Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Nutr ; 148(3): 389-400, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29546304

RESUMEN

Background: Epidemiologic and in vitro studies suggest independent linkages between poor folate and/or vitamin B-12 nutrition, genomic human papillomavirus (HPV) type 16 viral integration, and cancer. However, there is no direct evidence in vivo to support the causative role of poor folate nutrition in HPV16 integration into the cellular genome. Objective: We tested the hypothesis that folate deficiency enables the integration of HPV16 into the genome of HPV16-harboring keratinocytes, and could thereby influence earlier transformation of these cells to cancer in an animal model. Methods: HPV16-harboring human keratinocytes [(HPV16)BC-1-Ep/SL] were differentiated into 3-dimensional HPV16-organotypic rafts under either folate-replete or folate-deficient conditions in vitro. These were then subcutaneously implanted in severely immunocompromised female Beige Nude XID (Hsd: NIHS-LystbgFoxn1nuBtkxid) mice (4-6 wk old, 16-18 g) fed either a folate-replete diet (1200 nmol folate/kg diet) or a progressively folate-deficient diet (600 or 400 nmol folate/kg diet) for 2 mo prior to raft-implantation surgery, and indefinitely thereafter. The tumors that subsequently developed were characterized for onset, pattern of growth, morphology, HPV16 oncogene expression, and HPV16-genomic integration. Results: All HPV16-organotypic rafts developed in either folate-replete or physiologic low-folate media in vitro and subsequently implanted in folate-replete mice eventually transformed into aggressive malignancies within weeks. When compared to HPV16-high folate-organotypic raft-derived tumors from mice fed either a 1200 or 600 nmol folate/kg diet, those raft-derived cancers that developed in mice fed a 400 nmol folate/kg diet expressed significantly more HPV16 E6 (1.8-fold more) and E7 (2.8-fold more) oncogenic proteins (P = 0.001), and revealed significantly more HPV16-integration sites in genomic DNA (2-fold more), either directly into, or in the vicinity of, cellular genes (P < 0.05). Conclusions: This unprecedented animal model for the consistent rapid transformation of differentiated (HPV16)BC-1-Ep/SL-derived organotypic raft-keratinocytes to cancer in Beige Nude XID mice confirms that dietary folate deficiency can profoundly influence and modulate events leading to HPV16-induced carcinogenesis, and facilitates genomic integration of HPV16 DNA in vivo.


Asunto(s)
Carcinogénesis/genética , Deficiencia de Ácido Fólico/complicaciones , Ácido Fólico/administración & dosificación , Genoma , Papillomavirus Humano 16/genética , Neoplasias/etiología , Integración Viral , Animales , ADN , Modelos Animales de Enfermedad , Femenino , Humanos , Queratinocitos/virología , Ratones Desnudos , Neoplasias/genética , Neoplasias/virología , Estado Nutricional , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/etiología , Infecciones por Papillomavirus/virología , Proteínas Represoras/metabolismo , Neoplasias del Cuello Uterino
2.
J Nutr ; 147(4): 482-498, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28250194

RESUMEN

Background: Previously, we determined that heterogeneous nuclear ribonucleoprotein E1 (hnRNP-E1) functions as an intracellular physiologic sensor of folate deficiency. In this model, l-homocysteine, which accumulates intracellularly in proportion to the extent of folate deficiency, covalently binds to and thereby activates homocysteinylated hnRNP-E1 to interact with folate receptor-α mRNA; this high-affinity interaction triggers the translational upregulation of cell surface folate receptors, which enables cells to optimize folate uptake from the external milieu. However, integral to this model is the need for ongoing generation of hnRNP-E1 to replenish homocysteinylated hnRNP-E1 that is degraded.Objective: We searched for an interrelated physiologic mechanism that could also maintain the steady-state concentration of hnRNP-E1 during prolonged folate deficiency.Methods: A novel RNA-protein interaction was functionally characterized by using molecular and biochemical approaches in vitro and in vivo.Results: l-homocysteine triggered a dose-dependent high-affinity interaction between hnRNP-E1 and a 25-nucleotide cis element within the 5'-untranslated region of hnRNP-E1 mRNA; this led to a proportionate increase in these RNA-protein complexes, and translation of hnRNP-E1 both in vitro and within placental cells. Targeted perturbation of this RNA-protein interaction either by specific 25-nucleotide antisense oligonucleotides or mutation within this cis element or by small interfering RNA to hnRNP-E1 mRNA significantly reduced cellular biosynthesis of hnRNP-E1. Conversely, transfection of hnRNP-E1 mutant proteins that mimicked homocysteinylated hnRNP-E1 stimulated both cellular hnRNP-E1 and folate receptor biosynthesis. In addition, ferrous sulfate heptahydrate [iron(II)], which also binds hnRNP-E1, significantly perturbed this l-homocysteine-triggered RNA-protein interaction in a dose-dependent manner. Finally, folate deficiency induced dual upregulation of hnRNP-E1 and folate receptors in cultured human cells and tumor xenografts, and more selectively in various fetal tissues of folate-deficient dams.Conclusions: This novel positive feedback loop amplifies hnRNP-E1 during prolonged folate deficiency and thereby maximizes upregulation of folate receptors in order to restore folate homeostasis toward normalcy in placental cells. It will also functionally impact several other mRNAs of the nutrition-sensitive, folate-responsive posttranscriptional RNA operon that is orchestrated by homocysteinylated hnRNP-E1.


Asunto(s)
Receptor 2 de Folato/metabolismo , Deficiencia de Ácido Fólico/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Placenta/citología , Regulación hacia Arriba/efectos de los fármacos , Animales , Línea Celular , Proteínas de Unión al ADN , Femenino , Receptor 2 de Folato/genética , Ácido Fólico/farmacología , Ribonucleoproteínas Nucleares Heterogéneas/genética , Humanos , Ratones , Ratones Desnudos , Neoplasias Experimentales/metabolismo , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN , Neoplasias del Cuello Uterino/metabolismo
3.
J Biol Chem ; 287(15): 12559-77, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22351779

RESUMEN

Although HPV16 transforms infected epithelial tissues to cancer in the presence of several co-factors, there is insufficient molecular evidence that poor nutrition has any such role. Because physiological folate deficiency led to the intracellular homocysteinylation of heterogeneous nuclear ribonucleoprotein E1 (hnRNP-E1) and activated a nutrition-sensitive (homocysteine-responsive) posttranscriptional RNA operon that included interaction with HPV16 L2 mRNA, we investigated the functional consequences of folate deficiency on HPV16 in immortalized HPV16-harboring human (BC-1-Ep/SL) keratinocytes and HPV16-organotypic rafts. Although homocysteinylated hnRNP-E1 interacted with HPV16 L2 mRNA cis-element, it also specifically bound another HPV16 57-nucleotide poly(U)-rich cis-element in the early polyadenylation element (upstream of L2L1 genes) with greater affinity. Together, these interactions led to a profound reduction of both L1 and L2 mRNA and proteins without effects on HPV16 E6 and E7 in vitro, and in cultured keratinocyte monolayers and HPV16-low folate-organotypic rafts developed in physiological low folate medium. In addition, HPV16-low folate-organotypic rafts contained fewer HPV16 viral particles, a similar HPV16 DNA viral load, and a much greater extent of integration of HPV16 DNA into genomic DNA when compared with HPV16-high folate-organotypic rafts. Subcutaneous implantation of 18-day old HPV16-low folate-organotypic rafts into folate-replete immunodeficient mice transformed this benign keratinocyte-derived raft tissue into an aggressive HPV16-induced cancer within 12 weeks. Collectively, these studies establish a likely molecular linkage between poor folate nutrition and HPV16 and predict that nutritional folate and/or vitamin-B(12) deficiency, which are both common worldwide, will alter the natural history of HPV16 infections and also warrant serious consideration as reversible co-factors in oncogenic transformation of HPV16-infected tissues to cancer.


Asunto(s)
Transformación Celular Viral , Deficiencia de Ácido Fólico , Papillomavirus Humano 16/fisiología , Queratinocitos/virología , Neoplasias Experimentales/virología , Infecciones por Papillomavirus/virología , Animales , Secuencia de Bases , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Catalasa/biosíntesis , Catalasa/genética , Técnicas de Cultivo de Célula , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN , Femenino , Ácido Fólico/metabolismo , Genes Reporteros , Ribonucleoproteínas Nucleares Heterogéneas/química , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Homocisteína/química , Homocisteína/metabolismo , Interacciones Huésped-Patógeno , Papillomavirus Humano 16/genética , Humanos , Queratinocitos/metabolismo , Queratinocitos/trasplante , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Neoplasias Experimentales/patología , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Unión Proteica , Proteolisis , ARN Mensajero/biosíntesis , ARN Mensajero/química , ARN Viral/química , ARN Viral/metabolismo , Proteínas de Unión al ARN , Compuestos de Sulfhidrilo/metabolismo , Carga Tumoral , Integración Viral
4.
Diabetes Metab Syndr Obes ; 16: 3641-3654, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38028988

RESUMEN

Introduction: Hypoglycemia is an acute complication that can appear in people with type 2 diabetes treated with drugs which are associated with a risk of hypoglycemia, and it may lead to individual's fear of hypoglycemia (FoH). FoH adversely affects diabetic self-management, psychological well-being, and quality of life, and it is associated with sleep quality. Nevertheless, this problem is often underestimated. The purpose of this study was to explore the heterogeneous characteristics of FoH in middle-aged and elderly hospitalized patients with type 2 diabetes and assess its relationship with the sleeping quality. Patients and Methods: A cross-sectional study was performed on 263 middle-aged and elderly patients with type 2 diabetes hospitalized at the Zhejiang Veteran Hospital in Jiaxing, China, from May to August 2022, selected by a convenient sampling method. A questionnaire containing general information, fear of hypoglycemia scale-15 (FH-15) and the Pittsburgh sleep quality index (PSQI) was provided to the participants. Latent profile analysis was performed to examine the potential latent groups in the distribution of answers on the individual FoH items. Results: The results identified three latent classes: Class 1 - Low FoH group (33%); Class 2 - No FoH group (61%); Class 3 - High FoH group (6%). The latent FoH class was the influencing factor of sleep quality, which was better in the No FoH group than in the Low FoH group, while the sleep quality of the Low FoH group was better than that in the High FoH group. Conclusion: A heterogeneity was found in the FoH of middle-aged and elderly hospitalized patients with type 2 diabetes. Health-care providers should pay more attention to these patients with high FoH and develop intervention strategies to mitigate it and improve their sleep quality.

5.
J Biol Chem ; 286(45): 39100-15, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-21930702

RESUMEN

The mechanism underlying the sensing of varying degrees of physiological folate deficiency, prior to adaptive optimization of cellular folate uptake through the translational up-regulation of folate receptors (FR) is unclear. Because homocysteine, which accumulates intracellularly during folate deficiency, stimulated interactions between heterogeneous nuclear ribonucleoprotein E1 (hnRNP-E1) and an 18-base FR-α mRNA cis-element that led to increased FR biosynthesis and net up-regulation of FR at cell surfaces, hnRNP-E1 was a plausible candidate sensor of folate deficiency. Accordingly, using purified components, we evaluated the physiological basis whereby L-homocysteine triggered these RNA-protein interactions to stimulate FR biosynthesis. L-homocysteine induced a concentration-dependent increase in RNA-protein binding affinity throughout the range of physiological folate deficiency, which correlated with a proportionate increase in translation of FR in vitro and in cultured human cells. Targeted reduction of newly synthesized hnRNP-E1 proteins by siRNA to hnRNP-E1 mRNA reduced both constitutive and L-homocysteine-induced rates of FR biosynthesis. Furthermore, L-homocysteine covalently bound hnRNP-E1 via multiple protein-cysteine-S-S-homocysteine mixed disulfide bonds within K-homology domains known to interact with mRNA. These data suggest that a concentration-dependent, sequential disruption of critical cysteine-S-S-cysteine bonds by covalently bound L-homocysteine progressively unmasks an underlying RNA-binding pocket in hnRNP-E1 to optimize interaction with FR-α mRNA cis-element preparatory to FR up-regulation. Collectively, such data incriminate hnRNP-E1 as a physiologically relevant, sensitive, cellular sensor of folate deficiency. Because diverse mammalian and viral mRNAs also interact with this RNA-binding domain with functional consequences to their protein expression, homocysteinylated hnRNP-E1 also appears well positioned to orchestrate a novel, nutrition-sensitive (homocysteine-responsive), posttranscriptional RNA operon in folate-deficient cells.


Asunto(s)
Receptor 1 de Folato/biosíntesis , Deficiencia de Ácido Fólico/metabolismo , Ácido Fólico/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Homocisteína/metabolismo , Unión Proteica , Proteínas de Unión al ADN , Disulfuros/metabolismo , Receptor 1 de Folato/genética , Deficiencia de Ácido Fólico/genética , Células HeLa , Ribonucleoproteínas Nucleares Heterogéneas/genética , Homocisteína/genética , Humanos , Estructura Terciaria de Proteína , ARN Mensajero , Proteínas de Unión al ARN , Regulación hacia Arriba
6.
J Integr Med ; 19(5): 460-466, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34154967

RESUMEN

There is currently no drug or therapy that can cure the coronavirus disease 2019 (COVID-19), which is highly contagious and can be life-threatening in severe cases. Therefore, seeking potential effective therapies is an urgent task. An older female at the Leishenshan Hospital in Wuhan, China, with a severe case of COVID-19 with significant shortness of breath and decrease in peripheral oxygen saturation (SpO2), was treated using manual acupuncture and Chinese herbal medicine granule formula Fuzheng Rescue Lung with Xuebijing Injection in addition to standard care. The patient's breath rate, SpO2, heart rate, ratio of neutrophil/lymphocyte (NLR), ratio of monocyte/lymphocyte (MLR), C-reactive protein (CRP), and chest computed tomography were monitored. Acupuncture significantly improved the patient's breathing function, increased SpO2, and decreased her heart rate. Chinese herbal medicine might make the effect of acupuncture more stable; the use of herbal medicine also seemed to accelerate the absorption of lung infection lesions when its dosage was increased. The combination of acupuncture and herbs decreased NLR from 14.14 to 5.83, MLR from 1.15 to 0.33 and CRP from 15.25 to 6.01 mg/L. These results indicate that acupuncture and Chinese herbal medicine, as adjuvants to standard care, might achieve better results in treating severe cases of COVID-19.


Asunto(s)
Terapia por Acupuntura , COVID-19 , Medicamentos Herbarios Chinos , COVID-19/terapia , Femenino , Humanos , Resultado del Tratamiento
7.
Biochem J ; 377(Pt 2): 317-26, 2004 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-14516279

RESUMEN

NMNAT (nicotinamide 5'-mononucleotide adenylyltransferase; EC 2.7.7.1) catalyses the transfer of the adenylyl group from ATP to NMN to form NAD. We have cloned a novel human NMNAT cDNA, designated hNMNAT-2, from human brain. The cDNA contains a 924 bp open reading frame that encodes a 307 amino acid peptide that was expressed as a histidine-patch-containing thioredoxin fusion protein. Expressed hNMNAT-2 shared only 35% amino acid sequence homology with the human NMNAT enzyme (hNMNAT-1), but possessed enzymic activity comparable with hNMNAT-1. Using human genomic databases, hNMNAT-2 was localized to chromosome 1q25 within a 171 kb gene, whereas hNMNAT-1 is on chromosome 1p32-35. Northern blot analysis revealed highly restricted expression of hNMNAT-2 to brain, heart and muscle tissues, which contrasts with the wide tissue expression of hNMNAT-1; different regions of the brain exhibited differential expression of hNMNAT-2. Substitution mutations of either of two invariant residues, His-24 or Trp-92, abolished enzyme activity. Anti-peptide antibody to a unique epitope within hNMNAT-2 was produced, and immunohistochemical analysis of sections of normal adult human pancreas revealed that hNMNAT-2 protein was markedly expressed in the islets of Langerhans. However, the pancreatic exocrine cells exhibited weak expression of hNMNAT-2 protein. Sections of pancreas from insulinoma patients showed strong expression of hNMNAT-2 protein in the insulin-producing tumour cells, whereas acinar cells exhibited relatively low expression of hNMNAT-2 protein. These data suggest that the unique tissue-expression patterns of hNMNAT-2 reflect distinct functions for the isoforms in the regulation of NAD metabolism.


Asunto(s)
Encéfalo/enzimología , Nicotinamida-Nucleótido Adenililtransferasa/genética , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo , Páncreas/enzimología , Secuencia de Aminoácidos , Northern Blotting , Línea Celular Tumoral , Clonación Molecular , Humanos , Inmunohistoquímica , Insulinoma/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Filogenia , ARN Mensajero/metabolismo , Alineación de Secuencia , Distribución Tisular
8.
Med Chem ; 7(6): 718-26, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22313312

RESUMEN

Nicotinamide adenine dinucleotide (NAD+) is synthesized by the action of nicotinamide mononucleotide adenylyltransferase (NMNAT) from NMN and ATP. The mouse homolog of NMNAT-2 (mmNMNAT-2) was cloned, expressed, and subsequently identified using MALDI-TOF in conjunction with the ProFound database. Circular dichroism analyses of recombinant mmNMNAT-2 showed α helical and ß sheet secondary structures, consistent with the known structure of the human isoform. Competition experiments using mouse pancreatic tissue lysates with recombinant mmNMNAT-2 demonstrated that the activity of the expressed protein was similar to the human isoform. Immunohistochemistry of mouse embryonic tissues with hNMNAT-2 also showed a tissue- and cellular-specific expression of this isoform. Therefore, our studies demonstrate for the first time the clear biological evidence for the existence of a mouse isoform of hNMNAT-2. These studies may help in future investigations aimed at understanding the regulation of this gene and its pathway, and in turn, will spur the development of novel therapies for diseases such as cancer and diabetes since mice are the most frequently used experimental system for in vivo studies.


Asunto(s)
Clonación Molecular , Nicotinamida-Nucleótido Adenililtransferasa/genética , Animales , Dicroismo Circular , Humanos , Inmunohistoquímica , Ratones , NAD/biosíntesis , NAD/química , Nicotinamida-Nucleótido Adenililtransferasa/química , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
9.
Artículo en Inglés | MEDLINE | ID: mdl-15641086

RESUMEN

BACKGROUND: Homocysteine, which increases in folate deficiency, can upregulate folate receptors (FR) at the translational level by increasing the interaction between a short cis-element in the 5'-untranslated region of FR-alpha mRNA and heterogeneous nuclear ribonucleoprotein-E1 (hnRNP-E1). Perturbation of this RNA-protein interaction on GD8.5 induces neural tube defects and neurocristopathies in mice. FR upregulation can also reduce cell proliferation independently of folate deficiency in some human cells. Accordingly, we tested the hypothesis that sustained murine maternal folate deficiency would negatively impact pregnancy outcomes, upregulate FR, and selectively reduce fetal cell proliferation. METHODS: Dams were fed chow with various levels of folic acid added for eight weeks before and throughout pregnancy. Following sacrifice on GD17, dams were compared for folate and homocysteine status as well as pregnancy outcomes. Fetuses from some groups were evaluated by specific biochemical, molecular, and immunohistochemical studies for FR, hnRNP-E1, and apoptosis. RESULTS: When compared to dams fed a folate-replete diet, those dams on a folate-depleted diet developed reduced red cell folates and hyperhomocysteinemia and an inverse dose-dependent upregulation of FR and hnRNP-E1 on GD17 without alterations in cell number in the majority of tissues. However, FR overexpression was accompanied by a significant reduction in the net number of cells in the midgut, lung, pons, tongue, and olfactory epithelium, and with premature differentiation in dorsal root ganglion cells and dysplasia of taste buds. By contrast, in the brain, spinal cord, diaphragm, and primordium of follicles of vibrissae, there was less FR expression, which accompanied a net reduction in number of cells and architectural anomalies. Subtle "immunohistochemical footprints" of apoptosis on GD17 fetuses corresponded with net cell loss in the lung and olfactory epithelium. Upregulation of FR could be explained by a homocysteine-induced RNA-protein interaction in folate-depleted fetuses that led to a proportionate increase in murine FR biosynthesis. CONCLUSIONS: Maternal folate deficiency results in selective upregulation of FR and hnRNP-E1 associated with multiple aberrations in fetal tissues that include increased cell loss, architectural anomalies, and premature differentiation. The potential significance of these findings to explain the wide spectrum of folate-responsive birth defects in humans is discussed.


Asunto(s)
Proteínas Portadoras/metabolismo , Feto/patología , Ácido Fólico/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Diferenciación Celular/fisiología , Electroforesis en Gel de Poliacrilamida , Ensayo de Cambio de Movilidad Electroforética , Femenino , Receptores de Folato Anclados a GPI , Inmunohistoquímica , Hígado/metabolismo , Ratones , Embarazo , Regulación hacia Arriba , Vitamina B 12/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA