Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1145080, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37180167

RESUMO

Background: The role of copper in cancer treatment is multifaceted, with copper homeostasis-related genes associated with both breast cancer prognosis and chemotherapy resistance. Interestingly, both elimination and overload of copper have been reported to have therapeutic potential in cancer treatment. Despite these findings, the exact relationship between copper homeostasis and cancer development remains unclear, and further investigation is needed to clarify this complexity. Methods: The pan-cancer gene expression and immune infiltration analysis were performed using the Cancer Genome Atlas Program (TCGA) dataset. The R software packages were employed to analyze the expression and mutation status of breast cancer samples. After constructing a prognosis model to separate breast cancer samples by LASSO-Cox regression, we examined the immune statement, survival status, drug sensitivity and metabolic characteristics of the high- and low-copper related genes scoring groups. We also studied the expression of the constructed genes using the human protein atlas database and analyzed their related pathways. Finally, copper staining was performed with the clinical sample to investigate the distribution of copper in breast cancer tissue and paracancerous tissue. Results: Pan-cancer analysis showed that copper-related genes are associated with breast cancer, and the immune infiltration profile of breast cancer samples is significantly different from that of other cancers. The essential copper-related genes of LASSO-Cox regression were ATP7B (ATPase Copper Transporting Beta) and DLAT (Dihydrolipoamide S-Acetyltransferase), whose associated genes were enriched in the cell cycle pathway. The low-copper related genes scoring group presented higher levels of immune activation, better probabilities of survival, enrichment in pathways related to pyruvate metabolism and apoptosis, and higher sensitivity to chemotherapy drugs. Immunohistochemistry staining showed high protein expression of ATP7B and DLAT in breast cancer samples. The copper staining showed copper distribution in breast cancer tissue. Conclusion: This study displayed the potential impacts of copper-related genes on the overall survival, immune infiltration, drug sensitivity and metabolic profile of breast cancer, which could predict patients' survival and tumor statement. These findings may serve to support future research efforts aiming at improving the management of breast cancer.


Assuntos
Neoplasias da Mama , ATPases Transportadoras de Cobre , Cobre , Di-Hidrolipoil-Lisina-Resíduo Acetiltransferase , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Humanos , Cobre/análise , Cobre/metabolismo , Perfilação da Expressão Gênica , Análise de Sobrevida , ATPases Transportadoras de Cobre/análise , ATPases Transportadoras de Cobre/genética , Di-Hidrolipoil-Lisina-Resíduo Acetiltransferase/análise , Di-Hidrolipoil-Lisina-Resíduo Acetiltransferase/genética , Di-Hidrolipoil-Lisina-Resíduo Acetiltransferase/metabolismo , Prognóstico , Resistencia a Medicamentos Antineoplásicos , Modelos Biológicos
2.
Commun Biol ; 6(1): 194, 2023 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-36804628

RESUMO

Hepatic ischemia-reperfusion (I/R) injury is a common complication in liver transplantation. The connection between I/R-induced injury response and liver heterogeneity has yet to be fully understood. In this study, we converge histopathological examination with spatial transcriptomics to dissect I/R injury patterns and their associated molecular changes, which reveal that the pericentral zones are most sensitive to I/R injury in terms of histology, transcriptomic changes, and cell type dynamics. Bioinformatic analysis of I/R injury-related pathways predicts that celastrol can protect against liver I/R injury by inducing ischemic pre-conditioning, which is experimentally validated. Mechanistically, celastrol likely implements its protective effect against I/R injury by activating HIF1α signaling and represents a potential strategy for resolving liver I/R.


Assuntos
Traumatismo por Reperfusão , Transcriptoma , Camundongos , Animais , Modelos Animais de Doenças , Fígado/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia
3.
J Virol ; 96(8): e0202821, 2022 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-35389233

RESUMO

BK polyomavirus (PyV) infects the genitourinary tract of >90% of the adult population. Immunosuppression increases the risk of viral reactivation, making BKPyV a leading cause of graft failure in kidney transplant recipients. Polyomaviruses have a small double-stranded DNA (dsDNA) genome that requires host replication machinery to amplify the viral genome. Specifically, polyomaviruses promote S phase entry and delay S phase exit by activating the DNA damage response (DDR) pathway via an uncharacterized mechanism requiring viral replication. BKPyV infection elevates expression of MutSα, a mismatch repair (MMR) pathway protein complex that senses and repairs DNA mismatches and can activate the DDR. Thus, we investigated the role of the MMR pathway by silencing the MutSα component, Msh6, in BKPyV-infected primary cells. This resulted in severe DNA damage that correlated with weak DNA damage response activation and a failure to arrest the cell cycle to prevent mitotic entry during infection. Furthermore, silencing Msh6 expression resulted in significantly fewer infectious viral particles due to significantly lower levels of VP2, a minor capsid protein important for trafficking during subsequent infections. Since viral assembly occurs in the nucleus, our findings are consistent with a model in which entry into mitosis disrupts viral assembly due to nuclear envelope breakdown, which disperses VP2 throughout the cell, reducing its availability for encapsidation into viral particles. Thus, the MMR pathway may be required to activate the ATR (ATM-Rad3-related) pathway during infection to maintain a favorable environment for both viral replication and assembly. IMPORTANCE Since there are no therapeutics that target BKPyV reactivation in organ transplant patients, it is currently treated by decreasing immunosuppression to allow the natural immune system to fight the viral infection. Antivirals would significantly improve patient outcomes since reducing immunosuppression carries the risk of graft failure. PyVs activate the DDR, for which there are several promising inhibitors. However, a better understanding of how PyVs activate the DDR and what role the DDR plays during infection is needed. Here, we show that a component of the mismatch repair pathway is required for DDR activation during PyV infection. These findings show that the mismatch repair pathway is important for DDR activation during PyV infection and that inhibiting the DDR reduces viral titers by generating less infectious virions that lack the minor capsid protein VP2, which is important for viral trafficking.


Assuntos
Vírus BK , Reparo de Erro de Pareamento de DNA , Vírus BK/genética , Proteínas do Capsídeo/genética , Dano ao DNA , Reparo de Erro de Pareamento de DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Infecções por Polyomavirus/virologia , Replicação Viral/genética
4.
Molecules ; 26(22)2021 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-34833997

RESUMO

The soft rot disease caused by Rhizopus stolonifer is an important disease in cherry tomato fruit. In this study, the effect of iturin A on soft rot of cherry tomato and its influence on the storage quality of cherry tomato fruit were investigated. The results showed that 512 µg/mL of iturin A could effectively inhibit the incidence of soft rot of cherry tomato fruit. It was found that iturin A could induce the activity of resistance-related enzymes including phenylalanine ammonia lyase (PAL), polyphenol oxidase (PPO), peroxidase (POD), glucanase (GLU), and chitinase (CHI), and active oxygen-related enzymes including ascorbate peroxidases (APX), superoxide dismutases (SOD), catalases (CAT), and glutathione reductase (GR) of cherry tomato fruit. In addition, iturin A treatment could slow down the weight loss of cherry tomato and soften the fruit. These results indicated that iturin A could retard the decay and improve the quality of cherry tomato fruit by both the inhibition growth of R. stolonifera and the inducing the resistance.


Assuntos
Resistência a Medicamentos/efeitos dos fármacos , Frutas/metabolismo , Peptídeos Cíclicos/farmacologia , Doenças das Plantas/microbiologia , Raízes de Plantas/metabolismo , Solanum lycopersicum/metabolismo , Frutas/microbiologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Solanum lycopersicum/microbiologia , Proteínas de Plantas/biossíntese , Raízes de Plantas/microbiologia , Rhizopus/crescimento & desenvolvimento
5.
Gene ; 757: 144926, 2020 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-32621951

RESUMO

Endometriosis is a frequent gynecologic disease in the world. CircRNAs can exert a crucial role in various diseases. Nevertheless, little is known about its roles in endometriosis. We investigated the involvement of circ_0061140 in endometriosis. Tissues from endometriosis women displayed an increased expression of circ_0061140. Then, we found loss of circ_0061140 significantly repressed ectopic endometrial cell proliferation, migration and invasion. Meanwhile,miR-140-3pcan demonstrate an important role in several cancers.Here, we reported miR-140-3p was reduced in ectopic endometrial cells and it acted as a target of circ_0061140. Moreover, miR-140-3p was able to reverse the effect of circ_0061140 on ectopic endometrial cells. Furthermore, Notch2 was predicted as a putative target of miR-140-3p. A positive correlation between circ_0061140 and Notch2 was indicated. miR-140-3p and Notch2 were operated as downstream effectors in the circ_0061140 mediated signaling in endometriosis. Decrease of circ_0061140 could depress endometriosis progression through modulating miR-140-3p and Notch2.


Assuntos
Movimento Celular , Proliferação de Células , Endometriose/genética , RNA Circular/genética , Células Cultivadas , Regulação para Baixo , Endometriose/metabolismo , Endometriose/patologia , Endométrio/metabolismo , Endométrio/patologia , Feminino , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Circular/metabolismo , Receptor Notch2/genética , Receptor Notch2/metabolismo
6.
Int J Biol Macromol ; 143: 775-784, 2020 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-31726133

RESUMO

In this study, we investigated the structural characterization and antidiabetic effects of Lactobacillus H31 exopolysaccharide (EPS) H31-2 from pickled cabbage. The BLAST result indicated that Lactobacillus H31 was closely related to Lactobacillusplantarum S1S2L2. In the spectrum of Fourier-transform infrared spectroscopy (FT-IR), 3370 cm-1 was the characteristic band of polysaccharide. A structural analysis showed that the monosaccharides of EPS H31-2 included d-mannose (Man) and d-glucose (Glc). The molecular weight of EPS H31-2 was around 10.75 kDa. All these results suggested EPS H31-2 was a novel polysaccharide. The inhibition ratios of crude EPS H31 and the pure fraction H31-2 of pancreatic α-amylase were 89.1 ±â€¯2.59% and 69.2 ±â€¯8.95%, respectively. In addition, the supernatant glucose concentration of insulin-resistant HepG2 cells decreased by treatment with 10-8 M of EPS H31-2, which meant EPS H31-2 could promote the glucose uptake of insulin-resistant HepG2 cell. Furthermore, EPS H31-2 upregulated the expression of the GLUT-4, Akt-2, and AMPK, which play important roles in glycometabolism. These results suggested that Lactobacillus plantarum EPS H31-2 could effectively inhibit the activity of pancreas α-amylase and has potential applications in the prevention and alleviation of diabetes mellitus.


Assuntos
Biomarcadores Tumorais/metabolismo , Brassica/microbiologia , Lactobacillus plantarum/química , Pâncreas/enzimologia , Polissacarídeos Bacterianos/farmacologia , Transdução de Sinais , alfa-Amilases/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/metabolismo , Glicemia/análise , Células Hep G2 , Humanos , Hipoglicemiantes/farmacologia , Metilação , Monossacarídeos/análise , Fosfatidilinositol 3-Quinases/metabolismo , Filogenia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espectroscopia de Prótons por Ressonância Magnética , Transdução de Sinais/efeitos dos fármacos , Espectroscopia de Infravermelho com Transformada de Fourier , alfa-Amilases/metabolismo
7.
mBio ; 10(1)2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30723127

RESUMO

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


Assuntos
Antígenos Virais de Tumores/metabolismo , Citidina Desaminase/biossíntese , Interações Hospedeiro-Patógeno , Antígenos de Histocompatibilidade Menor/biossíntese , Polyomavirus/crescimento & desenvolvimento , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima , Antígenos Virais de Tumores/genética , Sítios de Ligação , Células Cultivadas , Fatores de Transcrição E2F/metabolismo , Perfilação da Expressão Gênica , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neoplasias/patologia , Proteínas de Ligação a Retinoblastoma/metabolismo
8.
Endocrinology ; 159(9): 3365-3377, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30060148

RESUMO

Steroid sulfatase (STS), a desulfating enzyme that converts steroid sulfates to hormonally active steroids, plays an important role in the homeostasis of sex hormones. STS is expressed in the adipose tissue of both male and female mice, but the role of STS in the development and function of adipose tissue remains largely unknown. In this report, we show that the adipose expression of Sts was induced in the high-fat diet (HFD) and ob/ob models of obesity and type 2 diabetes. Transgenic overexpression of the human STS in the adipose tissue of male mice exacerbated the HFD-induced metabolic phenotypes, including increased body weight gain and fat mass, and worsened insulin sensitivity, glucose tolerance, and energy expenditure, which were accounted for by adipocyte hypertrophy, increased adipose inflammation, and dysregulation of adipogenesis. The metabolic harm of the STS transgene appeared to have resulted from increased androgen activity in the adipose tissue, and castration abolished most of the phenotypes. Interestingly, the transgenic effects were sex specific, because the HFD-fed female STS transgenic mice exhibited improved metabolic functions, which were associated with attenuated adipose inflammation. The metabolic benefit of the STS transgene in female mice was accounted for by increased estrogenic activity in the adipose tissue, whereas such benefit was abolished upon ovariectomy. Our results revealed an essential role of the adipose STS in energy homeostasis in sex- and sex hormone-dependent manner. The adipose STS may represent a therapeutic target for the management of obesity and type 2 diabetes.


Assuntos
Tecido Adiposo/metabolismo , Androgênios/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/genética , Estrogênios/metabolismo , Obesidade/metabolismo , Caracteres Sexuais , Esteril-Sulfatase/genética , Adipogenia , Tecido Adiposo/imunologia , Tecido Adiposo/patologia , Animais , Peso Corporal , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Teste de Tolerância a Glucose , Hormônios Esteroides Gonadais/metabolismo , Humanos , Inflamação , Resistência à Insulina , Lipólise , Masculino , Camundongos , Camundongos Obesos , Camundongos Transgênicos , Obesidade/imunologia , Orquiectomia , Ovariectomia , Esteril-Sulfatase/metabolismo , Transcriptoma
9.
Adv Exp Med Biol ; 1043: 455-469, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29224107

RESUMO

Sulfonation and desulfation are two opposing processes that represent an important layer of regulation of estrogenic activity via ligand supplies. Enzymatic activities of families of enzymes, known as sulfotransferases and sulfatases, lead to structural and functional changes of the steroids, thyroids, xenobiotics, and neurotransmitters. Estrogen sulfotransferase (EST) and steroid sulfatase (STS) represent negative and positive regulation of the estrogen activity, respectively. This is because EST-mediated sulfation deactivates estrogens, whereas STS-mediated desulfation converts the inactive estrogen sulfates to active estrogens. In addition to the known functions of estrogens, EST and STS in reproductive processes, regulation of estrogens and other signal molecules especially at the local tissue levels has gained increased attention in the context of metabolic disease in recent years. EST expression is detectable in the subcutaneous adipose tissue in both obese women and men, and the expression of EST is markedly induced in the livers of rodent models of obesity and type 2 diabetes. STS was found to be upregulated in patients with chronic inflammatory liver diseases. Interestingly, the tissue distribution and the transcriptional regulation of EST and STS exhibit obvious sex and species specificity. EST ablation produces completely opposite metabolic phenotype in female and male obese mice. Adipogenesis is also differentially regulated by EST in murine and human adipocytes. This chapter focuses on the recent progress in our understanding of the expression and regulation EST and STS in the context of metabolic homeostasis.


Assuntos
Metabolismo Energético , Estrogênios/metabolismo , Esteril-Sulfatase/metabolismo , Sulfotransferases/metabolismo , Animais , Feminino , Homeostase , Humanos , Masculino , Caracteres Sexuais , Fatores Sexuais , Transdução de Sinais , Especificidade da Espécie
10.
Endocrinology ; 158(11): 4093-4104, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28938414

RESUMO

Estrogen sulfotransferase catalyzes the sulfoconjugation and deactivation of estrogens. Previously, we showed that loss of Est in male ob/ob mice, but not in female ob/ob mice, exacerbated the diabetic phenotype, but the underlying mechanism was unclear. In this study, we show that transgenic reconstitution of Est in the adipose tissue, but not in the liver, attenuated diabetic phenotype in Est-deficient ob/ob mice (obe mice). Mechanistically, adipose reconstitution of Est in obe mice (oae mice) resulted in reduced local and systemic inflammation, improved insulin sensitivity, and increased energy expenditure. At the molecular level, adipose induction of lipocalin-2 (Lcn2) in oae males may have contributed to the inhibition of inflammation because the level of Lcn2 was negatively associated with tumor necrosis factor (Tnf) α expression, and treatment of differentiated adipocytes with Lcn2 antagonized Tnfα-responsive inhibition of insulin signaling. The metabolic benefit of adipose reconstitution of Est was sex specific, because adipose reconstitution of Est in obe females had little effect. Interestingly, despite their improved metabolic functions, obe male mice with reconstituted Est in their adipose tissue failed to ameliorate the impairment of the structure and function of the pancreatic islets. In summary, our study uncovers a crucial adipose- and male-specific role of Est in maintaining the whole-body energy homeostasis.


Assuntos
Metabolismo Energético/genética , Resistência à Insulina/genética , Sulfotransferases/fisiologia , Células 3T3-L1 , Tecido Adiposo/metabolismo , Adiposidade/genética , Animais , Células Cultivadas , Feminino , Homeostase/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Camundongos Transgênicos , Especificidade de Órgãos/genética , Fatores Sexuais
11.
Annu Rev Virol ; 3(1): 517-532, 2016 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-27501263

RESUMO

Mammalian polyomaviruses are characterized by establishing persistent infections in healthy hosts and generally causing clinical disease only in hosts whose immune systems are compromised. Despite the fact that these viruses were discovered decades ago, our knowledge of the mechanisms that govern viral persistence and reactivation is limited. Whereas mouse polyomavirus has been studied in a fair amount of detail, our understanding of the human viruses in particular is mostly inferred from experiments aimed at addressing other questions. In this review, we summarize the state of our current knowledge, draw conclusions when possible, and suggest areas that are in need of further study.


Assuntos
Vírus BK/crescimento & desenvolvimento , Vírus JC/crescimento & desenvolvimento , Infecções por Polyomavirus/virologia , Vírus 40 dos Símios/crescimento & desenvolvimento , Infecções Tumorais por Vírus/virologia , Animais , Vírus BK/genética , Vírus BK/imunologia , DNA Viral/genética , Humanos , Vírus JC/genética , Vírus JC/imunologia , Camundongos , Vírus 40 dos Símios/genética , Vírus 40 dos Símios/imunologia , Replicação Viral/genética
12.
J Virol ; 90(14): 6379-6386, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27147740

RESUMO

UNLABELLED: The APOBEC3 family of DNA cytosine deaminases has important roles in innate immunity and cancer. It is unclear how DNA tumor viruses regulate these enzymes and how these interactions, in turn, impact the integrity of both the viral and cellular genomes. Polyomavirus (PyVs) are small DNA pathogens that contain oncogenic potentials. In this study, we examined the effects of PyV infection on APOBEC3 expression and activity. We demonstrate that APOBEC3B is specifically upregulated by BK polyomavirus (BKPyV) infection in primary kidney cells and that the upregulated enzyme is active. We further show that the BKPyV large T antigen, as well as large T antigens from related polyomaviruses, is alone capable of upregulating APOBEC3B expression and activity. Furthermore, we assessed the impact of A3B on productive BKPyV infection and viral genome evolution. Although the specific knockdown of APOBEC3B has little short-term effect on productive BKPyV infection, our informatics analyses indicate that the preferred target sequences of APOBEC3B are depleted in BKPyV genomes and that this motif underrepresentation is enriched on the nontranscribed stand of the viral genome, which is also the lagging strand during viral DNA replication. Our results suggest that PyV infection upregulates APOBEC3B activity to influence virus sequence composition over longer evolutionary periods. These findings also imply that the increased activity of APOBEC3B may contribute to PyV-mediated tumorigenesis. IMPORTANCE: Polyomaviruses (PyVs) are a group of emerging pathogens that can cause severe diseases, including cancers in immunosuppressed individuals. Here we describe the finding that PyV infection specifically induces the innate immune DNA cytosine deaminase APOBEC3B. The induced APOBEC3B enzyme is fully functional and therefore may exert mutational effects on both viral and host cell DNA. We provide bioinformatic evidence that, consistent with this idea, BK polyomavirus genomes are depleted of APOBEC3B-preferred target motifs and enriched for the corresponding predicted reaction products. These data imply that the interplay between PyV infection and APOBEC proteins may have significant impact on both viral evolution and virus-induced tumorigenesis.


Assuntos
Citidina Desaminase/metabolismo , Regulação da Expressão Gênica , Genoma Viral , Túbulos Renais/enzimologia , Antígenos de Histocompatibilidade Menor/metabolismo , Infecções por Polyomavirus/virologia , Polyomavirus/patogenicidade , Replicação Viral , Células Cultivadas , Citidina Desaminase/antagonistas & inibidores , Citidina Desaminase/genética , Humanos , Túbulos Renais/virologia , Antígenos de Histocompatibilidade Menor/genética , Polyomavirus/genética , Infecções por Polyomavirus/patologia , RNA Interferente Pequeno/genética , Ativação Transcricional , Regulação para Cima
13.
J Hepatol ; 64(1): 44-52, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26220752

RESUMO

BACKGROUND & AIMS: Chronic inflammatory liver diseases are associated with estrogen excess and feminization in men, which is thought to be due to compromised liver function to break down estrogens. The goal of this study is to determine whether the inflammatory induction of steroid sulfatase (STS), which converts inactive estrogen sulfates to active estrogens, may have contributed to the estrogen excess in chronic liver disease. METHODS: We performed bioinformatic analysis, real-time PCR, immunohistochemistry, and UPLC/MS-MS to analyze hepatic STS expression and serum estrogen levels in patients with chronic liver diseases. The crosstalk between NF-κB pathway and STS-regulated estrogen signaling was investigated by electrophoretic mobility shift assay, chromatin immunoprecipitation, luciferase assay and gene knockdown experiments in human hepatocytes. RESULTS: Hepatic STS was induced in patients with chronic inflammatory liver diseases, which was accompanied by increased circulating estrogen levels. The human STS gene, but not the mouse Sts gene, was induced by inflammatory stimuli in hepatic cells. Mechanistically, STS was established as a novel NF-κB target gene, whose induction facilitated the conversion of inactive estrogen sulfates to active estrogens, and consequently attenuated the inflammatory response. In contrast, genetic or pharmacological inhibition of STS or a direct blockade of estrogen signaling sensitized liver cells to the transcriptional activation of NF-κB and inflammatory response, possibly through the inhibition of IκB kinase activation. CONCLUSIONS: Our results suggest a negative feedback loop in chronic inflammatory liver diseases, in which the inflammatory activation of NF-κB induces STS gene expression. The induced STS facilitates the conversion of inactive estrogen sulfates to active estrogens, which in return attenuates the NF-κB-mediated inflammation.


Assuntos
Estrogênios/metabolismo , Homeostase , Inflamação/etiologia , Hepatopatias/metabolismo , Esteril-Sulfatase/fisiologia , Células Cultivadas , Doença Crônica , Biologia Computacional , Humanos , Cirrose Hepática Alcoólica/metabolismo , NF-kappa B/fisiologia , Transdução de Sinais
14.
J Proteome Res ; 14(10): 4413-24, 2015 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-26354146

RESUMO

Polyomaviruses are a family of small DNA viruses that are associated with a number of severe human diseases, particularly in immunocompromised individuals. The detailed virus-host interactions during lytic polyomavirus infection are not fully understood. Here, we report the first nuclear proteomic study with BK polyomavirus (BKPyV) in a primary renal proximal tubule epithelial cell culture system using stable isotope labeling by amino acids in cell culture (SILAC) proteomic profiling coupled with liquid chromatography-tandem mass spectrometry. We demonstrated the feasibility of SILAC labeling in these primary cells and subsequently performed reciprocal labeling-infection experiments to identify proteins that are altered by BKPyV infection. Our analyses revealed specific proteins that are significantly up- or down-regulated in the infected nuclear proteome. The genes encoding many of these proteins were not identified in a previous microarray study, suggesting that differential regulation of these proteins may be independent of transcriptional control. Western blotting experiments verified the SILAC proteomic findings. Finally, pathway and network analyses indicated that the host cell DNA damage response signaling and DNA repair pathways are among the cellular processes most affected at the protein level during polyomavirus infection. Our study provides a comprehensive view of the host nuclear proteomic changes during polyomavirus lytic infection and suggests potential novel host factors required for a productive polyomavirus infection.


Assuntos
Vírus BK/fisiologia , Núcleo Celular/metabolismo , Reparo do DNA , Células Epiteliais/metabolismo , Proteoma/metabolismo , Núcleo Celular/química , Núcleo Celular/patologia , Núcleo Celular/virologia , Cromatografia Líquida , Dano ao DNA , Células Epiteliais/patologia , Células Epiteliais/virologia , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Interações Hospedeiro-Patógeno , Humanos , Marcação por Isótopo , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Túbulos Renais Proximais/virologia , Anotação de Sequência Molecular , Cultura Primária de Células , Proteoma/genética , Proteoma/isolamento & purificação , Transdução de Sinais , Espectrometria de Massas em Tandem , Transcrição Gênica
15.
Nat Commun ; 6: 7979, 2015 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-26259151

RESUMO

Sepsis is the host's deleterious systemic inflammatory response to microbial infections. Here we report an essential role for the oestrogen sulfotransferase (EST or SULT1E1), a conjugating enzyme that sulfonates and deactivates estrogens, in sepsis response. Both the caecal ligation and puncture (CLP) and lipopolysaccharide models of sepsis induce the expression of EST and compromise the activity of oestrogen, an anti-inflammatory hormone. Surprisingly, EST ablation sensitizes mice to sepsis-induced death. Mechanistically, EST ablation attenuates sepsis-induced inflammatory responses due to compromised oestrogen deactivation, leading to increased sepsis lethality. In contrast, transgenic overexpression of EST promotes oestrogen deactivation and sensitizes mice to CLP-induced inflammatory response. The induction of EST by sepsis is NF-κB dependent and EST is a NF-κB-target gene. The reciprocal regulation of inflammation and EST may represent a yet-to-be-explored mechanism of endocrine regulation of inflammation, which has an impact on the clinical outcome of sepsis.


Assuntos
Sepse/genética , Sulfotransferases/metabolismo , Animais , Ceco/patologia , Estrogênios/sangue , Estrogênios/metabolismo , Estrogênios/farmacologia , Feminino , Predisposição Genética para Doença , Células Hep G2 , Humanos , Células de Kupffer/efeitos dos fármacos , Células de Kupffer/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Knockout , Sulfotransferases/genética , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Útero/efeitos dos fármacos
16.
J Hepatol ; 63(4): 855-62, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26070408

RESUMO

BACKGROUND & AIMS: Fatty acid binding protein 4 (FABP4) has been known as a mediator of inflammatory response in the macrophages and adipose tissue, but its hepatic function is poorly understood. The goal of this study is to investigate the role of FABP4 in liver ischemia/reperfusion (I/R), a clinical condition that involves both hypoxia and inflammation. METHODS: To examine the I/R regulation of FABP4, mice were subjected to I/R surgery before being measured for FABP4 gene expression. Both loss-of-function (by using a pharmacological FABP4 inhibitor) and gain-of-function (by adenoviral overexpression of FABP4) were used to determine the functional relevance of FABP4 expression and its regulation during I/R. To determine the hypoxia responsive regulation of FABP4, primary mouse hepatocytes were exposed to hypoxia. The FABP4 gene promoter was cloned and its regulation by hypoxia inducible factor 1α (HIF-1α) was characterized by luciferase reporter gene, electrophoretic mobility shift, and chromatin immunoprecipitation assays. RESULTS: We found that the hepatic expression of FABP4 was markedly induced by I/R. At the functional level, pharmacological inhibition of FABP4 alleviated the I/R injury, whereas adenoviral overexpression of FABP4 sensitized mice to I/R injury. We also showed that exposure of primary hepatocytes to hypoxia or transgenic overexpression of HIF-1α in the mouse liver was sufficient to induce the expression of FABP4. Our promoter analysis established FABP4 as a novel transcriptional target of HIF-1α. CONCLUSIONS: FABP4 is a hypoxia inducible gene that sensitizes mice to liver I/R injury. FABP4 may represent a novel therapeutic target, and FABP4 inhibitors may be used as therapeutic agents to manage hepatic I/R injury.


Assuntos
DNA/genética , Proteínas de Ligação a Ácido Graxo/genética , Regulação da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Isquemia/genética , Fígado/irrigação sanguínea , Traumatismo por Reperfusão/genética , Animais , Western Blotting , Hipóxia Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Proteínas de Ligação a Ácido Graxo/biossíntese , Feminino , Hepatócitos/metabolismo , Hepatócitos/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Isquemia/etiologia , Isquemia/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/metabolismo
17.
J Biol Chem ; 290(23): 14754-64, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-25922074

RESUMO

Estrogen sulfotransferase (EST) regulates estrogen homeostasis by sulfonating and deactivating estrogens. Liver ischemia and reperfusion (I/R) involves both hypoxia during the ischemic phase and oxidative damage during the reperfusion phase. In this report, we showed that the expression of EST was markedly induced by I/R. Mechanistically, oxidative stress-induced activation of Nrf2 was responsible for the EST induction, which was abolished in Nrf2(-/-) mice. EST is a direct transcriptional target of Nrf2. In female mice, the I/R-responsive induction of EST compromised estrogen activity. EST ablation attenuated I/R injury as a result of decreased estrogen deprivation, whereas this benefit was abolished upon ovariectomy. The effect of EST ablation was sex-specific because the EST(-/-) males showed heightened I/R injury. Reciprocally, both estrogens and EST regulate the expression and activity of Nrf2. Estrogen deprivation by ovariectomy abolished the I/R-responsive Nrf2 accumulation, whereas the compromised estrogen deprivation in EST(-/-) mice was associated with increased Nrf2 accumulation. Our results suggested a novel I/R-responsive feedback mechanism to limit the activity of Nrf2 in which Nrf2 induces the expression of EST, which subsequently increases estrogen deactivation and limits the estrogen-responsive activation of Nrf2. Inhibition of EST, at least in females, may represent an effective approach to manage hepatic I/R injury.


Assuntos
Fígado/patologia , Estresse Oxidativo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Sulfotransferases/genética , Animais , Células Cultivadas , Estrogênios/metabolismo , Feminino , Deleção de Genes , Células Hep G2 , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Traumatismo por Reperfusão/metabolismo , Fatores Sexuais , Sulfotransferases/metabolismo , Regulação para Cima
18.
Virol Sin ; 30(2): 122-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25910481

RESUMO

Viruses are obligate intracellular parasites that subvert cellular metabolism and pathways to mediate their own replication-normally at the expense of the host cell. Polyomaviruses are a group of small DNA viruses, which have long been studied as a model for eukaryotic DNA replication. Polyomaviruses manipulate host replication proteins, as well as proteins involved in DNA maintenance and repair, to serve as essential cofactors for productive infection. Moreover, evidence suggests that polyomavirus infection poses a unique genotoxic threat to the host cell. In response to any source of DNA damage, cells must initiate an effective DNA damage response (DDR) to maintain genomic integrity, wherein two protein kinases, ataxia telangiectasia mutated (ATM) and ATM- and Rad3-related (ATR), are major regulators of DNA damage recognition and repair. Recent investigation suggests that these essential DDR proteins are required for productive polyomavirus infection. This review will focus on polyomaviruses and their interaction with ATM- and ATR-mediated DNA damage responses and the effect of this interaction on host genomic stability.


Assuntos
Dano ao DNA , Reparo do DNA , Instabilidade Genômica , Interações Hospedeiro-Patógeno , Polyomavirus/fisiologia , Replicação Viral , Humanos
19.
J Virol ; 89(9): 5032-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25694603

RESUMO

UNLABELLED: BK polyomavirus (BKPyV) reactivation is associated with severe human disease in kidney and bone marrow transplant patients. The interplay between viral and host factors that regulates the productive infection process remains poorly understood. We have previously reported that the cellular DNA damage response (DDR) is activated upon lytic BKPyV infection and that its activation is required for optimal viral replication in primary kidney epithelial cells. In this report, we set out to determine what viral components are responsible for activating the two major phosphatidylinositol 3-kinase-like kinases (PI3KKs) involved in the DDR: ataxia telangiectasia mutated (ATM) kinase and ATM and Rad3-related (ATR) kinase. Using a combination of UV treatment, lentivirus transduction, and mutant virus infection experiments, our results demonstrate that neither the input virus nor the expression of large T antigen (TAg) alone is sufficient to trigger the activation of ATM or ATR in our primary culture model. Instead, our data suggest that the activation of both the ATM- and ATR-mediated DDR pathways is linked to viral DNA replication. Intriguingly, a TAg mutant virus that is unable to activate the DDR causes substantial host DNA damage. Our study provides insight into how DDRs are activated by polyomaviruses in primary cells with intact cell cycle checkpoints and how the activation might be linked to the maintenance of host genome stability. IMPORTANCE: Polyomaviruses are opportunistic pathogens that are associated with several human diseases under immunosuppressed conditions. BK polyomavirus (BKPyV) affects mostly kidney and bone marrow transplant patients. The detailed replication mechanism of these viruses remains to be determined. We have previously reported that BKPyV activates the host DNA damage response (DDR), a response normally used by the host cell to combat genotoxic stress, to aid its own replication. In this study, we identified that the trigger for DDR activation is viral replication. Furthermore, we show that the virus is able to cause host DNA damage in the absence of viral replication and DDR activation. These results suggest an intricate relationship between viral replication, DDR activation, and host genome instability.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Vírus BK/fisiologia , Dano ao DNA , Reparo do DNA , Replicação Viral , Células Cultivadas , Células Epiteliais/virologia , Humanos , Mutação , Transdução Genética , Raios Ultravioleta
20.
J Virol ; 89(4): 1948-50, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25473048

RESUMO

Understanding the life cycle and pathogenesis of animal viruses requires that we have systems in which the viruses can replicate and cause disease. For the latter, we rely upon animal models or information that we can obtain from studying natural infections of humans and other animals. For the former, however, we are largely dependent on the availability of cell culture systems in which viruses can be propagated to investigate the molecular mechanisms of viral replication. For many years, it was assumed that replication in culture provided an accurate description of the life cycle of the organism. In this Gem, we will discuss two viruses, polyomavirus and cytomegalovirus, in which cell culture systems have accidentally provided unique potential insights into viral replication and persistence in their hosts.


Assuntos
Citomegalovirus/fisiologia , Rearranjo Gênico , Genoma Viral , Polyomavirus/fisiologia , Replicação Viral , Animais , Citomegalovirus/genética , DNA Viral/genética , Humanos , Polyomavirus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA