mesothelioma (MPM), although resistance and poor response pose a significant challenge. Cytidine deaminase (CDA) is a key enzyme in the nucleotide salvage pathway and is involved in the adaptive stress response to chemotherapy. The cytidine analog capecitabine and its metabolite 5'-deoxy-5-fluorocytidine (5'-DFCR) are converted via CDA to 5-fluorouracil, which affects DNA and RNA metabolism. This study investigated a schedule-dependent treatment strategy, proposing that initial chemotherapy induces CDA expression, sensitizing cells to subsequent capecitabine treatment. Basal CDA protein expression was low in different mesothelioma cell lines but increased in the corresponding xenografts. Standard chemotherapy increased CDA protein levels in MPM cells in vitro and in vivo in a schedule-dependent manner. This was associated with epithelial-to-mesenchymal transition and with HIF-1alpha expression at the transcriptional level. In addition, pretreatment with cisplatin and pemetrexed in combination sensitized MPM xenografts to capecitabine. Analysis of a tissue microarray (TMA) consisting of samples from 98 human MPM patients revealed that most human MPM samples had negative CDA expression. While survival curves based on CDA expression in matched samples clearly separated, significance was not reached due to the limited sample size. In non-matched samples, CDA expression before but not after neoadjuvant therapy was significantly associated with worse overall survival. In conclusion, chemotherapy increases CDA expression in xenografts, which is consistent with our in vitro results in MPM and lung cancer. A subset of matched patient samples showed increased CDA expression after therapy, suggesting that a schedule-dependent treatment strategy based on chemotherapy and capecitabine may benefit a selected MPM patient population.


Assuntos
Capecitabina , Citidina Desaminase , Mesotelioma Maligno , Pemetrexede , Neoplasias Pleurais , Ensaios Antitumorais Modelo de Xenoenxerto , Humanos , Capecitabina/farmacologia , Animais , Linhagem Celular Tumoral , Mesotelioma Maligno/tratamento farmacológico , Mesotelioma Maligno/metabolismo , Mesotelioma Maligno/patologia , Citidina Desaminase/metabolismo , Citidina Desaminase/genética , Camundongos , Pemetrexede/farmacologia , Neoplasias Pleurais/tratamento farmacológico , Neoplasias Pleurais/metabolismo , Neoplasias Pleurais/patologia , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Mesotelioma/tratamento farmacológico , Mesotelioma/metabolismo , Mesotelioma/patologia , Feminino , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos
12.
Nat Commun ; 15(1): 7452, 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39198414

RESUMO

The global epidemic of Mpox virus (MPXV) continues, and a local outbreak has occurred in Shenzhen city since June 2023. Herein, the evolutionary trajectory and characteristics of MPXV in 2023 were analyzed using 92 MPXV sequences from the Shenzhen outbreak and the available genomes from GISAID and GenBank databases. Phylogenetic tracing of the 92 MPXVs suggests that MPXVs in Shenzhen may have multiple sources of importation, and two main transmission chains have been established. The combination of phylogenetic relationships, epidemiological features, and mutation characteristics supports the emergence of a new lineage C.1.1. Together with the B.1 lineage diverging from the A.1 lineage, C.1.1 lineage diverging from the C.1 lineage may serve as another significant evolutionary events of MPXV. Moreover, increasing apolipoprotein B mRNA-editing catalytic polypeptide-like 3 (APOBEC3) related mutations, higher rate of missense mutations, and less mutations in the non-coding regions have been shown during MPXV evolution. Host regulation proteins of MPXV have accumulated considerable amino acid mutations since the B.1 lineage, and a lineage-defining APOBEC3-related mutation that disrupts the N2L gene encoding a viral innate immune modulator has been identified in the C.1.1 lineage. In summary, our study provides compelling evidence for the ongoing evolution of MPXV with specific features.


Assuntos
Evolução Molecular , Genoma Viral , Monkeypox virus , Filogenia , Humanos , Desaminases APOBEC/genética , China/epidemiologia , Citidina Desaminase/genética , Surtos de Doenças , Genoma Viral/genética , Genômica/métodos , Mutação , Monkeypox virus/genética
13.
G3 (Bethesda) ; 14(10)2024 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-39150943

RESUMO

Human APOBEC single-strand (ss) specific DNA and RNA cytidine deaminases change cytosines to uracils (U's) and function in antiviral innate immunity and RNA editing and can cause hypermutation in chromosomes. The resulting U's can be directly replicated, resulting in C to T mutations, or U-DNA glycosylase can convert the U's to abasic (AP) sites which are then fixed as C to T or C to G mutations by translesion DNA polymerases. We noticed that in yeast and in human cancers, contributions of C to T and C to G mutations depend on the origin of ssDNA mutagenized by APOBECs. Since ssDNA in eukaryotic genomes readily binds to replication protein A (RPA) we asked if RPA could affect APOBEC-induced mutation spectrum in yeast. For that purpose, we expressed human APOBECs in the wild-type (WT) yeast and in strains carrying a hypomorph mutation rfa1-t33 in the large RPA subunit. We confirmed that the rfa1-t33 allele can facilitate mutagenesis by APOBECs. We also found that the rfa1-t33 mutation changed the ratio of APOBEC3A-induced T to C and T to G mutations in replicating yeast to resemble a ratio observed in long persistent ssDNA in yeast and in cancers. We present the data suggesting that RPA may shield APOBEC formed U's in ssDNA from Ung1, thereby facilitating C to T mutagenesis through the accurate copying of U's by replicative DNA polymerases. Unexpectedly, we also found that for U's shielded from Ung1 by WT RPA, the mutagenic outcome is reduced in the presence of translesion DNA polymerase zeta.


Assuntos
Mutagênese , Mutação , Proteína de Replicação A , Saccharomyces cerevisiae , Proteína de Replicação A/metabolismo , Proteína de Replicação A/genética , Humanos , Saccharomyces cerevisiae/genética , Desaminases APOBEC/metabolismo , Desaminases APOBEC/genética , Citidina Desaminase/metabolismo , Citidina Desaminase/genética , DNA de Cadeia Simples/metabolismo , Desaminase APOBEC-1/genética , Desaminase APOBEC-1/metabolismo , Subunidades Proteicas/metabolismo , Subunidades Proteicas/genética
14.
Genetics ; 227(4)2024 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-38946641

RESUMO

APOBEC proteins are cytidine deaminases that restrict the replication of viruses and transposable elements. Several members of the APOBEC3 family, APOBEC3A, APOBEC3B, and APOBEC3H-I, can access the nucleus and cause what is thought to be indiscriminate deamination of the genome, resulting in mutagenesis and genome instability. Although APOBEC3C is also present in the nucleus, the full scope of its deamination target preferences is unknown. By expressing human APOBEC3C in a yeast model system, I have defined the APOBEC3C mutation signature, as well as the preferred genome features of APOBEC3C targets. The APOBEC3C mutation signature is distinct from those of the known cancer genome mutators APOBEC3A and APOBEC3B. APOBEC3C produces DNA strand-coordinated mutation clusters, and APOBEC3C mutations are enriched near the transcription start sites of active genes. Surprisingly, APOBEC3C lacks the bias for the lagging strand of DNA replication that is seen for APOBEC3A and APOBEC3B. The unique preferences of APOBEC3C constitute a mutation profile that will be useful in defining sites of APOBEC3C mutagenesis in human genomes.


Assuntos
Citidina Desaminase , Mutação , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Humanos , Genoma Humano , Replicação do DNA , Proteínas/genética , Proteínas/metabolismo , Mutagênese , Saccharomyces cerevisiae/genética , Antígenos de Histocompatibilidade Menor
15.
DNA Repair (Amst) ; 141: 103734, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39047499

RESUMO

Enzymes of the apolipoprotein B mRNA editing catalytic polypeptide like (APOBEC) family are cytosine deaminases that convert cytosine to uracil in DNA and RNA. Among these proteins, APOBEC3 sub-family members, APOBEC3A (A3A) and APOBEC3B (A3B), are prominent sources of mutagenesis in cancer cells. The aberrant expression of A3A and A3B in cancer cells leads to accumulation of mutations with specific single-base substitution (SBS) signatures, characterized by C→T and C→G changes, in a number of tumor types. In addition to fueling mutagenesis, A3A and A3B, particularly A3A, induce DNA replication stress, DNA damage, and chromosomal instability through their catalytic activities, triggering a range of cellular responses. Thus, A3A/B have emerged as key drivers of genome evolution during cancer development, contributing to tumorigenesis, tumor heterogeneity, and therapeutic resistance. Yet, the expression of A3A/B in cancer cells presents a cancer vulnerability that can be exploited therapeutically. In this review, we discuss the recent studies that shed light on the mechanisms regulating A3A expression and the impact of A3A in cancer. We also review recent advances in the development of A3A inhibitors and provide perspectives on the future directions of A3A research.


Assuntos
Citidina Desaminase , Neoplasias , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Citidina Desaminase/metabolismo , Regulação Neoplásica da Expressão Gênica , Animais , Proteínas/metabolismo , Proteínas/genética , Dano ao DNA , Mutagênese
16.
PLoS Genet ; 20(7): e1011367, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39058749

RESUMO

The pathway for axon regeneration in Caenorhabditis elegans is activated by SVH-1, a growth factor belonging to the HGF/plasminogen family. SVH-1 is a dual-function factor that acts as an HGF-like growth factor to promote axon regeneration and as a protease to regulate early development. It is important to understand how SVH-1 is converted from a protease to a growth factor for axon regeneration. In this study, we demonstrate that cytidine deaminase (CDD) SVH-17/CDD-2 plays a role in the functional conversion of SVH-1. We find that the codon exchange of His-755 to Tyr in the Asp-His-Ser catalytic triad of SVH-1 can suppress the cdd-2 defect in axon regeneration. Furthermore, the stem hairpin structure around the His-755 site in svh-1 mRNA is required for the activation of axon regeneration by SVH-1. These results suggest that CDD-2 promotes axon regeneration by transforming the function of SVH-1 from a protease to a growth factor through modification of svh-1 mRNA.


Assuntos
Axônios , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Citidina Desaminase , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Axônios/metabolismo , Axônios/fisiologia , Citidina Desaminase/metabolismo , Citidina Desaminase/genética , Regeneração Nervosa/genética , Regeneração Nervosa/fisiologia , Fator de Crescimento de Hepatócito/metabolismo , Fator de Crescimento de Hepatócito/genética , Regeneração/genética
17.
Viruses ; 16(7)2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39066304

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has acquired multiple mutations since its emergence. Analyses of the SARS-CoV-2 genomes from infected patients exhibit a bias toward C-to-U mutations, which are suggested to be caused by the apolipoprotein B mRNA editing enzyme polypeptide-like 3 (APOBEC3, A3) cytosine deaminase proteins. However, the role of A3 enzymes in SARS-CoV-2 replication remains unclear. To address this question, we investigated the effect of A3 family proteins on SARS-CoV-2 replication in the myeloid leukemia cell line THP-1 lacking A3A to A3G genes. The Wuhan, BA.1, and BA.5 variants had comparable viral replication in parent and A3A-to-A3G-null THP-1 cells stably expressing angiotensin-converting enzyme 2 (ACE2) protein. On the other hand, the replication and infectivity of these variants were abolished in A3A-to-A3G-null THP-1-ACE2 cells in a series of passage experiments over 20 days. In contrast to previous reports, we observed no evidence of A3-induced SARS-CoV-2 mutagenesis in the passage experiments. Furthermore, our analysis of a large number of publicly available SARS-CoV-2 genomes did not reveal conclusive evidence for A3-induced mutagenesis. Our studies suggest that A3 family proteins can positively contribute to SARS-CoV-2 replication; however, this effect is deaminase-independent.


Assuntos
Desaminases APOBEC , COVID-19 , Citidina Desaminase , SARS-CoV-2 , Replicação Viral , Humanos , SARS-CoV-2/genética , SARS-CoV-2/fisiologia , SARS-CoV-2/metabolismo , Desaminases APOBEC/metabolismo , Desaminases APOBEC/genética , COVID-19/virologia , COVID-19/metabolismo , Citidina Desaminase/metabolismo , Citidina Desaminase/genética , Células THP-1 , Mutação , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Genoma Viral
18.
PLoS Biol ; 22(7): e3002718, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38976757

RESUMO

Cancer initiates as a consequence of genomic mutations and its subsequent progression relies in part on increased production of ribosomes to maintain high levels of protein synthesis for unchecked cell growth. Recently, cytidine deaminases have been uncovered as sources of mutagenesis in cancer. In an attempt to form a connection between these 2 cancer driving processes, we interrogated the cytidine deaminase family of proteins for potential roles in human ribosome biogenesis. We identified and validated APOBEC3A and APOBEC4 as novel ribosome biogenesis factors through our laboratory's established screening platform for the discovery of regulators of nucleolar function in MCF10A cells. Through siRNA depletion experiments, we highlight APOBEC3A's requirement in making ribosomes and specific role within the processing and maturation steps that form the large subunit 5.8S and 28S ribosomal (r)RNAs. We demonstrate that a subset of APOBEC3A resides within the nucleolus and associates with critical ribosome biogenesis factors. Mechanistic insight was revealed by transient overexpression of both wild-type and a catalytically dead mutated APOBEC3A, which both increase cell growth and protein synthesis. Through an innovative nuclear RNA sequencing methodology, we identify only modest predicted APOBEC3A C-to-U target sites on the pre-rRNA and pre-mRNAs. Our work reveals a potential direct role for APOBEC3A in ribosome biogenesis likely independent of its editing function. More broadly, we found an additional function of APOBEC3A in cancer pathology through its function in ribosome biogenesis, expanding its relevance as a target for cancer therapeutics.


Assuntos
Nucléolo Celular , Proliferação de Células , Citidina Desaminase , Ribossomos , Humanos , Citidina Desaminase/metabolismo , Citidina Desaminase/genética , Nucléolo Celular/metabolismo , Ribossomos/metabolismo , Proliferação de Células/genética , RNA Ribossômico/metabolismo , RNA Ribossômico/genética , Linhagem Celular Tumoral , Proteínas/metabolismo , Proteínas/genética
19.
Nat Commun ; 15(1): 6331, 2024 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-39068148

RESUMO

Activation-induced cytidine deaminase (AID) is a B cell-specific mutator required for antibody diversification. However, it is also implicated in the etiology of several B cell malignancies. Evaluating the AID-induced mutation load in patients at-risk for certain blood cancers is critical in assessing disease severity and treatment options. We have developed a digital PCR (dPCR) assay that allows us to quantify mutations resulting from AID modification or DNA double-strand break (DSB) formation and repair at sites known to be prone to DSBs. Implementation of this assay shows that increased AID levels in immature B cells increase genome instability at loci linked to chromosomal translocation formation. This includes the CRLF2 locus that is often involved in translocations associated with a subtype of acute lymphoblastic leukemia (ALL) that disproportionately affects Hispanics, particularly those with Latin American ancestry. Using dPCR, we characterize the CRLF2 locus in B cell-derived genomic DNA from both Hispanic ALL patients and healthy Hispanic donors and found increased mutations in both, suggesting that vulnerability to DNA damage at CRLF2 may be driving this health disparity. Our ability to detect and quantify these mutations will potentiate future risk identification, early detection of cancers, and reduction of associated cancer health disparities.


Assuntos
Citidina Desaminase , Hispânico ou Latino , Mutação , Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores de Citocinas , Humanos , Citidina Desaminase/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Hispânico ou Latino/genética , Receptores de Citocinas/genética , Quebras de DNA de Cadeia Dupla , Linfócitos B/metabolismo , Linfócitos B/imunologia , Disparidades nos Níveis de Saúde , Translocação Genética , Loci Gênicos , América Latina , Feminino
20.
BMC Biol ; 22(1): 151, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38977974

RESUMO

BACKGROUND: RNA-DNA hybrids or R-loops are associated with deleterious genomic instability and protective immunoglobulin class switch recombination (CSR). However, the underlying phenomenon regulating the two contrasting functions of R-loops is unknown. Notably, the underlying mechanism that protects R-loops from classic RNase H-mediated digestion thereby promoting persistence of CSR-associated R-loops during CSR remains elusive. RESULTS: Here, we report that during CSR, R-loops formed at the immunoglobulin heavy (IgH) chain are modified by ribose 2'-O-methylation (2'-OMe). Moreover, we find that 2'-O-methyltransferase fibrillarin (FBL) interacts with activation-induced cytidine deaminase (AID) associated snoRNA aSNORD1C to facilitate the 2'-OMe. Moreover, deleting AID C-terminal tail impairs its association with aSNORD1C and FBL. Disrupting FBL, AID or aSNORD1C expression severely impairs 2'-OMe, R-loop stability and CSR. Surprisingly, FBL, AID's interaction partner and aSNORD1C promoted AID targeting to the IgH locus. CONCLUSION: Taken together, our results suggest that 2'-OMe stabilizes IgH-associated R-loops to enable productive CSR. These results would shed light on AID-mediated CSR and explain the mechanism of R-loop-associated genomic instability.


Assuntos
Citidina Desaminase , Switching de Imunoglobulina , Estruturas R-Loop , Switching de Imunoglobulina/genética , Citidina Desaminase/metabolismo , Citidina Desaminase/genética , Citidina Desaminase/química , Animais , Camundongos , Metilação , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/metabolismo , Recombinação Genética , RNA/metabolismo , RNA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA