Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nucleic Acids Res ; 51(5): 2151-2176, 2023 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-36715342

RESUMO

How the diverse neural cell types emerge from multipotent neural progenitor cells during central nervous system development remains poorly understood. Recent scRNA-seq studies have delineated the developmental trajectories of individual neural cell types in many neural systems including the neural retina. Further understanding of the formation of neural cell diversity requires knowledge about how the epigenetic landscape shifts along individual cell lineages and how key transcription factors regulate these changes. In this study, we dissect the changes in the epigenetic landscape during early retinal cell differentiation by scATAC-seq and identify globally the enhancers, enriched motifs, and potential interacting transcription factors underlying the cell state/type specific gene expression in individual lineages. Using CUT&Tag, we further identify the enhancers bound directly by four key transcription factors, Otx2, Atoh7, Pou4f2 and Isl1, including those dependent on Atoh7, and uncover the sequential and combinatorial interactions of these factors with the epigenetic landscape to control gene expression along individual retinal cell lineages such as retinal ganglion cells (RGCs). Our results reveal a general paradigm in which transcription factors collaborate and compete to regulate the emergence of distinct retinal cell types such as RGCs from multipotent retinal progenitor cells (RPCs).


Assuntos
Retina , Fatores de Transcrição , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Retina/citologia , Retina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
Ecotoxicol Environ Saf ; 259: 115010, 2023 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-37211000

RESUMO

Inorganic arsenic (iAs) contamination in drinking water is a global public health problem, and exposure to iAs is a known risk factor for bladder cancer. Perturbation of urinary microbiome and metabolome induced by iAs exposure may have a more direct effect on the development of bladder cancer. The aim of this study was to determine the impact of iAs exposure on urinary microbiome and metabolome, and to identify microbiota and metabolic signatures that are associated with iAs-induced bladder lesions. We evaluated and quantified the pathological changes of bladder, and performed 16S rDNA sequencing and mass spectrometry-based metabolomics profiling on urine samples from rats exposed to low (30 mg/L NaAsO2) or high (100 mg/L NaAsO2) iAs from early life (in utero and childhood) to puberty. Our results showed that iAs induced pathological bladder lesions, and more severe effects were noticed in the high-iAs group and male rats. Furthermore, six and seven featured urinary bacteria genera were identified in female and male offspring rats, respectively. Several characteristic urinary metabolites, including Menadione, Pilocarpine, N-Acetylornithine, Prostaglandin B1, Deoxyinosine, Biopterin, and 1-Methyluric acid, were identified significantly higher in the high-iAs groups. In addition, the correlation analysis demonstrated that the differential bacteria genera were highly correlated with the featured urinary metabolites. Collectively, these results suggest that exposure to iAs in early life not only causes bladder lesions, but also perturbs urinary microbiome composition and associated metabolic profiles, which shows a strong correlation. Those differential urinary genera and metabolites may contribute to bladder lesions, suggesting a potential for development of urinary biomarkers for iAs-induced bladder cancer.


Assuntos
Arsênio , Arsenicais , Microbiota , Neoplasias da Bexiga Urinária , Masculino , Feminino , Animais , Ratos , Arsênio/metabolismo , Bexiga Urinária/metabolismo , Arsenicais/metabolismo , Neoplasias da Bexiga Urinária/induzido quimicamente
3.
Arch Toxicol ; 94(3): 749-760, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31844926

RESUMO

Exposure to fluoride (F) or arsenite (As) through contaminated drinking water has been associated with chronic nephrotoxicity in humans. Autophagy is a regulated mechanism ubiquitous for the body in a toxic environment with F and As, but the underlying mechanisms of autophagy in the single or combined nephrotoxicity of F and As are unclear. In the present study, we established a rat model of prenatal and postnatal exposure to F and As with the aim of investigating the mechanism underlying nephrotoxicity of these pollutants in offspring. Rats were randomly divided into four groups that received NaF (100 mg/L), NaAsO2 (50 mg/L), or NaF (100 mg/L) with NaAsO2 (50 mg/L) in drinking water or clean water during pregnancy and lactation; after weaning, pups were exposed to the same treatment as their mothers until puberty. The results revealed that F and As exposure (alone or combined) led to significant increases of arsenic and fluoride levels in blood and bone, respectively. In this context, F and/or As disrupted histopathology and ultrastructure in the kidney, and also altered creatinine (CRE), urea nitrogen (BUN) and uric acid (UA) levels. Intriguingly, F and/or As uptake induced the formation of autophagosomes in kidney tissue and resulted in the upregulation of genes encoding autophagy-related proteins. Collectively, these results suggest that nephrotoxicity of F and As for offspring exposed to the pollutants from in utero to puberty is associated with deregulation of autophagy and there is an antagonism between F and As in the toxicity autophagy process.


Assuntos
Arsenitos/toxicidade , Autofagia/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Fluoretos/toxicidade , Insuficiência Renal/induzido quimicamente , Animais , Feminino , Rim/efeitos dos fármacos , Masculino , Gravidez , Ratos , Maturidade Sexual/efeitos dos fármacos
4.
Chem Res Toxicol ; 32(8): 1572-1582, 2019 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-31240907

RESUMO

Cisplatin is a platinum-based chemotherapeutic drug widely used in the treatment of various cancers such as testicular, ovarian, lung, bladder, and cervical cancers. However, its use and the dosage range applied have been limited by severe side effects (e.g., nephrotoxicity and ototoxicity) and by the development of resistance to cisplatin in patients during treatment. Metal chelators have shown promising potential in overcoming these problems often associated with platinum drugs. Previously, a new chelating agent, sodium (S)-2-(dithiocarboxylato((2S,3R,4R,5R)-2,3,4,5,6-pentahydroxyhexyl)amino)-4(methylthio)butanoate (GMDTC), was developed. In this study, we examined the effect of GMDTC in modifying cisplatin-induced toxicities following in vitro and in vivo exposures. GMDTC treatment dramatically reduced cisplatin-induced apoptosis and cytotoxicity in HK2 cells by decreasing the amount of intracellular platinum. In the 4T1 breast cancer mouse model, GMDTC reduced cisplatin-induced nephrotoxicity by reducing cisplatin deposition in the kidney. GMDTC attenuated cisplatin-induced elevations in blood urea nitrogen and plasma creatinine, ameliorated renal tubular dilation and vacuolation, and prevented necrosis of glomeruli and renal tubular cells. GMDTC also inhibited cisplatin-induced ototoxicity as shown by improved hearing loss which was assessed using the auditory brainstem response test. Furthermore, GMDTC attenuated cisplatin-induced hematotoxicity and hepatotoxicity. Importantly, co-treatment of cisplatin with GMDTC did not affect cisplatin antitumor efficacy. Tumor growth, size, and metastasis were all comparable between the cisplatin only and cisplatin-GMDTC co-treatment groups. In conclusion, the current study suggests that GMDTC reduces cisplatin-induced systemic toxicity by preventing the accumulation and assisting in the removal of intracellular cisplatin, without compromising cisplatin therapeutic activity. These results support the development of GMDTC as a chemotherapy protector and rescue agent to overcome the toxicity of and resistance to platinum-based antineoplastic drugs.


Assuntos
Antineoplásicos/toxicidade , Neoplasias da Mama/tratamento farmacológico , Quelantes/farmacologia , Cisplatino/farmacologia , Cisplatino/toxicidade , Glucosamina/análogos & derivados , Metionina/análogos & derivados , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/sangue , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/antagonistas & inibidores , Feminino , Glucosamina/farmacologia , Metionina/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Células Tumorais Cultivadas
5.
Carcinogenesis ; 38(4): 378-390, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28182198

RESUMO

Our previous studies have shown that chronic exposure to low doses of monomethylarsonous acid (MMAIII) causes global histone acetylation dysregulation in urothelial cells (UROtsa cells) during the course of malignant transformation. To reveal the relationship between altered histone acetylation patterns and aberrant gene expression, more specifically, the carcinogenic relevance of these alterations, we performed a time-course analysis of the binding patterns of histone 3 lysine 18 acetylation (H3K18ac) across the genome and generated global gene-expression profiles from this UROtsa cell malignant transformation model. We showed that H3K18ac, one of the most significantly upregulated histone acetylation sites following MMAIII exposure, was enriched at gene promoter-specific regions across the genome and that MMAIII-induced upregulation of H3K18ac led to an altered binding pattern in a large number of genes that was most significant during the critical window for MMAIII-induced UROtsa cells' malignant transformation. Some genes identified as having a differential binding pattern with H3K18ac, acted as upstream regulators of critical gene networks with known functions in tumor development and progression. The altered H3K18ac binding patterns not only led to changes in expression of these directly affected upstream regulators but also resulted in gene-expression changes in their regulated networks. Collectively, our data suggest that MMAIII-induced alteration of histone acetylation patterns in UROtsa cells led to a time- and malignant stage-dependent aberrant gene-expression pattern, and that some gene regulatory networks were altered in accordance with their roles in carcinogenesis, probably contributing to MMAIII-induced urothelial cell malignant transformation and carcinogenesis.


Assuntos
Acetilação/efeitos dos fármacos , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/genética , Expressão Gênica/genética , Histonas/genética , Compostos Organometálicos/farmacologia , Bexiga Urinária/patologia , Animais , Células Cultivadas , Feminino , Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Redes Reguladoras de Genes/genética , Genoma/efeitos dos fármacos , Genoma/genética , Humanos , Lisina/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
6.
Biol Trace Elem Res ; 201(10): 4870-4881, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36692655

RESUMO

Co-contamination of arsenic (As) and fluoride (F) is widely distributed in groundwater, which are known risk factors for the nephrotoxicity. Emerging evidence has linked environmentally associated nephrotoxicity with the disturbance of gut microbiota and blood metabolites. In this study, we generated gut microbiota and blood metabolomic profile and identified multiple serum metabolites and gut bacteria species, which were associated with kidney injury on rat model exposed to As and F alone or combined. Combined As and F exposure significantly increased creatinine level. Abnormal autophagosomes and lysosome were observed, and the autophagic genes were enhanced in kidney tissue after single and combined As and F exposure. The metabolome data showed that single and combined As and F exposure remarkably altered the serum metabolites associated with the proximal tubule reabsorption function pathway, with glutamine and alpha-ketoglutarate level decreased in all exposed group. Furthermore, phosphatidylethanolamine (PE), the key contributor of autophagosomes, was decreased significantly in As and F + As exposed groups during the screen of autophagy-animal pathway. Multiple altered gut bacterial microbiota at phylum and species levels post As and F exposure were associated with targeted kidney injury, including p_Bacteroidetes, s_Chromohalobacter_unclassified, s_Halomonas_unclassified, s_Ignatzschineria_unclassified, s_Bacillus_subtilis, and s_Brevundimonas_sp._NA6. Meanwhile, our analysis indicated that As and F co-exposure possessed an interactive influence on gut microbiota. In conclusion, single or combined As and F exposure leads to the disruption of serum metabolic and gut microbiota profiles. Multiple metabolites and bacterial species are identified and associated with nephrotoxicity, which have potential to be developed as biomarkers of As and/or F-induced kidney damage.


Assuntos
Arsênio , Microbioma Gastrointestinal , Insuficiência Renal , Ratos , Animais , Arsênio/toxicidade , Fluoretos/toxicidade , Microbioma Gastrointestinal/genética , Bactérias
7.
Sci Total Environ ; 767: 144924, 2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-33636766

RESUMO

Co-exposure to inorganic arsenic (iAs) and fluoride (F-) and their collective actions on cardiovascular systems have been recognized as a global public health concern. Emerging studies suggest an association between the perturbation of gut bacterial microbiota and adverse cardiovascular effects (CVEs), both of which are the consequence of iAs and F- exposure in human and experimental animals. The aim of this study was to fill the gap of understanding the relationship among co-exposure to iAs and F-, gut microbiota perturbation, and adverse CVEs. We systematically assessed cardiac morphology and functions (blood pressure, echocardiogram, and electrocardiogram), and generated gut microbiota profiles using 16S rRNA gene sequencing on rats exposed to iAs (50 mg/L NaAsO2), F- (100 mg/L NaF) or combined iAs and F- (50 mg/L NaAsO2 + 100 mg/L NaF), in utero and during early postnatal periods (postnatal day 90). Correlation analysis was then performed to examine relationship between significantly altered microbiota and cardiac performance indices. Our results showed that co-exposure to iAs and F- resulted in more prominent effects in CVEs and perturbation of gut microbiota profiles, compared to iAs or F- treatment alone. Furthermore, nine bacterial genera (Adlercreutzia, Clostridium sensu stricto 1, Coprococcus 3, Romboutsia, [Bacteroides] Pectinophilus group, Lachnospiraceae NC2004 group, Desulfovibrio, and two unidentified genera in Muribaculaceae and Ruminococcaceae family), which differed significantly in relative abundance between control and iAs and F- co-exposure group, were strongly correlated with the higher risk of CVEs (correlation coefficient = 0.70-0.88, p < 0.05). Collectively, these results suggest that co-exposure to iAs and F- poses a higher risk of CVEs, and the part of the mode of action is potentially through inducing gut microbiota disruption, and the strong correlations between them indicate a high potential for the development of novel microbiome-based biomarkers of iAs and/or F- associated CVEs.


Assuntos
Arsênio , Sistema Cardiovascular , Microbioma Gastrointestinal , Animais , Arsênio/toxicidade , Fluoretos , RNA Ribossômico 16S/genética , Ratos
8.
Environ Int ; 140: 105763, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32371304

RESUMO

Many "hot spot" geographic areas across the world with drinking water co-contaminated with inorganic arsenic (iAs) and fluoride (F-), two of the most common natural contaminants in drinking water. Both iAs and F- are known neurotoxins and affect neurodevelopment of children. However, very few studies have investigated the neurodevelopmental effects of concurrent exposure to iAs and F-, which could potentially pose a greater risk than iAs or F- exposure alone. Further, perturbations of gut microbiota, which plays a regulatory role in neurodevelopment, resulting from iAs and F- exposure has been reported in numerous studies. There is lacking of information regarding to the relationship among concurrent iAs and F- exposure, microbiome disruption, and neurodevelopmental impacts. To fill these gaps, we treated offspring rats to iAs (50 mg/L NaAsO2) and F- (100 mg/L NaF), alone or combined from early life (in utero and childhood) to puberty. We applied Morris water maze test to assess spatial learning and memory of these rats and generated gut microbiome profiles using 16S rRNA gene sequencing. We showed that concurrent iAs and F- exposure caused more prominent neurodevelopmental effects in rats than either iAs or F- exposure alone. Moreover, Unsupervised Principal Coordinates Analysis (PCoA) and Linear Discriminant Analysis Effect Size (LEfSe) analysis of gut microbiome sequencing results separated concurrent exposure group from others, indicating a more sophisticated change of gut microbial communities occurred under the concurrent exposure condition. Further, a correlation analysis between indices of the water maze test and microbial composition at the genus level identified featured genera that were clearly associated with neurobehavioral performance of rats. 75% (9 out of 12) genera, which had a remarkable difference in relative abundance between the control and combined iAs and F- exposure groups, showed significantly strong correlations (r = 0.70-0.90) with the water maze performance indicators. Collectively, these results suggest that concurrent iAs and F- exposure led to more prominent effects on neurodevelopment and gut microbiome composition structures in rats, and the strong correlation between them indicates a high potential for the development of novel microbiome-based biomarkers of iAs and/or F- associated neurodevelopmental deficits.


Assuntos
Arsênio , Arsenicais , Microbioma Gastrointestinal , Animais , Arsênio/toxicidade , Criança , Fluoretos/toxicidade , Humanos , RNA Ribossômico 16S/genética , Ratos
9.
Front Oncol ; 9: 1279, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31824855

RESUMO

The Fry gene, located on chromosome 13, is an evolutionarily conserved large protein from yeast to human. Our previous study genetically linked the Fry gene with differential susceptibility to mammary carcinogenesis, but whether Fry affects mammary gland development and function, as well as the growth of breast cancer cells, is largely unknown. To define the consequences of Fry loss in the mammary glands, we have generated mice conditionally deficient of the Fry gene in the mammary glands using the Cre-loxP recombination system. We examined multiple phenotypes with male and female homozygous Fry conditional knockout mice (Mfry) and control mice (WT), including body weight, preliminary observations (health and neurological flexes), open field locomotion, sensory abilities, auditory threshold, and glucose metabolism. The loss of Fry in the mammary glands didn't cause a significant difference in these genotypes between Mfry and WT mice. However, our data showed that Fry was required during pregnancy, while it was functionally dispensable in virgin mammary gland development. Loss of Fry led to more lateral buds, and the lobuloalveoli were smaller and showed undistended morphology in mammary glands during late pregnancy. in vitro experiment, ectopic expression of FRY could alter the morphology and significantly suppress the growth and proliferation of the breast cancer cell lines, MDA-MB-231 (ER-/PR-/HER2-, Basal-like) and BT474 (ER+/PR+/HER2+, Luminal B). The following genome-wide transcriptomic analysis of these cells suggested that FRY interacted with protein kinases relevant signaling pathways and induced massive changes in gene expression, including the activation of the Hippo/Yap pathway. Together, our data suggest that the FRY is required for mammary glands developments during pregnant periods, and affects breast cancer cell growth and proliferation.

10.
Environ Int ; 119: 250-263, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29982128

RESUMO

As a nonmutagenic human carcinogen, arsenic (As)'s carcinogenic activity is likely the result of epigenetic changes, particularly alterations in DNA methylation. While increasing studies indicate a potentially important role for timing of As exposure on DNA methylation patterns and the subsequent differential risks for As toxicity and carcinogenesis, there is a lack of research that tackles these critical questions, particularly in human based populations. Here we reported a family-based study including three generations, in which each generation living in the same household had a distinctive timing of As exposure: in adulthood, in utero and during early childhood, and in germlines exposure for grandparents, parents, and grandchildren, respectively. We generated genome-wide DNA methylation data for 18 As-exposed families, nine control families, as well as 18 arsenical skin lesion patients. Our analysis showed that As exposure may leave detectable DNA methylation changes even though exposure occurred decades ago, and the most significant changes of global DNA methylation were observed among patients afflicted with arsenical skin lesions. As exposure across generations shared common differentially methylated DNA loci and regions (744 DML and 15 DMRs) despite the distinctive exposure timing in each generation. Importantly, based on these DML, clustering analysis grouped skin lesion patients together with grandparents in exposed families in the same cluster, separated from grandparents in control families. Further analysis identified a number of DML and several molecular pathways that were significantly distinguished between controls, exposed populations, as well as skin lesion patients. Finally, our exploratory analysis suggested that some of these DML altered by As exposure, may have the potential to be inherited affecting not only those directly exposed but also later generations. Together, our results suggest that common DML and/or DMRs associated with an increased risk for disease development could be identified regardless of when exposure to As occurred during their life span, and thus may be able to serve as biomarkers for identifying individuals at risk for As-induced skin lesions and possible cancers.


Assuntos
Intoxicação por Arsênico , Arsênio/toxicidade , Metilação de DNA , Exposição Ambiental , Dermatopatias/induzido quimicamente , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Exposição Ambiental/análise , Exposição Ambiental/estatística & dados numéricos , Família , Feminino , Humanos , Gravidez , Efeitos Tardios da Exposição Pré-Natal
11.
Oncol Rep ; 37(2): 803-812, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27959429

RESUMO

Human NudC nuclear distribution protein (hNUDC) is differentially expressed between normal and cancer cells. Based on its marked altered expression and its roles in modulating cell division, cytokineses and migration, a detailed understanding of the mechanisms regulating hNUDC expression in cancer cells is critical. In this study, we identified miR-194 as a downstream target of hNUDC and linked its expression to reduced metastatic capacity and tumorigenicity of prostate cancer (PCa) cells. Using miRNA target prediction programs, hNUDC mRNA was found to contain a potential binding site for miR-194 within its 3'UTR. A Reporter assay confirmed that post-transcriptional regulation of hNUDC was dependent on the miR-194 binding site. Forced expression of miR-194 in PCa cell lines, PC-3 and DU-145, led to a decrease in the mRNA and protein levels of hNUDC. Overexpression of miR-194 in these cells inhibited cell migration and invasion, and induced multinucleated cells. Our data showed that hNUDC knockdown by siRNA significantly reduced the migration and invasion in the PC-3 and DU-145 cells, phenocopying the results of miR-194 overexpression. Furthermore, lentivirus-mediated stable expression of miR-194 in PCa cells reduced the ability of colony formation as detected by a soft agar assay and exhibited significantly less tumorigenic ability in vivo. Our results suggest a novel role for miR-194 in effectively controlling cell metastatic processes in PCa cells via the regulation of hNUDC expression.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Movimento Celular , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Proteínas Nucleares/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Animais , Apoptose , Western Blotting , Proteínas de Ciclo Celular/genética , Proliferação de Células , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Proteínas Nucleares/genética , Neoplasias da Próstata/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Toxicol Sci ; 155(1): 124-134, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27637898

RESUMO

Chronic arsenic exposure via drinking water has become a worldwide public health concern. In humans, inorganic arsenic (iAs) is metabolized to monomethylarsonic acid (MMA) and dimethylarsinic acid (DMA) mainly mediated by arsenic (+3 oxidation state) methyltransferase (As3MT). We reported recently that N-6 adenine-specific DNA methyltransferase 1 (N6AMT1) was involved in arsenic metabolism, and examined its interactive effect with As3MT on arsenic metabolism in vitro To further evaluate the interactive effect of N6AMT1 and As3MT on arsenic biomethylation in humans, we conducted a human population-based study including 289 subjects living in rural villages in Inner Mongolia, China, and assessed their urinary arsenic metabolites profiles in relation to genetic polymorphisms and haplotypes of N6AMT1 and As3MT Five N6AMT1 single nucleotide polymorphisms (SNPs; rs1003671, rs7282257, rs2065266, rs2738966, rs2248501) and the N6AMT1 haplotype 2_GGCCAT were significantly associated with the percentage of iAs (% iAs) in urine (e.g., for rs7282257, mean was 9.62% for TT, 6.73% for AA). Rs1003671 was also in a significant relationship with urinary MMA and DMA (the mean of %MMA was 24.95% for GA, 31.69% for GG; the mean of % DMA was 69.21% for GA, 59.82% for GG). The combined effect of N6AMT1 haplotype 2_GGCCAT and As3MT haplotype 2_GCAC showed consistence with the additive significance of each haplotype on % iAs: the mean was 5.47% and 9.36% for carriers with both and null haplotypes, respectively. Overall, we showed that N6AMT1 genetic polymorphisms were associated with arsenic biomethylation in the Chinese population, and its interaction with As3MT was observed in specific haplotype combinations.


Assuntos
Arsênio/metabolismo , Variação Genética , Metiltransferases/genética , DNA Metiltransferases Sítio Específica (Adenina-Específica)/genética , Adulto , China , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único
13.
Toxicol Sci ; 146(2): 354-62, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25997655

RESUMO

In humans, arsenic is primarily metabolized by arsenic (+3 oxidation state) methyltransferase (As3MT) to yield both trivalent and pentavalent methylated metabolites. We recently reported that the putative N-6 adenine-specific DNA methyltransferase 1 (N6AMT1) can biotransform monomethylarsonous acid (MMA(III)) to dimethylarsinic acid, conferring resistance of human cells to arsenic exposure. To further decipher the role of N6AMT1 and its interaction with As3MT in arsenic biomethylation, we examined the relative contribution of N6AMT1 and As3MT in metabolizing arsenic using several newly modified UROtsa human urothelial cells, ie, UROtsa cells with either a constant level of N6AMT1 or As3MT in combination with an inducible level of As3MT or N6AMT1, respectively. Our analysis confirmed the involvement of N6AMT1 in MMA(III) biomethylation but not for inorganic arsenic. In a comparable level of N6AMT1 and As3MT, the effect of N6AMT1 mediated MMA(III) biomethylation was obscured by the action of As3MT. Furthermore, we showed that the levels of N6AMT1 and As3MT proteins varied among and within human normal and cancerous tissues. Overall, the data showed that N6AMT1 has a role in MMA(III) biomethylation, but its effect is relatively minor and limited compared with As3MT. In addition, the varied levels and distributions of N6AMT1 and As3MT among human tissues may potentially contribute to the tissue specificity and susceptibility to arsenic toxicity and carcinogenicity.


Assuntos
Arsênio/farmacocinética , Metiltransferases/metabolismo , DNA Metiltransferases Sítio Específica (Adenina-Específica)/metabolismo , Linhagem Celular Transformada , Cromatografia Líquida de Alta Pressão , Humanos , Espectrometria de Massas , Metilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA