Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nucleic Acids Res ; 51(21): e108, 2023 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-37870443

RESUMEN

DNA methylation is essential for a wide variety of biological processes, yet the development of a highly efficient and robust technology remains a challenge for routine single-cell analysis. We developed a multiplex scalable single-cell reduced representation bisulfite sequencing (msRRBS) technology. It allows cell-specific barcoded DNA fragments of individual cells to be pooled before bisulfite conversion, free of enzymatic modification or physical capture of the DNA ends, and achieves read mapping rates of 62.5 ± 3.9%, covering 60.0 ± 1.4% of CpG islands and 71.6 ± 1.6% of promoters in K562 cells. Its reproducibility is shown in duplicates of bulk cells with close to perfect correlation (R = 0.97-0.99). At a low 1 Mb of clean reads, msRRBS provides highly consistent coverage of CpG islands and promoters, outperforming the conventional methods with orders of magnitude reduction in cost. Here, we use this method to characterize the distinct methylation patterns and cellular heterogeneity of six cell lines, plus leukemia and hepatocellular carcinoma models. Taking 4 h of hands-on time, msRRBS offers a unique, highly efficient approach for dissecting methylation heterogeneity in a variety of multicellular systems.


Asunto(s)
Metilación de ADN , ADN , Humanos , Islas de CpG/genética , Metilación de ADN/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Células K562 , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN/métodos , Línea Celular Tumoral
2.
Anim Biotechnol ; 34(8): 3808-3818, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37435839

RESUMEN

In the dairy industry, glucose (Glu) is used as bioactive substance to increase milk yield. However, the molecular regulation underneath needs further clarification. Here, the regulation and its molecular mechanism of Glu on cell growth and casein synthesis of dairy cow mammary epithelial cells (DCMECs) were investigated. When Glu was added from DCMECs, both cell growth, ß-casein expression and the mechanistic target of rapamycin complex 1 (mTORC1) pathway were increased. Overexpression and silencing of mTOR revealed that Glu promoted cell growth and ß-casein expression through the mTORC1 pathway. When Glu was added from DCMECs, both Adenosine 5'-monophosphate-activated protein kinase α (AMPKα) and Sestrin2 (SESN2) expression were decreased. Overexpression and silencing of AMPKα or SESN2 uncovered that AMPKα suppressed cell growth and ß-casein synthesis through inhibiting mTORC1 pathway, and SESN2 suppressed cell growth and ß-casein synthesis through activating AMPK pathway. When Glu was depleted from DCMECs, both activating transcription factor 4 (ATF4) and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) expression were increased. Overexpression or silencing of ATF4 or Nrf2 demonstrated that Glu depletion promoted SESN2 expression through ATF4 and Nrf2. Together, these results indicate that in DCMECs, Glu promoted cell growth and casein synthesis via ATF4/Nrf2-SESN2-AMPK-mTORC1 pathway.


Asunto(s)
Factor de Transcripción Activador 4 , Caseínas , Femenino , Bovinos , Animales , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Factor de Transcripción Activador 4/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Glucosa/farmacología , Glucosa/metabolismo , Glándulas Mamarias Animales/metabolismo , Células Epiteliales/metabolismo
3.
Cancer Cell Int ; 23(1): 145, 2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37480002

RESUMEN

Papillary thyroid carcinoma (PTC) is one of the most common thyroid carcinomas. The gross extrathyroidal extension and extensive metastases of PTC lead to high rates of recurrence and poor clinical outcomes. However, the mechanisms underlying PTC development are poorly understood. In this study, using single-cell RNA sequencing, the transcriptome profiles of two PTC patients were addressed, including PTC1 with low malignancy and good prognosis and PTC2 with high malignancy and poor prognosis. We found that epithelial subcluster Epi02 was the most associated with the malignant development of PTC cells, with which the fold change of Chitinase 3-like 1 (CHI3L1) is on the top of the differentially expressed genes between PTC1 and PTC2 (P < 0.001). However CHI3L1 is rarely investigated in PTC as far. We then studied its role in PTC with a series of experiments. Firstly, qRT-PCR analysis of 14 PTC patients showed that the expression of CHI3L1 was positively correlated with malignancy. In addition, overexpression or silencing of CHI3L1 in TPC-1 cells, a PTC cell line, cultured in vitro showed that the proliferation, invasion, and metastasis of the cells were promoted or alleviated by CHI3L1. Further, immunohistochemistry analysis of 110 PTC cases revealed a significant relationship between CHI3L1 protein expression and PTC progression, especially the T (P < 0.001), N (P < 0.001), M stages (P = 0.007) and gross ETE (P < 0.001). Together, our results prove that CHI3L1 is a positive regulator of malignant development of PTC, and it promotes proliferation, invasion, and metastasis of PTC cells. Our study improves understanding of the molecular mechanisms underlying the progression of PTC and provides new insights for the clinical diagnosis and treatment of PTC.

4.
Anim Nutr ; 13: 126-136, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37123620

RESUMEN

Short-chain fatty acids are important nutrients that regulate milk fat synthesis. They regulate milk synthesis via the sterol regulatory element binding protein 1 (SREBP1) pathway; however, the details are still unknown. Here, the regulation and mechanism of sodium acetate (SA) in milk fat synthesis in bovine mammary epithelial cells (BMECs) were assessed. BMECs were treated with SA supplementation (SA+) or without SA supplementation (SA-), and milk fat synthesis and activation of the SREBP1 pathway were increased (P = 0.0045; P = 0.0042) by SA+ and decreased (P = 0.0068; P = 0.0031) by SA-, respectively. Overexpression or inhibition of SREBP1 demonstrated that SA promoted milk fat synthesis (P = 0.0045) via the SREBP1 pathway. Overexpression or inhibition of TATA element modulatory factor 1 (TMF1) demonstrated that TMF1 suppressed activation of the SREBP1 pathway (P = 0.0001) and milk fat synthesis (P = 0.0022) activated by SA+. Overexpression or inhibition of TMF1 and SREBP1 showed that TMF1 suppressed milk fat synthesis (P = 0.0073) through the SREBP1 pathway. Coimmunoprecipitation analysis revealed that TMF1 interacted with SREBP1 in the cytoplasm and suppressed the nuclear localization of SREBP1 (P = 0.0066). The absence or presence of SA demonstrated that SA inhibited the expression of TMF1 (P = 0.0002) and the interaction between TMF1 and SREBP1 (P = 0.0001). Collectively, our research suggested that TMF1 was a new negative regulator of milk fat synthesis. In BMECs, SA promoted the SREBP1 pathway and milk fat synthesis by suppressing TMF1. This study enhances the current understanding of the regulation of milk fat synthesis and provides new scientific data for the regulation of milk fat synthesis.

5.
RNA Biol ; 18(sup1): 232-243, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34586036

RESUMEN

RNA-seq has been widely used to reveal the molecular mechanism of variants of life process. We have developed an alternative method, MustSeq, which generates multiple second strands along a single 1st strand cDNA by random-priming initiation, immediately after reverse transcription for each RNA extract using sample-barcoded poly-dT primers, then 3' ends-enriching PCR is applied to construct the library. Unlike the conventional RNA seq, MustSeq avoids procedures such as mRNA isolation, fragmentation and RNA 5'-end capture, enables early pooling of multiple samples, and requires only one twentieth of sequencing reads of full-length sequencing. We demonstrate the power and features of MustSeq comparing with TruSeq and NEBNext RNA-seq, two conventional full-length methods and QuantSeq, an industrial 3' end method. In cancer cell lines, the reads distribution of CDS-exon as well as genes, lncRNAs and GO terms detected by MustSeq are closer than QuantSeq to TruSeq. In mouse hepatocarcinoma and healthy livers, MustSeq enriches the same pathways as by NEBNext, and reveals the molecular profile of carcinogenesis. Overall MustSeq is a robust and accurate RNA-seq method allowing efficient library construction, sequencing and analysis, particularly valuable for analysis of differentially expressed genes with a large number of samples. MustSeq will greatly accelerate the application of bulk RNA-seq on different fields, and potentially applicable for single cell RNA-seq.


Asunto(s)
Regiones no Traducidas 3'/genética , ARN Mensajero/genética , RNA-Seq/métodos , Análisis de Secuencia de ARN/métodos , Transcriptoma , Animales , Biblioteca de Genes , Células HeLa , Humanos , Células Jurkat , Células K562 , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/análisis
6.
J Agric Food Chem ; 69(24): 6810-6819, 2021 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-34096300

RESUMEN

In the dairy industry, glutamine (Gln) is often used as a feed additive to increase milk yield and quality; however, the molecular regulation underneath needs further clarification. Here, with bovine mammary epithelial cells (BMECs), the effects and mechanisms of Gln on cell growth and casein synthesis were assessed. When Gln was added or depleted from BMECs, both cell growth and ß-casein (CSN2) expression were increased or decreased, respectively. Overexpressing or inhibiting the mechanistic target of rapamycin (mTOR) revealed that Gln regulated cell growth and CSN2 synthesis through the mTORC1 pathway. A similar intervention of ADP-ribosylation factor 1 (Arf1) uncovered that Gln activated the mTORC1 pathway through Arf1. We next observed that both guanine nucleotide exchange factors, Cytohesin-1/2/3 (CYTH1/2/3, CYTHs) and ADP-ribosylation factor GTPase activating protein 1 (ARFGAP1), interacted with Arf1. Inhibiting CYTHs or ARFGAP1 showed that Gln supplement or depletion activated or inactivated Arf1 through CYTHs or ARFGAP1, respectively. Collectively, this study demonstrated that Gln positively regulated cell growth and casein synthesis in BMECs, which works through the CYTHs/ARFGAP1-Arf1-mTORC1 pathway. These results greatly enhanced current understanding regarding the regulation of the mTOR pathway and provided new insights for the processes of cell growth and casein synthesis by amino acids, particularly Gln.


Asunto(s)
Factor 1 de Ribosilacion-ADP , Caseínas , Animales , Caseínas/metabolismo , Bovinos , Células Epiteliales/metabolismo , Glutamina , Glándulas Mamarias Animales/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Transducción de Señal
7.
Biomed Pharmacother ; 126: 110074, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32163746

RESUMEN

Cardiovascular disease (CVD) is a significant cause of death worldwide. Because of its major individual differences in genetic background, pathogenesis, and disease progression pattern, the mortality risk rate remains high following conventional Western medicine diagnosis under current guidelines. Traditional Chinese medicine (TCM) has important multi-target, multi-pathway, and multi-layer benefits that can effectively address western medicine deficiencies. It was therefore commonly used in CVD diagnosis. Oxidative stress is also one of the main factors of CVD. Likewise, this main reaction regulator is the nuclear factor erythroid-2-related (Nrf2) factor. When activated, it can be transferred to the nucleus and initiated in the downstream pathway, thus playing an anti-oxidant stress role. As one of the most crucial endogenous protection systems in the body, Nrf2-related / heme oxygenase 1 (Nrf2/HO-1) signaling pathway is Nrf2's most classic approach to playing roles. Recently, various advances have been made to research and explain TCM by manipulating this pathway to treat CVD using modern molecular biology and other approaches. This analysis summarized the relationship between Nrf2/HO-1 signaling route, CVD and TCM. Further, Autodock calculation was also conducted to determine the binding amino acid on this TCM to Nrf2 and HO-1.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Hemo-Oxigenasa 1/metabolismo , Medicina Tradicional China , Factor 2 Relacionado con NF-E2/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Biomarcadores , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Medicamentos Herbarios Chinos/uso terapéutico , Humanos , Modelos Biológicos , Terapia Molecular Dirigida , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
8.
Biochem Biophys Res Commun ; 523(4): 847-852, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-31954518

RESUMEN

Glycyl-tRNA synthetase (GlyRS) has non-canonical roles beyond aminoacylation, but the molecular mechanism is largely unknown. We have previously found that GlyRS is phosphorylated in the cytoplasm of bovine mammary epithelial cells (bMECs) in response to amino acid stimulation, and the phosphorylated GlyRS enters nucleus to stimulate gene expression for milk synthesis. In this study, we aim to uncover the upstream kinase of GlyRS and reveal the signaling pathways that methionine (Met) stimulates GlyRS phosphorylation. We show that mitogen-activated protein kinase 10 (MAP3K10) interacts with GlyRS in bMECs by Co-IP, mass spectrometry, and Western blotting analysis. We further identify that MAP3K10 is an upstream kinase of GlyRS by in vitro kinase assay and MAP3K10 stimulates NFκB1 phosphorylation via activating GlyRS. We also uncover that Met stimulates GlyRS phosphorylation via the GPR87-CDC42/Rac1-MAP3K10 signaling pathway. Our findings help to understand the molecular mechanism of GlyRS in cellular signaling transduction.


Asunto(s)
Glicina-ARNt Ligasa/metabolismo , Metionina/farmacología , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Bovinos , Activación Enzimática/efectos de los fármacos , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
9.
J Agric Food Chem ; 67(17): 4849-4859, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-30969118

RESUMEN

Sestrin2 (SESN2) negatively regulates the mammalian target of rapamycin complex 1 (mTORC1) pathway and casein synthesis in response to amino acid (AA) depletion in cow mammary epithelial cells (CMECs); however, the underlying mechanism is unclear. In the current study, the regulation of SESN2 on AA-mediated ß-casein (CSN2) synthesis in CMECs and its mechanism were investigated. Overexpression and silencing of SESN2 demonstrated that SESN2 negatively regulated AA-mediated expression of CSN2 and mTORC1 pathway. Co-immunoprecipitation analysis showed that SESN2 interacted with SH3 domain-binding protein 4 (SH3BP4). Overexpression and silencing of SH3BP4 demonstrated that SH3BP4 negatively regulated AA-mediated expression of CSN2 and mTORC1 pathway and that SESN2 negatively regulated expression of CSN2 and mTORC1 pathway through the SH3BP4 in the presence and absence of AA. The absence or presence of AA demonstrated that AA negatively regulated expression and nuclear localization of activating transcription factor 4 (ATF4). Overexpression and silencing of ATF4 demonstrated that AA negatively regulated SESN2 expression through ATF4. Together, these results indicate that SESN2 negatively regulates the mTORC1 pathway and subsequent CSN2 synthesis through the SH3BP4 in response to AA absence or presence in CMECs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aminoácidos/metabolismo , Caseínas/biosíntesis , Bovinos/metabolismo , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Bovinos/genética , Femenino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Proteínas Nucleares/genética , Unión Proteica , Transducción de Señal
10.
J Cell Physiol ; 234(3): 2973-2983, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30171693

RESUMEN

Amino acids are required for the mammalian target of rapamycin (mTOR) signaling pathway and milk synthesis in bovine mammary epithelial cells (BMECs). However, the mechanism through which amino acids activate this pathway is largely unknown. Here we show that glycyl-tRNA synthetase (GlyRS) mediates amino acid-induced activation of the mTOR-S6K1/4EBP1 pathway, and milk protein and fat synthesis in BMECs. Among 19 aminoacyl-tRNA synthetases, only the mRNA expression of GlyRS and Leucyl-tRNA synthetase (LeuRS) were significantly increased by several amino acids including Met and Leu. We then observed that GlyRS knockdown abolished the stimulation of Met on milk protein and fat synthesis in BMECs, whereas GlyRS overexpression led to more significantly increased milk synthesis in cells treated with Met. By western blotting and qualitative real time-polymerase chain reaction analysis (qRT-PCR) analysis, we next revealed that GlyRS is required for amino acid-induced activation of the mTOR-S6K1/4EBP1 pathway. Thus, this study establishes that GlyRS mediates amino acid-induced activation of the mTOR pathway, thereby regulating milk protein and fat synthesis.


Asunto(s)
Células Epiteliales/metabolismo , Glicina-ARNt Ligasa/genética , Glándulas Mamarias Animales/metabolismo , Leche/metabolismo , Aminoácidos/genética , Aminoácidos/metabolismo , Animales , Bovinos , Femenino , Leucina/metabolismo , Glándulas Mamarias Animales/crecimiento & desarrollo , Metionina/metabolismo , Proteínas de la Leche/biosíntesis , Proteínas Quinasas S6 Ribosómicas 70-kDa , Transducción de Señal , Serina-Treonina Quinasas TOR/genética
11.
J Cell Physiol ; 234(5): 7608-7621, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30471104

RESUMEN

Amino acids are required for the activation of mammalian target of rapamycin (mTOR) to increase cell growth, protein and lipid synthesis, and inhibit autophagy. However, the mechanism through which amino acids activate the mTOR signaling is still largely unknown. In our previous study, we discovered that glycyl-tRNA synthetase (GlyRS) is a key mediator of amino-acid-induced mTOR expression and activation in bovine mammary epithelial cells (BMECs). Here we show that amino acids stimulate GlyRS nuclear localization for mTOR expression in BMECs. Met stimulates GlyRS nuclear localization, and the nuclear GlyRS is cleaved into a C-terminus-containing truncated form. We prove that GlyRS has a bipartite nuclear leading sequences, and GlyRS is phosphorylated at Thr544 and Ser704 in the cytoplasm under the stimulation of amino acids (Met, Leu, and Lys). The nuclear GlyRS physically binds to nuclear factor kappa B1, triggers its phosphorylation, thereby enhancing mRNA expression of its target genes including mTOR, S6K1, and 4EBP1. We further demonstrate that GlyRS is required for the inhibition of autophagy by Met. Thus our work elucidates that amino acids trigger GlyRS phosphorylation and nuclear localization to enhance the mRNA expression of mTOR.


Asunto(s)
Aminoácidos/metabolismo , Células Epiteliales/metabolismo , Glicina-ARNt Ligasa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Autofagia/fisiología , Bovinos , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Femenino , Glándulas Mamarias Animales/metabolismo , Fosforilación/fisiología , Transducción de Señal/fisiología
12.
J Agric Food Chem ; 66(49): 13041-13049, 2018 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-30499671

RESUMEN

Lactation of bovine mammary epithelial cells (BMEC) is a complex biological process that involves in various organelles. Studies have shown that lysosome and lysosomal membrane proteins (LMP) plays an important role in lactation of BMEC. But the LMP of BMEC remains poorly understood. To obtain a global view of the LMP of BMEC and the affect of lysosome on lactation, the LMP of BMEC was identified using sequential windowed acquisition of all theoretical mass spectra (LC-SWATH/MS). 1214 LMP were identified and 559 were reported to be localized on lysosomal membrane for the first time in BMEC. Gene ontology annotation of these identified proteins showed that both previously reported casein synthesis-related LMP, such as LAMTOR1, 2, 3, and rRagC, and newly identified casein and milk fat synthesis-related LMP, such as EIF4E and ACAA1, were found. KEGG pathway analysis of these identified proteins showed that some pathways involved in lactation, such as PI3K-Akt, mTOR, insulin, PPAR, and JAK-STAT pathway, were found. The lysosomal location of five proteins (PRKCA, EIF4E, ACAA1, HRAS, and THBS1) was analyzed by laser confocal microscopy, and all five were associated with the lysosomal membrane. These findings help to elucidate lysosome functions in the regulation of lactation. The results implicate lysosomes as important organelles in regulation of lactation of BMEC that have been previously undervalued.


Asunto(s)
Bovinos , Lactancia/fisiología , Proteínas de Membrana de los Lisosomas/análisis , Lisosomas/fisiología , Glándulas Mamarias Animales/química , Proteómica , Animales , Células Epiteliales/química , Femenino , Proteínas de Membrana de los Lisosomas/fisiología , Microscopía Confocal/veterinaria
13.
Biochim Biophys Acta Proteins Proteom ; 1866(11): 1092-1101, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30282607

RESUMEN

In cow mammary epithelial cells (CMECs), cell growth and casein synthesis are regulated by amino acids (AAs), and lysosomes are important organelles in this regulatory process, but the mechanisms remain unclear. Herein, lysosomal membrane proteins (LMPs) in CMECs in the presence (Leu+) and absence (Leu-) of leucine were quantitatively analysed using Sequential Windowed Acquisition of All Theoretical Fragment Ion (SWATH) mass spectrometry. In identified LMPs, Guanine nucleotide-binding protein subunit gamma-12 (GNG12) was a markedly up-regulated protein in Leu+ group. CMECs were treated with Leu+ or Leu-, expression and lysosomal localization of GNG12 were decreased in response to Leu absence. Overexpressing or inhibiting GNG12 demonstrated that cell growth, casein synthesis and activation of the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway were all up-regulated by GNG12. Cell growth, casein synthesis and mTORC1 signaling pathway were decreased in response to Leu absence, but these decreases were partially restored by GNG12 overexpression, and those effects were partially reversed by inhibiting GNG12. Co-immunoprecipitation analysis showed that GNG12 activates the mTORC1 pathway via interaction with Ragulator. Taken together, these results suggest that GNG12 is a positive regulator of the Leu-mediated mTORC1 signaling pathway in CMECs that promotes cell growth and casein synthesis.


Asunto(s)
Proliferación Celular , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Leucina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Animales , Caseínas/biosíntesis , Bovinos , Células Cultivadas , Células Epiteliales/metabolismo , Femenino , Subunidades gamma de la Proteína de Unión al GTP/genética , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Glándulas Mamarias Animales/citología , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Proteómica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal
14.
J Proteome Res ; 17(9): 3128-3142, 2018 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-30079738

RESUMEN

Aflatoxin M1 (AFM1) is a common mycotoxin in dairy milk, and it is typically concurrently present with other mycotoxins that may represent a threat to food safety. However, knowledge of how AFM1, alone or in combination with other mycotoxins, may affect human intestinal epithelial integrity remain to be established. We employed transcriptome and proteome analysis integrated with biological validation to reveal the molecular basis underlining the effect of exposure to AFM1, ochratoxin A (OTA), or both on the intestinal epithelial integrity of differentiated Caco-2 cells. Exposure to 4 µg/mL of OTA was found to disrupt human gut epithelial integrity, whereas 4 µg/mL of AFM1 did not. The integrated transcriptome and proteome analysis of AFM1 and OTA, alone or in combination, indicate the synergistic effect of the two mycotoxins in disrupting intestinal integrity. This effect was mechanistically linked to a broad range of pathways related to intestinal integrity enriched by down-regulated genes and proteins, associated with focal adhesion, adheren junction, and gap junction pathways. Furthermore, the cross-omics analysis of mixed AFM1 and OTA compared to OTA alone suggest that kinase family members, including myosin light-chain kinase, mitogen-activated protein kinases, and protein kinase C, are the potential key regulators in modulating intestinal epithelial integrity. These findings provide novel insight into the synergistic detrimental role of multiple mycotoxins in disrupting intestinal integrity and, therefore, identify potential targets to improve milk safety related to human health.


Asunto(s)
Aflatoxina M1/toxicidad , Adhesiones Focales/efectos de los fármacos , Ocratoxinas/toxicidad , Proteoma/genética , Transcriptoma , Uniones Adherentes/efectos de los fármacos , Células CACO-2 , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Uniones Comunicantes/efectos de los fármacos , Regulación de la Expresión Génica , Humanos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Mapas de Interacción de Proteínas , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Proteoma/clasificación , Proteoma/metabolismo
15.
Sci Rep ; 8(1): 3912, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29500367

RESUMEN

Amino acids (AA) are one of the key nutrients that regulate cell proliferation and casein synthesis in cow mammary epithelial cells (CMEC), but the mechanism of this regulation is not yet clear. In this study, the effect of SESN2 on AA-mediated cell proliferation and casein synthesis in CMEC was assessed. After 12 h of AA starvation, CMECs were cultured in the absence of all AA (AA-), in the presences of only essential AA (EAA+), or of all AA (AA+). Cell proliferation, casein expression, and activation of the mammalian target of rapamycin complex 1 (mTORC1) pathway were increased; but SESN2 expression was decreased in response to increased EAA or AA supply. Overexpressing or inhibiting SESN2 demonstrated that cell proliferation, casein expression, and activation of the mTORC1 pathway were all controlled by SESN2 expression. Furthermore, the increase in cell proliferation, casein expression, and activation of the mTORC1 pathway in response to AA supply was inhibited by overexpressing SESN2, and those effects were reversed by inhibiting SESN2. These results indicate that SESN2 is an important inhibitor of mTORC1 in CMEC blocking AA-mediated cell proliferation and casein synthesis.


Asunto(s)
Aminoácidos/farmacología , Caseínas/metabolismo , Proliferación Celular , Células Epiteliales/citología , Glándulas Mamarias Animales/citología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Nucleares/metabolismo , Animales , Bovinos , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Proteínas Nucleares/genética , Fosforilación , Transducción de Señal
16.
Toxins (Basel) ; 10(1)2017 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-29280945

RESUMEN

Aflatoxin M1 (AFM1) and ochratoxin A (OTA) are mycotoxins commonly found in milk; however, their effects on intestinal epithelial cells have not been reported. In the present study, we show that AFM1 (0.12 and 12 µM) and OTA (0.2 and 20 µM) individually or collectively increased the paracellular flux of lucifer yellow and fluorescein isothiocyanate (FITC)-dextrans (4 and 40 kDa) and decreased transepithelial electrical resistance values in differentiated Caco-2 cells after 48 h of exposure, indicating increased epithelial permeability. Immunoblotting and immunofluorescent analysis revealed that AFM1, OTA, and their combination decreased the expression levels of tight junction (TJ) proteins and disrupted their structures, namely, claudin-3, claudin-4, occludin, and zonula occludens-1 (ZO-1), and p44/42 mitogen-activated protein kinase (MAPK) partially involved in the mycotoxins-induced disruption of intestinal barrier. The effects of a combination of AFM1 and OTA on intestinal barrier function were more significant (p < 0.05) than those of AFM1 and OTA alone, yielding additive or synergistic effects. The additive or synergistic effects of AFM1 and OTA on intestinal barrier function might affect human health, especially in children, and toxin risks should be considered.


Asunto(s)
Aflatoxina M1/farmacología , Mucosa Intestinal/efectos de los fármacos , Ocratoxinas/farmacología , Células CACO-2 , Diferenciación Celular , Dextranos/farmacología , Sinergismo Farmacológico , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/farmacología , Colorantes Fluorescentes/farmacología , Humanos , Mucosa Intestinal/metabolismo , Isoquinolinas/farmacología , Permeabilidad/efectos de los fármacos , Proteínas de Uniones Estrechas/metabolismo
17.
J Agric Food Chem ; 65(48): 10464-10472, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29112400

RESUMEN

To investigate the effect and potential mechanisms of lactoferrin on colon cancer cells and tumors, HT29 and HCT8 cells were exposed to varying concentrations of lactoferrin, and the impacts on cell proliferation, migration, and invasion were observed. Cell proliferation test showed that high dosage of lactoferrin (5-100 mg/mL) inhibited cell viability in a dose-dependent manner, with the 50% concentration of inhibition at 81.3 ± 16.7 mg/mL and 101 ± 23.8 mg/mL for HT29 and HCT8 cells, respectively. Interestingly, migration and invasion of the cells were inhibited dramatically by 20 mg/mL lactoferrin, consistent with the significant down regulation of VEGFR2, VEGFA, pPI3K, pAkt, and pErk1/2 proteins. HT29 was chosen as the sensitive cell line to construct a tumor-bearing nude mice model. Notably, HT29 tumor weight was greatly reduced in both the lactoferrin group (26.5 ± 6.7 mg) and the lactoferrin/5-Fu group (14.5 ± 5.1 mg), compared with the control one (39.3 ± 6.5 mg), indicating that lactoferrin functioned as a tumor growth inhibitor. Considering lactoferrin also reduced the growth of blood vessels and the degree of malignancy, we concluded that HT29 tumors were effectively suppressed by lactoferrin, which might be achieved by regulation of phosphorylation from various kinases and activation of the VEGFR2-PI3K/Akt-Erk1/2 pathway.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Antineoplásicos/administración & dosificación , Neoplasias del Colon/tratamiento farmacológico , Lactoferrina/administración & dosificación , Inhibidores de la Angiogénesis/química , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/fisiopatología , Modelos Animales de Enfermedad , Células HT29 , Humanos , Lactoferrina/química , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/fisiopatología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Int J Mol Sci ; 16(12): 29936-47, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26694361

RESUMEN

Tudor staphylococcal nuclease (Tudor-SN) is a highly conserved and ubiquitously expressed multifunctional protein, related to multiple and diverse cell type- and species-specific cellular processes. Studies have shown that Tudor-SN is mainly expressed in secretory cells, however knowledge of its role is limited. In our previous work, we found that the protein level of Tudor-SN was upregulated in the nucleus of bovine mammary epithelial cells (BMEC). In this study, we assessed the role of Tudor-SN in milk synthesis and cell proliferation of BMEC. We exploited gene overexpression and silencing methods, and found that Tudor-SN positively regulates milk synthesis and proliferation via Stat5a activation. Both amino acids (methionine) and estrogen triggered NFκB1 to bind to the gene promoters of Tudor-SN and Stat5a, and this enhanced the protein level and nuclear localization of Tudor-SN and p-Stat5a. Taken together, these results suggest the key role of Tudor-SN in the transcriptional regulation of milk synthesis and proliferation of BMEC under the stimulation of amino acids and hormones.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/citología , Leche/metabolismo , Proteínas Nucleares/metabolismo , Animales , Bovinos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Estrógenos/farmacología , Femenino , Silenciador del Gen/efectos de los fármacos , Metionina/farmacología , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
19.
In Vitro Cell Dev Biol Anim ; 51(7): 697-704, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26183263

RESUMEN

14-3-3 proteins are an acidic protein family that is highly conserved and widely distributed in eukaryotic cells. Recent studies have found that 14-3-3 proteins play critical roles in cell signal transductions, cell growth and differentiation, and protein synthesis. 14-3-3γ is an important member of 14-3-3 protein family. In our previous study, we found that 14-3-3γ was upregulated by estrogen in dairy cow mammary epithelial cell (DCMEC), but the function and mechanism of 14-3-3γ is not known. In this experiment, we first cultured and purified the primary DCMEC and found 14-3-3γ located both in the cytoplasm and nucleus by using immunofluorescence assay. Methionine, lysine, estrogen, and prolactin could upregulate the expression of 14-3-3γ, stimulate the secretion of ß-casein and triglyceride, and raise the cell viability of DCMEC. We constructed a stable 14-3-3γ overexpression cell line of DCMEC and found that the expressions of mTOR and p-mTOR, the secretion of triglyceride and ß-casein (CSN2), and the cell viability of DCMEC were all upregulated. We also observed the effects of 14-3-3γ gene silencing and gained consistent results with 14-3-3γ overexpression. These findings reveal that 14-3-3γ affects the mTOR pathway and regulates lactogenesis in DCMECs.


Asunto(s)
Proteínas 14-3-3/metabolismo , Glándulas Mamarias Animales/citología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas 14-3-3/genética , Animales , Caseínas/metabolismo , Bovinos , Supervivencia Celular , Células Cultivadas , Industria Lechera , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Lactancia , Lisina/metabolismo , Lisina/farmacología , Glándulas Mamarias Animales/metabolismo , Metionina/metabolismo , Metionina/farmacología , Prolactina/metabolismo , Prolactina/farmacología , Transducción de Señal , Triglicéridos/metabolismo
20.
Arch Biochem Biophys ; 564: 142-55, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25281768

RESUMEN

14-3-3γ, an isoform of the 14-3-3 protein family, was proved to be a positive regulator of mTOR pathway. Here, we analyzed the function of 14-3-3γ in protein synthesis using bovine mammary epithelial cells (BMECs). We found that 14-3-3γ interacted with eIF1AX and RPS7 by 14-3-3γ coimmunoprecipitation (CoIP) and matrix-assisted laser desorption/ionization-time-of-flight/time-of-flight (MALDI-TOF/TOF) peptide mass fingerprinting analysis. These interactions of 14-3-3γ with eIF1AX and RPS7 were further confirmed by colocalization and fluorescence resonance energy transfer (FRET) analysis. We also found that methionine could promote protein synthesis and trigger the protein expression levels of 14-3-3γ, eIF1AX and RPS7. Analysis of overexpression and inhibition of 14-3-3γ confirmed that it positively affected the protein expression levels of eIF1AX, RPS7, Stat5 and mTOR pathway to promote protein synthesis and cell proliferation in BMECs. We further showed that overexpression of eIF1AX and RPS7 also triggered protein translation and cell proliferation. From these results, we conclude that molecular network including eIF1AX, RPS7, and 14-3-3γ regulates protein translation and cell proliferation in BMECs.


Asunto(s)
Proteínas 14-3-3/metabolismo , Proliferación Celular/fisiología , Células Epiteliales/metabolismo , Factor 1 Eucariótico de Iniciación/metabolismo , Glándulas Mamarias Animales/metabolismo , Biosíntesis de Proteínas/fisiología , Proteína S6 Ribosómica/metabolismo , Proteínas 14-3-3/genética , Animales , Bovinos , Células Cultivadas , Células Epiteliales/citología , Factor 1 Eucariótico de Iniciación/genética , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Glándulas Mamarias Animales/citología , Proteína S6 Ribosómica/genética , Resonancia por Plasmón de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA