Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 257
Filtrar
1.
Virol J ; 20(1): 148, 2023 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-37443068

RESUMO

BACKGROUND: The Epstein-Barr virus (EBV) is a prevalent oncovirus associated with a variety of human illnesses. BGLF5, an EBV DNase with alkaline nuclease (AN) activity, plays important roles in the viral life cycle and progression of human malignancies and has been suggested as a possible diagnostic marker and target for cancer therapy. Methods used conventionally for the detection of AN activity, radioactivity-based nuclease activity assay and DNA digestion detection by gel electrophoresis, are not suitable for screening AN inhibitors; the former approach is unsafe, and the latter is complicated. In the present study, a fluorescence-based nuclease activity assay was used to screen several natural compounds and identify an EBV DNase inhibitor. RESULTS: Fluorescence-based nuclease activity assays, in which the DNA substrate is labelled with PicoGreen dye, are cheaper, safer, and easier to perform. Herein, the results of the fluorescence-based nuclease activity assay were consistent with the results of the two conventional methods. In addition, the PicoGreen-labelling method was applied for the biochemical characterisation of viral nucleases. Using this approach, we explored EBV DNase inhibitors. After several rounds of screening, emodin, an anthraquinone derivative, was found to possess significant anti-EBV DNase activity. We verified the efficacy of emodin using the conventional DNA-cleavage assay. Furthermore, using comet assay and micronucleus formation detection, we confirmed that emodin can inhibit DNase-induced DNA damage and genomic instability. Additionally, emodin treatment inhibited EBV production. CONCLUSIONS: Using a PicoGreen-mediated nuclease activity assay, we successfully demonstrated that emodin has the potential to inhibit EBV DNase nuclease activity. Emodin also inhibits EBV DNase-related biological functions, suggesting that it is a potential inhibitor of EBV DNase.


Assuntos
Emodina , Infecções por Vírus Epstein-Barr , Humanos , Emodina/farmacologia , Herpesvirus Humano 4/genética , DNA , Desoxirribonucleases/química , Desoxirribonucleases/genética
2.
Gastroenterology ; 165(2): 357-373, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37178737

RESUMO

BACKGROUND & AIMS: The purpose of this study was to identify drivers of genomic evolution in esophageal adenocarcinoma (EAC) and other solid tumors. METHODS: An integrated genomics strategy was used to identify deoxyribonucleases correlating with genomic instability (as assessed from total copy number events in each patient) in 6 cancers. Apurinic/apyrimidinic nuclease 1 (APE1), identified as the top gene in functional screens, was either suppressed in cancer cell lines or overexpressed in normal esophageal cells and the impact on genome stability and growth was monitored in vitro and in vivo. The impact on DNA and chromosomal instability was monitored using multiple approaches, including investigation of micronuclei, acquisition of single nucleotide polymorphisms, whole genome sequencing, and/or multicolor fluorescence in situ hybridization. RESULTS: Expression of 4 deoxyribonucleases correlated with genomic instability in 6 human cancers. Functional screens of these genes identified APE1 as the top candidate for further evaluation. APE1 suppression in EAC, breast, lung, and prostate cancer cell lines caused cell cycle arrest; impaired growth and increased cytotoxicity of cisplatin in all cell lines and types and in a mouse model of EAC; and inhibition of homologous recombination and spontaneous and chemotherapy-induced genomic instability. APE1 overexpression in normal cells caused a massive chromosomal instability, leading to their oncogenic transformation. Evaluation of these cells by means of whole genome sequencing demonstrated the acquisition of changes throughout the genome and identified homologous recombination as the top mutational process. CONCLUSIONS: Elevated APE1 dysregulates homologous recombination and cell cycle, contributing to genomic instability, tumorigenesis, and chemoresistance, and its inhibitors have the potential to target these processes in EAC and possibly other cancers.


Assuntos
Adenocarcinoma , Resistencia a Medicamentos Antineoplásicos , Masculino , Animais , Camundongos , Humanos , Resistencia a Medicamentos Antineoplásicos/genética , Hibridização in Situ Fluorescente , Linhagem Celular Tumoral , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Carcinogênese/genética , Transformação Celular Neoplásica/genética , Recombinação Homóloga , Ciclo Celular , Instabilidade Genômica , Genômica , Instabilidade Cromossômica/genética , Desoxirribonucleases/genética , Evolução Molecular
3.
Proc Natl Acad Sci U S A ; 120(17): e2220982120, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37075072

RESUMO

Cell-free DNA (cfDNA) fragmentation is nonrandom, at least partially mediated by various DNA nucleases, forming characteristic cfDNA end motifs. However, there is a paucity of tools for deciphering the relative contributions of cfDNA cleavage patterns related to underlying fragmentation factors. In this study, through non-negative matrix factorization algorithm, we used 256 5' 4-mer end motifs to identify distinct types of cfDNA cleavage patterns, referred to as "founder" end-motif profiles (F-profiles). F-profiles were associated with different DNA nucleases based on whether such patterns were disrupted in nuclease-knockout mouse models. Contributions of individual F-profiles in a cfDNA sample could be determined by deconvolutional analysis. We analyzed 93 murine cfDNA samples of different nuclease-deficient mice and identified six types of F-profiles. F-profiles I, II, and III were linked to deoxyribonuclease 1 like 3 (DNASE1L3), deoxyribonuclease 1 (DNASE1), and DNA fragmentation factor subunit beta (DFFB), respectively. We revealed that 42.9% of plasma cfDNA molecules were attributed to DNASE1L3-mediated fragmentation, whereas 43.4% of urinary cfDNA molecules involved DNASE1-mediated fragmentation. We further demonstrated that the relative contributions of F-profiles were useful to inform pathological states, such as autoimmune disorders and cancer. Among the six F-profiles, the use of F-profile I could inform the human patients with systemic lupus erythematosus. F-profile VI could be used to detect individuals with hepatocellular carcinoma, with an area under the receiver operating characteristic curve of 0.97. F-profile VI was more prominent in patients with nasopharyngeal carcinoma undergoing chemoradiotherapy. We proposed that this profile might be related to oxidative stress.


Assuntos
Ácidos Nucleicos Livres , Humanos , Camundongos , Animais , Ácidos Nucleicos Livres/genética , Desoxirribonucleases/genética , Camundongos Knockout , Endonucleases/genética , Fragmentação do DNA , Endodesoxirribonucleases/genética
4.
Sci Rep ; 13(1): 1890, 2023 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-36732401

RESUMO

Adeno-associated viral (AAV) vector suspensions produced in either human derived HEK cells or in Spodoptera frugiperda (Sf9) insect cells differ in terms of residual host cell components as well as species-specific post-translational modifications displayed on the AAV capsid proteins. Here we analysed the impact of these differences on the immunogenic properties of the vector. We stimulated human plasmacytoid dendritic cells with various lots of HEK cell-produced and Sf9 cell-produced AAV-CMV-eGFP vectors derived from different manufacturers. We found that AAV8-CMV-eGFP as well as AAV2-CMV-eGFP vectors induced lot-specific but not production platform-specific or manufacturer-specific inflammatory cytokine responses. These could be reduced or abolished by blocking toll-like receptor 9 signalling or by enzymatically reducing DNA in the vector lots using DNase. Successful HEK cell transduction by DNase-treated AAV lots and DNA analyses demonstrated that DNase did not affect the integrity of the vector but degraded extra-viral DNA. We conclude that both HEK- and Sf9-cell derived AAV preparations can contain immunogenic extra-viral DNA components which can trigger lot-specific inflammatory immune responses. This suggests that improved strategies to remove extra-viral DNA impurities may be instrumental in reducing the immunogenic properties of AAV vector preparations.


Assuntos
Infecções por Citomegalovirus , DNA Viral , Humanos , Dependovirus/genética , Vetores Genéticos/genética , Receptor Toll-Like 9/genética , Imunidade Inata , Células Dendríticas , Desoxirribonucleases/genética , Transdução Genética
5.
Vet Microbiol ; 273: 109529, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35944391

RESUMO

Extracellular DNases/nucleases are important virulence factors in many bacteria. However, no DNase/nucleases have been reported in Mycobacterium avium subsp. paratuberculosis (MAP), which is a pathogen of paratuberculosis. Genome analyses of MAP K-10 revealed that the map3916c gene putatively encodes a nuclease. In this study, we show that MAP3916c is an extracellular nonspecific DNase requiring a divalent cation, especially Mg2+. The optimum DNase activity of MAP3916c was exhibited at 41 °C and pH 9.0. Site-directed mutagenesis studies indicated that 125-Histidine is necessary for MAP3916c DNase activity. In addition, MAP3916c DNase could destroy the neutrophil extracellular traps (NETs) induced by Phorbol 12-myristate 13-acetate in vitro and degrade the NETs induced by MAP K-10 upon infection. Furthermore, MAP3916c DNase promoted the colonization of MAP K-10, induced the formation of granulomas in the liver and small intestine and promoted the release of IL-1ß, IL-6 and TNF-α inflammatory cytokines during the infection of mice. These results indicated that MAP3916c is relevant to NETs escape and the pathogenicity of MAP. It also provides a basis for further study of the function of nuclease activity on the MAP immune evasion.


Assuntos
Desoxirribonucleases , Armadilhas Extracelulares , Mycobacterium avium subsp. paratuberculosis , Paratuberculose , Animais , Desoxirribonucleases/genética , Desoxirribonucleases/metabolismo , Armadilhas Extracelulares/metabolismo , Macrófagos/microbiologia , Camundongos , Mycobacterium avium subsp. paratuberculosis/enzimologia , Mycobacterium avium subsp. paratuberculosis/patogenicidade , Paratuberculose/microbiologia , Virulência
6.
Clin Chem ; 68(7): 917-926, 2022 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-35587043

RESUMO

BACKGROUND: Jagged ends of plasma DNA are a recently recognized class of fragmentomic markers for cell-free DNA, reflecting the activity of nucleases. A number of recent studies have also highlighted the importance of jagged ends in the context of pregnancy and oncology. However, knowledge regarding the generation of jagged ends is incomplete. METHODS: Jaggedness of plasma DNA was analyzed based on Jag-seq, which utilized the differential methylation signals introduced by the DNA end-repair process. We investigated the jagged ends in plasma DNA using mouse models by deleting the deoxyribonuclease 1 (Dnase1), DNA fragmentation factor subunit beta (Dffb), or deoxyribonuclease 1 like 3 (Dnase1l3) gene. RESULTS: Aberrations in the profile of plasma DNA jagged ends correlated with the type of nuclease that had been genetically deleted, depending on nucleosomal structures. The deletion of Dnase1l3 led to a significant reduction of jaggedness for those plasma DNA molecules involving more than 1 nucleosome (e.g., size ranges 240-290 bp, 330-380 bp, and 420-470 bp). However, less significant effects of Dnase1 and Dffb deletions were observed regarding different sizes of DNA fragments. Interestingly, the aberration in plasma DNA jagged ends related to multinucleosomes was observed in human subjects with familial systemic lupus erythematosus with Dnase1l3 deficiency and human subjects with sporadic systemic lupus erythematosus. CONCLUSIONS: Detailed understanding of the relationship between nuclease and plasma DNA jaggedness has opened up avenues for biomarker development.


Assuntos
Ácidos Nucleicos Livres , Lúpus Eritematoso Sistêmico , Animais , Biomarcadores , Ácidos Nucleicos Livres/genética , DNA/genética , Desoxirribonucleases/genética , Endodesoxirribonucleases/genética , Feminino , Humanos , Lúpus Eritematoso Sistêmico/genética , Camundongos , Nucleossomos/genética , Gravidez
7.
Mol Cell Biochem ; 477(9): 2213-2233, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35460011

RESUMO

DNA fragmentation factor 40 (DFF40), or the caspase-activated DNase (CAD), is an endonuclease specific for double-stranded DNA. Alterations in its function and expression have been linked to apoptosis resistance, a mechanism likely used by cancer cells. However, how the DFF40-related apoptosis resistance pathway occurs remains unclear. Here, we sought to determine if DFF40 expression could be linked to cell metabolism through the regulation of mitochondrial integrity and function. We demonstrated that DFF40-deficient cells are more resistant to staurosporine and tributyltin (TBT)-induced apoptosis, and express higher levels of Mcl-1 at basal state. Treatment with TBT induces higher Bcl-2 and caspase-9 mRNA transcripts in DFF40 KO Jurkat cells, as well as enhanced Bcl-2 phosphorylation. A loss of DFF40 expression induces a higher mitochondrial mass, mtDNA copy number, mitochondrial membrane potential, and glycolysis rates in resting T cells. DFF40-deficient cells exhibit the Warburg effect phenotype, where they rely significantly more on glycolysis than oxidative phosphorylation and have a higher proliferative state, demonstrated by a higher Ki-67 transcription factor expression and AKT phosphorylation. Finally, we demonstrated with cell fractioning that DFF40 can translocate to the mitochondria following apoptosis induction. Our study reveals that DFF40 may act as a regulator of mitochondria during cell death and its loss could compromise mitochondrial integrity and cause an energetic reprogramming in pathologies such as cancer.


Assuntos
Caspases , Neoplasias , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Caspases/metabolismo , Fragmentação do DNA , Desoxirribonucleases/genética , Desoxirribonucleases/metabolismo , Desoxirribonucleases/farmacologia , Humanos , Células Jurkat , Proteínas Proto-Oncogênicas c-bcl-2/genética
8.
mBio ; 12(6): e0255721, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34809467

RESUMO

Viruses have evolved a plethora of mechanisms to impair host innate immune responses. Herpes simplex virus type 1 (HSV-1), a double-stranded linear DNA virus, impairs the mitochondrial network and dynamics predominantly through the UL12.5 gene. We demonstrated that HSV-1 infection induced a remodeling of mitochondrial shape, resulting in a fragmentation of the mitochondria associated with a decrease in their volume and an increase in their sphericity. This damage leads to the release of mitochondrial DNA (mtDNA) to the cytosol. By generating a stable THP-1 cell line expressing the DNase I-mCherry fusion protein and a THP-1 cell line specifically depleted of mtDNA upon ethidium bromide treatment, we showed that cytosolic mtDNA contributes to type I interferon and APOBEC3A upregulation. This was confirmed by using an HSV-1 strain (KOS37 UL98-SPA) with a deletion of the UL12.5 gene that impaired its ability to induce mtDNA stress. Furthermore, by using an inhibitor of RNA polymerase III, we demonstrated that upon HSV-1 infection, cytosolic mtDNA enhanced type I interferon induction through the RNA polymerase III/RIG-I pathway. APOBEC3A was in turn induced by interferon. Deep sequencing analyses of cytosolic mtDNA mutations revealed an APOBEC3A signature predominantly in the 5'TpCpG context. These data demonstrate that upon HSV-1 infection, the mitochondrial network is disrupted, leading to the release of mtDNA and ultimately to its catabolism through APOBEC3-induced mutations. IMPORTANCE Herpes simplex virus 1 (HSV-1) impairs the mitochondrial network through the viral protein UL12.5. This leads to the fusion of mitochondria and simultaneous release of mitochondrial DNA (mtDNA) in a mouse model. We have shown that released mtDNA is recognized as a danger signal, capable of stimulating signaling pathways and inducing the production of proinflammatory cytokines. The expression of the human cytidine deaminase APOBEC3A is highly upregulated by interferon responses. This enzyme catalyzes the deamination of cytidine to uridine in single-stranded DNA substrates, resulting in the catabolism of edited DNA. Using human cell lines deprived of mtDNA and viral strains deficient in UL12, we demonstrated the implication of mtDNA in the production of interferon and APOBEC3A expression during viral infection. We have shown that HSV-1 induces mitochondrial network fragmentation in a human model and confirmed the implication of RNA polymerase III/RIG-I signaling in the capture of cytosolic mtDNA.


Assuntos
Proteína DEAD-box 58/metabolismo , Herpes Simples/metabolismo , Herpesvirus Humano 1/fisiologia , Interferon beta/metabolismo , Mitocôndrias/virologia , RNA Polimerase III/metabolismo , Receptores Imunológicos/metabolismo , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Proteína DEAD-box 58/genética , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Desoxirribonucleases/genética , Desoxirribonucleases/metabolismo , Herpes Simples/genética , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Interações Hospedeiro-Patógeno , Humanos , Interferon beta/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas/genética , Proteínas/metabolismo , RNA Polimerase III/genética , Receptores Imunológicos/genética , Transdução de Sinais , Proteínas Virais/genética , Proteínas Virais/metabolismo
9.
Biochem Pharmacol ; 194: 114801, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34678222

RESUMO

The regulation of the apoptotic pathway is one of the most studied mechanisms regarding cancer cell resistance. Many mutations have been linked to drug resistance. The DNA fragmentation factor 40 (DFF40) has been gaining interest regarding cancer cell response to chemotherapy and patient outcomes. Glioblastomas and uterine leiomyosarcomas have been shown to have a downregulation of DFF40 expression, conferring a poor patient prognosis. In concordance with these observations, in this study, we showed that DFF40 gene is also downregulated in breast, endocervical, ovarian, lung, pancreas and glioblastomas. DFF40 is the endonuclease responsible of DNA fragmentation during apoptosis. In this study, we sought to determine if a DFF40 deficiency in Jurkat T cells could impact the sensitivity to conventional chemotherapy drugs. CRISPR-cas9 generated DFF40 knockout (DFF40 KO) stable Jurkat cells and wild-type (DFF40 WT) cells were treated with different antimetabolites and topoisomerase II (TOP2) inhibitors, and cell viability was subsequently assessed. DFF40 deficient cells show chemoresistance to antimetabolites (e.g. methotrexate, 6-mercaptopurine and cytarabine) and surprisingly, they are more sensitive to TOP2 inhibitors (e.g. etoposide and teniposide). DFF40 deficient cells exposed to cytarabine present lower phosphatidylserine translocation levels to the outer cell membrane layer. Etoposide exposure in DFF40 deficient cells induces higher mortality levels and downregulation of Bcl-xL cells compared to DFF40 expressing T cells. The abolition of DFF40 expression in Jurkat cells significantly impairs histone H2AX phosphorylation following etoposide and cytarabine treatments. Our findings suggest that DFF40 is a novel key target in cancer cell resistance that potentially regulates genomic stability.


Assuntos
Apoptose/fisiologia , Desoxirribonucleases/deficiência , Resistencia a Medicamentos Antineoplásicos/fisiologia , Neoplasias/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/deficiência , Transdução de Sinais/fisiologia , Linfócitos T/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Desoxirribonucleases/genética , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Técnicas de Inativação de Genes , Células HeLa , Humanos , Células Jurkat , Neoplasias/tratamento farmacológico , Neoplasias/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos
10.
Biotechnol Lett ; 43(10): 1967-1976, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34482510

RESUMO

PURPOSE: DNA fragmenting factor (DFF40), an endonuclease inducing irreversible apoptosis protein, is down-regulated in many types of tumor cells. iRGD is a tumor-penetrating peptide with high affinity to cancer cells overexpressing αVß3 receptor. The aim of this study was to produce the recombinant DFF40-iRGD protein as a new molecule to selectively induce cytotoxicity in cancer cells and evaluate its biological effects. METHODS: The three-dimensional structure of DFF40-iRGD was predicted using Modeller software and its interaction with αVß3 receptor was evaluated by HADDOCK web-server. Recombinant DFF40 and DFF40-iRGD proteins were produced using intein fusion system in Escherichia coli BL21 (DE3). To improve the soluble expression, the inducer concentration, temperature and incubation time were optimized. After purification of DFF40 and DFF40-iRGD using chitin column, the cytotoxic and apoptotic effects of the proteins against MDA-MB-231 (αVß3 positive) and MCF-7 (αVß3 negative) cell lines were evaluated using cell viability assay and flow cytometric analysis. RESULTS: The results of molecular docking indicated the proper interaction of DFF40-iRGD with the integrin receptor comparable to iRGD. The optimum conditions of soluble expression of proteins were the induction by 0.5 mM and 0.1 mM of IPTG for DFF40 and DFF40-iRGD, respectively, at 7 °C for 24 h. After 48 h of incubation, DFF40-iRGD exhibited significantly higher cytotoxic effect against MDA-MB-231 cells than MCF-7 cells as IC50 values of 19.25 and 41 nM were found for MDA-MB-231 and MCF-7 cells, respectively. However, DFF40 cytotoxicity was not significantly different in two cell lines. Furthermore, Flow cytometry results showed that the fusion protein can induce remarkably apoptotic cell death in cancer cells. CONCLUSION: In this study, DFF40-iRGD protein was produced in soluble form and its inhibitory effects on cancer cell survival and induction of apoptosis were established; therefore, it has the potential to be used as a drug candidate for targeted treatment of breast cancer, especially Triple Negative Breast Cancer Cells.


Assuntos
Apoptose/efeitos dos fármacos , Desoxirribonucleases/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , Proteínas Recombinantes de Fusão , Neoplasias de Mama Triplo Negativas/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Simulação de Acoplamento Molecular , Oligopeptídeos/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia
11.
Int J Toxicol ; 40(4): 380-387, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33960240

RESUMO

Downregulation of the apoptotic protein DNA fragmentation factor 40 (DFF40) is correlated with poor overall survival in some malignancies, including melanoma. In this study, DFF40 gene expression driven by survivin promoter, a tumor-specific promoter, was used to selectively induce cytotoxicity in melanoma cells. The activity and strength of survivin promoter were examined in B16F10 murine melanoma, and L929 murine normal fibroblast cell lines using enhanced green fluorescent protein reporter assay and reverse transcription polymerase chain reaction. The effect of expression of DFF40 under the control of cytomegalovirus (CMV) or survivin promoter on viability of cancerous and normal cells was determined by MTT [3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide] assay. Apoptosis induction by expression of DFF40 was evaluated using Annexin-V/propidium iodide staining. Our findings showed high activity of survivin promoter comparable to the control promoter (ie, CMV) in melanoma cells, while survivin activity in normal cells was negligible. Survivin promoter-derived DFF40 gene expression led to selective inhibition of cell viability and induction of apoptosis in cancerous cells. Low and sublethal concentrations of a chemotherapeutic drug, dacarbazine, significantly enhanced the growth inhibitory effect of DFF40 gene therapy. Combination of survivin-driven gene therapy and chemotherapy could be considered as a potential therapeutic treatment for melanoma and possibly other malignancies with similar features.


Assuntos
Antineoplásicos/farmacologia , Desoxirribonucleases/metabolismo , Melanoma/tratamento farmacológico , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Survivina/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Desoxirribonucleases/genética , Fibroblastos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Transgênicos , Proteínas de Ligação a Poli-ADP-Ribose/genética , Regiões Promotoras Genéticas , Survivina/genética
12.
Trends Genet ; 37(8): 758-770, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34006390

RESUMO

Cell-free DNA (cfDNA) is a widely used noninvasive biomarker for diagnosis and prognosis of multiple disease states. Emerging evidence suggests that cfDNA might not just be passive waste products of cell death but could have a physiological and pathological function in inflammation and autoimmunity. The balance of cfDNA generation and clearance may thus be vital in health and disease. In particular, plasma nuclease activity has been linked to multiple pathologies including cancer and systemic lupus erythematosus (SLE) and associated with profound changes in the nonrandom fragmentation of cfDNA. Lastly, in this review, we explore the effects of DNA fragmentation factor B (DFFB), DNASE1L3, and DNASE1 on cfDNA levels and their fragmentomic profiles, and what these recent insights reveal about the biology of cfDNA.


Assuntos
Ácidos Nucleicos Livres/genética , Desoxirribonuclease I/genética , Desoxirribonucleases/genética , Endodesoxirribonucleases/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , Autoimunidade/genética , Ácidos Nucleicos Livres/sangue , Fragmentação do DNA , Desoxirribonuclease I/sangue , Desoxirribonucleases/sangue , Endodesoxirribonucleases/sangue , Humanos , Inflamação/sangue , Inflamação/genética , Inflamação/patologia , Proteínas de Ligação a Poli-ADP-Ribose/sangue
13.
PLoS One ; 16(4): e0250291, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33905417

RESUMO

Maintaining genome integrity is particularly important in germ cells to ensure faithful transmission of genetic information across generations. Here we systematically describe germ cell mutagenesis in wild-type and 61 DNA repair mutants cultivated over multiple generations. ~44% of the DNA repair mutants analysed showed a >2-fold increased mutagenesis with a broad spectrum of mutational outcomes. Nucleotide excision repair deficiency led to higher base substitution rates, whereas polh-1(Polη) and rev-3(Polζ) translesion synthesis polymerase mutants resulted in 50-400 bp deletions. Signatures associated with defective homologous recombination fall into two classes: 1) brc-1/BRCA1 and rad-51/RAD51 paralog mutants showed increased mutations across all mutation classes, 2) mus-81/MUS81 and slx-1/SLX1 nuclease, and him-6/BLM, helq-1/HELQ or rtel-1/RTEL1 helicase mutants primarily accumulated structural variants. Repetitive and G-quadruplex sequence-containing loci were more frequently mutated in specific DNA repair backgrounds. Tandem duplications embedded in inverted repeats were observed in helq-1 helicase mutants, and a unique pattern of 'translocations' involving homeologous sequences occurred in rip-1 recombination mutants. atm-1/ATM checkpoint mutants harboured structural variants specifically enriched in subtelomeric regions. Interestingly, locally clustered mutagenesis was only observed for combined brc-1 and cep-1/p53 deficiency. Our study provides a global view of how different DNA repair pathways contribute to prevent germ cell mutagenesis.


Assuntos
Caenorhabditis elegans/genética , Reparo do DNA , DNA de Helmintos/genética , Regulação da Expressão Gênica , Genoma Helmíntico , Células Germinativas/metabolismo , Mutação , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proliferação de Células , Mapeamento Cromossômico , Dano ao DNA , DNA Helicases/genética , DNA Helicases/metabolismo , Replicação do DNA , DNA de Helmintos/metabolismo , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Desoxirribonucleases/genética , Desoxirribonucleases/metabolismo , Endonucleases/genética , Endonucleases/metabolismo , Células Germinativas/citologia , Isoenzimas/genética , Isoenzimas/metabolismo , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo
14.
Nature ; 590(7847): 624-629, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33461211

RESUMO

In the type III CRISPR-Cas immune response of prokaryotes, infection triggers the production of cyclic oligoadenylates that bind and activate proteins that contain a CARF domain1,2. Many type III loci are associated with proteins in which the CRISPR-associated Rossman fold (CARF) domain is fused to a restriction  endonuclease-like domain3,4. However, with the exception of the well-characterized Csm6 and Csx1 ribonucleases5,6, whether and how these inducible effectors provide defence is not known. Here we investigated a type III CRISPR accessory protein, which we name cyclic-oligoadenylate-activated single-stranded ribonuclease and single-stranded deoxyribonuclease 1 (Card1). Card1 forms a symmetrical dimer that has a large central cavity between its CRISPR-associated Rossmann fold and restriction endonuclease domains that binds cyclic tetra-adenylate. The binding of ligand results in a conformational change comprising the rotation of individual monomers relative to each other to form a more compact dimeric scaffold, in which a manganese cation coordinates the catalytic residues and activates the cleavage of single-stranded-but not double-stranded-nucleic acids (both DNA and RNA). In vivo, activation of Card1 induces dormancy of the infected hosts to provide immunity against phage infection and plasmids. Our results highlight the diversity of strategies used in CRISPR systems to provide immunity.


Assuntos
Nucleotídeos de Adenina/metabolismo , Sistemas CRISPR-Cas/imunologia , DNA de Cadeia Simples/metabolismo , Desoxirribonucleases/metabolismo , Endorribonucleases/metabolismo , Oligorribonucleotídeos/metabolismo , RNA/metabolismo , Staphylococcus/enzimologia , Staphylococcus/imunologia , Nucleotídeos de Adenina/imunologia , Trifosfato de Adenosina/metabolismo , Bacteriófagos/imunologia , Bacteriófagos/fisiologia , Biocatálise , Domínio Catalítico , Desoxirribonucleases/química , Desoxirribonucleases/genética , Endorribonucleases/química , Endorribonucleases/genética , Ativação Enzimática , Ligantes , Manganês/química , Manganês/metabolismo , Modelos Moleculares , Oligorribonucleotídeos/imunologia , Plasmídeos/genética , Plasmídeos/metabolismo , Multimerização Proteica , Rotação , Staphylococcus/crescimento & desenvolvimento , Staphylococcus/virologia , Especificidade por Substrato
15.
Apoptosis ; 26(1-2): 9-23, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33387146

RESUMO

Maintenance of genomic stability in cells is primordial for cellular integrity and protection against tumor progression. Many factors such as ultraviolet light, oxidative stress, exposure to chemical reagents, particularly mutagens and radiation, can alter the integrity of the genome. Thus, human cells are equipped with many mechanisms that prevent these irreversible lesions in the genome, as DNA repair pathways, cell cycle checkpoints, and telomeric function. These mechanisms activate cellular apoptosis to maintain DNA stability. Emerging studies have proposed a new protein in the maintenance of genomic stability: the DNA fragmentation factor (DFF). The DFF40 is an endonuclease responsible of the oligonucleosomal fragmentation of the DNA during apoptosis. The lack of DFF in renal carcinoma cells induces apoptosis without oligonucleosomal fragmentation, which poses a threat to genetic information transfer between cancerous and healthy cells. In this review, we expose the link between the DFF and genomic instability as the source of disease development.


Assuntos
Desoxirribonucleases/metabolismo , Instabilidade Genômica , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Animais , Apoptose , Fragmentação do DNA , Reparo do DNA , Desoxirribonucleases/genética , Humanos , Neoplasias/enzimologia , Neoplasias/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética
16.
Life Sci ; 265: 118866, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33301810

RESUMO

AIMS: GnRH-DFF40 (gonadotropin releasing hormone-DNA fragmentation factor 40) humanized recombinant immunotoxin serves as a prospective candidate for targeted therapy of malignancies with over-expressed gonadotropin releasing hormone receptor (GnRHR). In this study, we attempted to generate a GnRH-based chimeric protein composed of human DFF40 fused with GnRH which encodes an apoptotic nuclease and specifically targets cancer cells displaying GnRH receptor overexpression. MATERIALS AND METHODS: A codon optimized, synthetic GnRH-DFF40 fusion gene and its single counterpart (DFF40) were constructed in pET28a expression vector. Cytotoxicity of these expressed proteins were evaluated on three breast cancer cell lines (MCF7, MDA-MB231, and SKBR3). The stability and biological activity of the recombinant proteins were investigated in the treated cell line and cell-free system. Also, the ability of this fusion and its single form in inducing apoptosis, and inhibiting metastasis and migration were evaluated by flow cytometry, migration assay and wound healing analysis, respectively. In silico analyses were also done to understand the specific interactions between GnRH and its receptor. KEY FINDINGS: GnRH-DFF40 fusion protein and DFF40 were successfully expressed. The purified chimeric protein showed dose-dependent cytotoxicity against all three cell lines. The recombinant fusion protein was biologically active with nucleolytic functionality and apoptosis induction ability. Moreover, the fusion could inhibit the invasion property of MDA-MB-231 cells. In silico analysis also showed that four residues from GnRH domain and 11 GnRHR residues had the most interaction sites for specific targeted delivery of the immunotoxin in cancer cells. SIGNIFICANCE: Fusion construct could be a prospective candidate for targeted therapy of cancers upregulating GnRH receptor.


Assuntos
Neoplasias da Mama/terapia , Desoxirribonucleases/genética , Imunotoxinas/farmacologia , Proteínas de Ligação a Poli-ADP-Ribose/genética , Receptores LHRH/genética , Proteínas Recombinantes de Fusão/farmacologia , Apoptose/fisiologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sistema Livre de Células , Simulação por Computador , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Humanos , Imunotoxinas/administração & dosagem , Células MCF-7 , Terapia de Alvo Molecular , Proteínas Recombinantes de Fusão/administração & dosagem
17.
Elife ; 92020 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-33191912

RESUMO

DNA viruses in the family Poxviridae encode poxin enzymes that degrade the immune second messenger 2'3'-cGAMP to inhibit cGAS-STING immunity in mammalian cells. The closest homologs of poxin exist in the genomes of insect viruses suggesting a key mechanism of cGAS-STING evasion may have evolved outside of mammalian biology. Here we use a biochemical and structural approach to discover a broad family of 369 poxins encoded in diverse viral and animal genomes and define a prominent role for 2'3'-cGAMP cleavage in metazoan host-pathogen conflict. Structures of insect poxins reveal unexpected homology to flavivirus proteases and enable identification of functional self-cleaving poxins in RNA-virus polyproteins. Our data suggest widespread 2'3'-cGAMP signaling in insect antiviral immunity and explain how a family of cGAS-STING evasion enzymes evolved from viral proteases through gain of secondary nuclease activity. Poxin acquisition by poxviruses demonstrates the importance of environmental connections in shaping evolution of mammalian pathogens.


Assuntos
Desoxirribonucleases/metabolismo , Nucleotídeos Cíclicos/metabolismo , Vaccinia virus/metabolismo , Proteínas Virais/metabolismo , Animais , Sítios de Ligação , Clonagem Molecular , Desoxirribonucleases/genética , Evolução Molecular , Genoma , Lepidópteros/virologia , Mamíferos/genética , Mamíferos/metabolismo , Modelos Moleculares , Nucleotídeos Cíclicos/genética , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Conformação Proteica , Vírus de RNA/enzimologia , Vaccinia virus/genética , Proteínas Virais/genética
18.
Nat Commun ; 11(1): 4148, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32811834

RESUMO

We evaluate gene editing of HSV in a well-established mouse model, using adeno-associated virus (AAV)-delivered meganucleases, as a potentially curative approach to treat latent HSV infection. Here we show that AAV-delivered meganucleases, but not CRISPR/Cas9, mediate highly efficient gene editing of HSV, eliminating over 90% of latent virus from superior cervical ganglia. Single-cell RNA sequencing demonstrates that both HSV and individual AAV serotypes are non-randomly distributed among neuronal subsets in ganglia, implying that improved delivery to all neuronal subsets may lead to even more complete elimination of HSV. As predicted, delivery of meganucleases using a triple AAV serotype combination results in the greatest decrease in ganglionic HSV loads. The levels of HSV elimination observed in these studies, if translated to humans, would likely significantly reduce HSV reactivation, shedding, and lesions. Further optimization of meganuclease delivery and activity is likely possible, and may offer a pathway to a cure for HSV infection.


Assuntos
Desoxirribonucleases/genética , Dependovirus/genética , Infecções Oculares/terapia , Edição de Genes/métodos , Herpes Simples/terapia , Herpesvirus Humano 1/genética , Latência Viral/genética , Animais , Sistemas CRISPR-Cas/genética , Células Cultivadas , Chlorocebus aethiops , Infecções Oculares/genética , Infecções Oculares/virologia , Feminino , Células HEK293 , Herpes Simples/genética , Herpesvirus Humano 1/patogenicidade , Humanos , Camundongos , Neurônios/metabolismo , Neurônios/virologia , RNA-Seq , Análise de Célula Única , Gânglio Cervical Superior/metabolismo , Gânglio Cervical Superior/virologia , Células Vero
19.
Biochim Biophys Acta Mol Basis Dis ; 1866(5): 165724, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32061775

RESUMO

Removal of nuclei in lens fiber cells is required for organelle-free zone (OFZ) formation during lens development. Defect in degradation of nuclear DNA leads to cataract formation. DNase2ß degrades nuclear DNA of lens fiber cells during lens differentiation in mouse. Hsf4 is the principal heat shock transcription factor in lens and facilitates the lens differentiation. Knockout of Hsf4 in mouse and zebrafish resulted in lens developmental defect that was characterized by retaining of nuclei in lens fiber cells. In previous in vitro studies, we found that Hsf4 promoted DNase2ß expression in human and mouse lens epithelial cells. In this study, it was found that, instead of DNase2ß, DNase1l1l is uniquely expressed in zebrafish lens and was absent in Hsf4-/- zebrafish lens. Using CRISPR-Cas9 technology, a DNase1l1l knockout zebrafish line was constructed, which developed cataract. Deletion of DNase1l1l totally abrogated lens primary and secondary fiber cell denucleation process, whereas had little effect on the clearance of other organelles. The transcriptional regulation of DNase1l1l was dramatically impaired in Hsf4-/- zebrafish lens. Rescue of DNase1l1l mRNA into Hsf4-/- zebrafish embryos alleviated its defect in lens fiber cell denucleation. Our results in vivo demonstrated that DNase1l1l is the primary DNase responsible for nuclear DNA degradation in lens fiber cells, and Hsf4 can transcriptionally activate DNase1l1l expression in zebrafish.


Assuntos
Catarata/genética , Desoxirribonucleases/genética , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição de Choque Térmico/metabolismo , Cristalino/embriologia , Proteínas de Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Sistemas CRISPR-Cas/genética , Catarata/patologia , Núcleo Celular/metabolismo , Desoxirribonucleases/metabolismo , Modelos Animais de Doenças , Embrião não Mamífero , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Técnicas de Inativação de Genes , Fatores de Transcrição de Choque Térmico/genética , Humanos , Cristalino/citologia , Cristalino/metabolismo , Cristalino/patologia , Masculino , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
20.
Front Immunol ; 10: 2552, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31772562

RESUMO

Staphylococcus aureus is one of the first and most prevalent pathogens cultured from the airways of cystic fibrosis (CF) patients, which can persist there for extended periods. Airway infections in CF patients are characterized by a strong inflammatory response of highly recruited neutrophils. One killing mechanism of neutrophils is the formation of neutrophil extracellular traps (NETs), which capture and eradicate bacteria by extracellular fibers of neutrophil chromatin decorated with antimicrobial granule proteins. S. aureus secretes nuclease, which can degrade NETs. We hypothesized, that S. aureus adapts to the airways of CF patients during persistent infection by escaping from NET-mediated killing via an increase of nuclease activity. Sputum samples of CF patients with chronic S. aureus infection were visualized by confocal microscopy after immuno-fluorescence staining for NET-specific markers, S. aureus bacteria and overall DNA structures. Nuclease activity was analyzed in sequential isogenic long persisting S. aureus isolates, as confirmed by whole genome sequencing, from an individual CF patient using a FRET-based nuclease activity assay. Additionally, some of these isolates were selected and analyzed by qRT-PCR to determine the expression of nuc1 and regulators of interest. NET-killing assays were performed with clinical S. aureus isolates to evaluate killing and bacterial survival depending on nuclease activity. To confirm the role of nuclease during NET-mediated killing, a clinical isolate with low nuclease activity was transformed with a nuclease expression vector (pCM28nuc). Furthermore, two sputa from an individual CF patient were subjected to RNA-sequence analysis to evaluate the activity of nuclease in vivo. In sputa, S. aureus was associated to extracellular DNA structures. Nuclease activity in clinical S. aureus isolates increased in a time-and phenotype-dependent manner. In the clinical isolates, the expression of nuc1 was inversely correlated to the activity of agr and was independent of saeS. NET-mediated killing was significantly higher in S. aureus isolates with low compared to isolates with high nuclease activity. Importantly, transformation of the clinical isolate with low nuclease activity with pCM28nuc conferred protection against NET-mediated killing confirming the beneficial role of nuclease for protection against NETs. Also, nuclease expression in in vivo sputa was high, which underlines the important role of nuclease within the highly inflamed CF airways. In conclusion, our data show that S. aureus adapts to the neutrophil-rich environment of CF airways with increasing nuclease expression most likely to avoid NET-killing during long-term persistence.


Assuntos
Proteínas de Bactérias/imunologia , Fibrose Cística/imunologia , Desoxirribonucleases/imunologia , Armadilhas Extracelulares/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/enzimologia , Proteínas de Bactérias/genética , Fibrose Cística/microbiologia , Desoxirribonucleases/genética , Humanos , Escarro/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA