Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Biol (Noisy-le-grand) ; 70(1): 99-109, 2024 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-38372107

RESUMEN

This study aimed to explore the involvement of Transmembrane and coiled-coil domains 1 (TMCO1) in ovarian cancer progression and its regulatory mechanisms in cisplatin resistance. Using the GEPIA database, we analyzed TMCO1 expression in ovarian cancer and normal tissues. In a cohort of 99 ovarian cancer patients, immunohistochemistry and immunofluorescence were employed to assess TMCO1 expression in tumor and adjacent tissues, correlating findings with clinical and pathological characteristics. TMCO1 overexpression and knockout cell models were constructed, and their impact on non-cisplatin-resistant (SK-OV-3) and cisplatin-resistant (SK-OV-3-CDDP) ovarian cancer cells was investigated through cloning, wound healing, Fluo 4, and Transwell experiments. Knocking down CALR and VDAC1 was performed to examine their effects on TMCO1, cell proliferation, and malignant markers. Subcutaneous tumor models in nude mice elucidated the in vivo role of TMCO1 in tumor growth. Expression levels of CALR, VDAC1, angiogenesis indicators (CD34), and epithelial-mesenchymal transition (EMT) markers were evaluated. TMCO1 expression in ovarian cancer tissue significantly differed from normal tissue, correlating with survival rates. TMCO1 overexpression was associated with lymph node metastases, late FIGO stage, and larger tumors. TMCO1 promoted proliferation, calcium ion elevation, cytoskeletal remodeling, and metastasis in SK-OV-3 and SK-OV-3-CDDP cells, upregulating VDAC1, CALR, Vimentin, N-cadherin, ß-catenin, and downregulating E-cadherin. Silencing TMCO1 inhibited cell growth, proliferation, and angiogenesis in vivo, suppressing the expression of CALR, VDAC1, Vimentin, N-cadherin, and ß-catenin. Overall, this study highlighted TMCO1 as a crucial regulator in ovarian cancer progression, influencing VDAC1 through CALR and impacting diverse cellular processes, offering potential as a targeted therapeutic strategy for ovarian cancer.


Asunto(s)
Canales de Calcio , Calreticulina , Neoplasias Ováricas , Animales , Femenino , Humanos , Ratones , beta Catenina/metabolismo , Cadherinas/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Cisplatino/farmacología , Cisplatino/uso terapéutico , Transición Epitelial-Mesenquimal/genética , Ratones Desnudos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Vimentina/metabolismo , Calreticulina/genética , Calreticulina/metabolismo
2.
J Sci Food Agric ; 103(4): 2175-2185, 2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36541582

RESUMEN

BACKGROUND: In recent years, an increasing interest in healthy functional foods has been documented among health-conscious consumers. Steam explosion (SE)-treated black-grained wheat (BGW) bran was explored for the development of chiffon cakes with high nutritional and functional value. RESULTS: The content of crude fat and total starch decreased with increasing SE pressure, whereas water-holding capacity and antioxidant activity increased, suggesting SE at 0.6-1.0 MPa could be an effective technique for enhancing the nutritional and functional properties of wheat bran. The protein, iron, zinc, manganese, selenium, and soluble dietary fiber contents, the water-holding, oil-binding, swelling, cholesterol binding, and cation-exchange capacities, and antioxidant activity of SE BGW bran were better than those of SE white-grained wheat bran. The addition of SE bran (0.8 MPa) to flour significantly decreased the peak viscosity, final viscosity, and setback and increased the pasting temperature. The effect of SE bran on the pasting properties of low-gluten and medium-gluten flour was stronger than that of high-gluten flour. SE BGW bran altered the physicochemical properties of chiffon cakes. When 6% SE BGW bran (0.8 MPa) was added, chiffon cakes exhibited good specific volume, hardness, chewiness, and other sensory qualities. CONCLUSIONS: These results indicate that SE at 0.6-1.0 MPa is an effective technique for enhancing the nutritional and functional properties of wheat bran. SE BGW bran can be alternatives to food materials for developing health functional cereal-based products. © 2022 Society of Chemical Industry.


Asunto(s)
Fibras de la Dieta , Vapor , Fibras de la Dieta/análisis , Antioxidantes , Agua , Harina/análisis , Glútenes
3.
Clin Genet ; 102(6): 483-493, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36029107

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD), caused by mutations in PKD1 and PKD2 (PKD1/2), has unexplained phenotypic variability likely affected by environmental and other genetic factors. Approximately 10% of individuals with ADPKD phenotype have no causal mutation detected, possibly due to unrecognized risk variants of PKD1/2. This study was designed to identify risk variants of PKD genes through population genetic analyses. We used Wright's F-statistics (Fst) to evaluate common single nucleotide variants (SNVs) potentially favored by positive natural selection in PKD1 from 1000 Genomes Project (1KG) and genotyped 388 subjects from the Rogosin Institute ADPKD Data Repository. The variants with >90th percentile Fst scores underwent further investigation by in silico analysis and molecular genetics analyses. We identified a deep intronic SNV, rs3874648G> A, located in a conserved binding site of the splicing regulator Tra2-ß in PKD1 intron 30. Reverse-transcription PCR (RT-PCR) of peripheral blood leukocytes (PBL) from an ADPKD patient homozygous for rs3874648-A identified an atypical PKD1 splice form. Functional analyses demonstrated that rs3874648-A allele increased Tra2-ß binding affinity and activated a cryptic acceptor splice-site, causing a frameshift that introduced a premature stop codon in mRNA, thereby decreasing PKD1 full-length transcript level. PKD1 transcript levels were lower in PBL from rs3874648-G/A carriers than in rs3874648-G/G homozygotes in a small cohort of normal individuals and patients with PKD2 inactivating mutations. Our findings indicate that rs3874648G > A is a PKD1 expression modifier attenuating PKD1 expression through Tra2-ß, while the derived G allele advantageously maintains PKD1 expression and is predominant in all subpopulations.


Asunto(s)
Riñón Poliquístico Autosómico Dominante , Canales Catiónicos TRPP , Humanos , Intrones , Mutación , Nucleótidos , Riñón Poliquístico Autosómico Dominante/genética , Sitios de Empalme de ARN , Canales Catiónicos TRPP/genética
4.
J Am Soc Nephrol ; 32(12): 3114-3129, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34716216

RESUMEN

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is a genetic disorder characterized by the development of multiple cysts in the kidneys. It is often caused by pathogenic mutations in PKD1 and PKD2 genes that encode polycystin proteins. Although the molecular mechanisms for cystogenesis are not established, concurrent inactivating germline and somatic mutations in PKD1 and PKD2 have been previously observed in renal tubular epithelium (RTE). METHODS: To further investigate the cellular recessive mechanism of cystogenesis in RTE, we conducted whole-genome DNA sequencing analysis to identify germline variants and somatic alterations in RTE of 90 unique kidney cysts obtained during nephrectomy from 24 unrelated participants. RESULTS: Kidney cysts were overall genomically stable, with low burdens of somatic short mutations or large-scale structural alterations. Pathogenic somatic "second hit" alterations disrupting PKD1 or PKD2 were identified in 93% of the cysts. Of these, 77% of cysts acquired short mutations in PKD1 or PKD2 ; specifically, 60% resulted in protein truncations (nonsense, frameshift, or splice site) and 17% caused non-truncating mutations (missense, in-frame insertions, or deletions). Another 18% of cysts acquired somatic chromosomal loss of heterozygosity (LOH) events encompassing PKD1 or PKD2 ranging from 2.6 to 81.3 Mb. 14% of these cysts harbored copy number neutral LOH events, while the other 3% had hemizygous chromosomal deletions. LOH events frequently occurred at chromosomal fragile sites, or in regions comprising chromosome microdeletion diseases/syndromes. Almost all somatic "second hit" alterations occurred at the same germline mutated PKD1/2 gene. CONCLUSIONS: These findings further support a cellular recessive mechanism for cystogenesis in ADPKD primarily caused by inactivating germline and somatic variants of PKD1 or PKD2 genes in kidney cyst epithelium.


Asunto(s)
Quistes , Riñón Poliquístico Autosómico Dominante , Humanos , Riñón Poliquístico Autosómico Dominante/genética , Mutación , Células Epiteliales , Canales Catiónicos TRPP/genética
5.
J Am Soc Nephrol ; 29(8): 2139-2156, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30042192

RESUMEN

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is a ciliopathy caused by mutations in PKD1 and PKD2 that is characterized by renal tubular epithelial cell proliferation and progressive CKD. Although the molecular mechanisms involved in cystogenesis are not established, concurrent inactivating constitutional and somatic mutations in ADPKD genes in cyst epithelium have been proposed as a cellular recessive mechanism. METHODS: We characterized, by whole-exome sequencing (WES) and long-range PCR techniques, the somatic mutations in PKD1 and PKD2 genes in renal epithelial cells from 83 kidney cysts obtained from nine patients with ADPKD, for whom a constitutional mutation in PKD1 or PKD2 was identified. RESULTS: Complete sequencing data by long-range PCR and WES was available for 63 and 65 cysts, respectively. Private somatic mutations of PKD1 or PKD2 were identified in all patients and in 90% of the cysts analyzed; 90% of these mutations were truncating, splice site, or in-frame variations predicted to be pathogenic mutations. No trans-heterozygous mutations of PKD1 or PKD2 genes were identified. Copy number changes of PKD1 ranging from 151 bp to 28 kb were observed in 12% of the cysts. WES also identified significant mutations in 53 non-PKD1/2 genes, including other ciliopathy genes and cancer-related genes. CONCLUSIONS: These findings support a cellular recessive mechanism for cyst formation in ADPKD caused primarily by inactivating constitutional and somatic mutations of PKD1 or PKD2 in kidney cyst epithelium. The potential interactions of these genes with other ciliopathy- and cancer-related genes to influence ADPKD severity merits further evaluation.


Asunto(s)
Células Epiteliales/metabolismo , Trasplante de Riñón/métodos , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/cirugía , Canales Catiónicos TRPP/genética , Adulto , Proliferación Celular/genética , Células Cultivadas , Estudios de Cohortes , Femenino , Humanos , Masculino , Mutación/genética , Podocitos/metabolismo , Riñón Poliquístico Autosómico Dominante/fisiopatología , Cuidados Preoperatorios , Pronóstico , Estudios Prospectivos , Reacción en Cadena en Tiempo Real de la Polimerasa , Secuenciación del Exoma
6.
Kidney Int ; 92(6): 1433-1443, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28739140

RESUMEN

Renal fibrosis is a common pathway leading to the progression of chronic kidney disease, and bone marrow-derived fibroblasts contribute significantly to the development of renal fibrosis. However, the signaling mechanisms underlying the activation of these fibroblasts are not completely understood. Here, we examined the role of IL-4 receptor α (IL-4Rα) in the activation of myeloid fibroblasts in two experimental models of renal fibrosis. Compared with wild-type mice, IL-4Rα knockout mice accumulated fewer bone marrow-derived fibroblasts and myofibroblasts in their kidneys. IL-4Rα deficiency suppressed the expression of α-smooth muscle actin, extracellular matrix proteins and the development of renal fibrosis. Furthermore, IL-4Rα deficiency inhibited the activation of signal transducer and activator of transcription 6 (STAT6) in the kidney. Moreover, wild-type mice engrafted with bone marrow cells from IL-4Rα knockout mice exhibited fewer myeloid fibroblasts in the kidney and displayed less severe renal fibrosis following ureteral obstructive injury compared with wild-type mice engrafted with wild-type bone marrow cells. In vitro, IL-4 activated STAT6 and stimulated expression of α-smooth muscle actin and fibronectin in mouse bone marrow monocytes. This was abolished in the absence of IL-4Rα. Thus, IL-4Rα plays an important role in bone marrow-derived fibroblast activation, resulting in extracellular matrix protein production and fibrosis development. Hence, the IL-4Rα/STAT6 signaling pathway may serve as a novel therapeutic target for chronic kidney disease.


Asunto(s)
Células de la Médula Ósea/inmunología , Fibroblastos/inmunología , Riñón/patología , Receptores de Superficie Celular/inmunología , Insuficiencia Renal Crónica/inmunología , Factor de Transcripción STAT6/metabolismo , Actinas/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibrosis , Ácido Fólico/toxicidad , Humanos , Riñón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Monocitos/inmunología , Monocitos/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/patología , Factor de Transcripción STAT6/inmunología , Transducción de Señal , Células Th2/inmunología , Células Th2/metabolismo
7.
J Am Soc Nephrol ; 26(12): 3060-71, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26032813

RESUMEN

Renal fibrosis is a final common manifestation of CKD resulting in progressive loss of kidney function. Bone marrow-derived fibroblast precursors contribute significantly to the pathogenesis of renal fibrosis. However, the signaling mechanisms underlying the activation of bone marrow-derived fibroblast precursors in the kidney are not fully understood. In this study, we investigated the role of the Janus kinase 3 (JAK3)/signal transducer and activator of transcription (STAT6) signaling pathway in the activation of bone marrow-derived fibroblasts. In cultured mouse monocytes, IL-4 or IL-13 activated STAT6 and induced expression of α-smooth muscle actin and extracellular matrix proteins (fibronectin and collagen I), which was abolished by a JAK3 inhibitor (CP690,550) in a dose-dependent manner or blocked in the absence of STAT6. In vivo, STAT6 was activated in interstitial cells of the obstructed kidney, an effect that was abolished by CP690,550. Mice treated with CP690,550 accumulated fewer bone marrow-derived fibroblasts in the obstructed kidneys compared with vehicle-treated mice. Treatment with CP690,550 also significantly reduced myofibroblast transformation, matrix protein expression, fibrosis development, and apoptosis in obstructed kidneys. Furthermore, STAT6-deficient mice accumulated fewer bone marrow-derived fibroblasts in the obstructed kidneys, produced less extracellular matrix protein, and developed much less fibrosis. Finally, wild-type mice engrafted with STAT6(-/-) bone marrow cells displayed fewer bone marrow-derived fibroblasts in the obstructed kidneys and showed less severe renal fibrosis compared with wild-type mice engrafted with STAT6(+/+) bone marrow cells. Our results demonstrate that JAK3/STAT6 has an important role in bone marrow-derived fibroblast activation, extracellular matrix production, and interstitial fibrosis development.


Asunto(s)
Fibroblastos/fisiología , Janus Quinasa 3/metabolismo , Riñón/patología , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Factor de Transcripción STAT6/fisiología , Actinas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Trasplante de Médula Ósea , Células Cultivadas , Colágeno Tipo I/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibronectinas/metabolismo , Fibrosis , Interleucina-13/farmacología , Interleucina-4/farmacología , Janus Quinasa 3/antagonistas & inhibidores , Janus Quinasa 3/genética , Riñón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos , Miofibroblastos/efectos de los fármacos , Miofibroblastos/fisiología , Transducción de Señal/efectos de los fármacos , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología
8.
J Biol Chem ; 289(4): 2072-83, 2014 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-24324267

RESUMEN

ZNF451 is a transcriptional cofactor localized to promyelocytic leukemia bodies. Here, we present evidence demonstrating that ZNF451 physically interacts with Smad3/4 and functionally inhibits TGF-ß signaling. Increased expression of ZNF451 attenuates TGF-ß-induced growth inhibitory and gene transcriptional responses, whereas depletion of ZNF451 enhances TGF-ß responses. Mechanistically, ZNF451 blocks the ability of Smad3/4 to recruit p300 in response to TGF-ß, which causes reduction of histone H3K9 acetylation on the promoters of TGF-ß target genes. Taken together, ZNF451 acts as a transcriptional corepressor for Smad3/4 and negatively regulates TGF-ß signaling.


Asunto(s)
Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Proteína smad3/metabolismo , Proteína Smad4/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Acetilación , Aminoaciltransferasas , Proteína p300 Asociada a E1A/genética , Proteína p300 Asociada a E1A/metabolismo , Células HEK293 , Células HeLa , Histonas/genética , Histonas/metabolismo , Humanos , Proteínas Represoras/genética , Proteína smad3/genética , Proteína Smad4/genética , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/genética
9.
J Biol Chem ; 289(38): 26441-26450, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25100727

RESUMEN

The bone morphogenetic protein (BMP) signaling pathway regulates a wide range of cellular responses in metazoans. A key step in the canonical BMP signaling is the phosphorylation and activation of transcription factors Smad1, Smad5, and Smad8 (collectively Smad1/5/8) by the type I BMP receptors. We previously identified PPM1A as a phosphatase toward dephosphorylation of all receptor-regulated Smads (R-Smads), including Smad1/5/8. Here we report another nuclear phosphatase named SCP4/CTDSPL2, belonging to the FCP/SCP family, as a novel Smad phosphatase in the nucleus. SCP4 physically interacts with and specifically dephosphorylates Smad1/5/8, and as a result attenuates BMP-induced transcriptional responses. Knockdown of SCP4 in multipotent mesenchymal C2C12 cells leads to increased expression of BMP target genes and consequently promotes BMP-induced osteogenic differentiation. Collectively, our results demonstrate that SCP4, as a Smad phosphatase, plays a critical role in BMP-induced signaling and cellular functions.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Diferenciación Celular , Fosfoproteínas Fosfatasas/fisiología , Procesamiento Proteico-Postraduccional , Proteína Smad1/metabolismo , Secuencias de Aminoácidos , Animales , Dominio Catalítico , Línea Celular , Expresión Génica , Humanos , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Mesodermo/citología , Ratones , Osteoblastos/fisiología , Fosfoproteínas Fosfatasas/química , Fosforilación , Unión Proteica , ARN Polimerasa II/metabolismo , Transducción de Señal , Proteína Smad1/química
10.
Int J Gynecol Cancer ; 24(3): 534-40, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24557437

RESUMEN

OBJECTIVE: AT-rich interactive domain 1A (ARID1A) is a tumor suppressor gene that encodes the BAF250a protein. Recent studies have shown the loss of ARID1A expression in several types of tumors. We aimed to investigate the clinical and pathologic role of BAF250a in endometrial carcinoma. METHODS: We examined the expression of BAF250a and its correlation with the expression of p53, estrogen receptor, progesterone receptor, glucocorticoid receptor, hypoxiainduciblefactor-1α, and vascular endothelial growth factor in normal and various malignant endometrial tissues. RESULTS: The expression of BAF250 was significantly down-regulated in endometrial carcinoma when compared with normal endometrial tissues. The loss of BAF250a expression was found in 25% of endometrial carcinoma samples but not in normal endometrial tissues, complex endometrial hyperplasia, and atypical endometrial hyperplasia samples. Subtypes of endometrial carcinoma, especially uterine endometrioid carcinoma and uterine clear cell carcinoma, had higher frequency of loss of BAF250a expression. In addition, the expression of BAF250a was positively correlated with estrogen receptor and negatively correlated with p53 in poorly differentiated endometrial adenocarcinoma. Moreover, the expression of BAF250a was significantly associated with the differentiation status of endometrial carcinoma but not associated with clinical stage, the depth of myometrial invasion, lymph node metastasis, and overall survival of patients with endometrial carcinoma. CONCLUSIONS: Our data showed that loss of BAF250a is frequently found in high-grade endometrioid and clear cell carcinomas but not in other types of endometrial carcinoma. The loss of BAF250a expression does not have prognostic value for endometrial carcinoma.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma/patología , Neoplasias Endometriales/patología , Endometrio/patología , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Proteínas de Unión al ADN , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Endometrio/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Pronóstico
11.
BMB Rep ; 57(2): 122, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38416113

RESUMEN

[Erratum to: BMB Reports 2023; 56(3): 184-189, PMID: 36617466, PMCID: PMC10068343] The BMB Reports would like to correct in BMB Rep. 56(3): 184-189, titled "circRNA circSnx12 confers Cisplatin chemoresistance to ovarian cancer by inhibiting ferroptosis through a miR-194-5p/SLC7A11 axis". The original version of this article unfortunately contained image error in the Fig. 3. This article has been updated to correct an error in the image in Fig. 3D. The author apologizes for any inconvenience or confusion this error may cause. Author information has been modified in the original PDF version.

12.
Polymers (Basel) ; 16(7)2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38611124

RESUMEN

The antioxidant activity of chitosan (CS) and three water-soluble derivatives was analyzed comparatively by in vitro and in vivo experiments, including hydroxypropyl chitosan (HPCS), quaternary ammonium salt of chitosan (HACC), and carboxymethyl chitosan (CMCS). The results show that chitosan and its water-soluble derivatives have a scavenging ability on DPPH radicals, superoxide radicals, and hydroxyl radicals, and a reducing ability. A remarkable difference (p < 0.05) was found for HACC and HPCS compared with CS on DPPH radicals, hydroxyl radicals, and reducing ability. The antioxidant ability of the four chitosan samples was in the order of HPCS > HACC > CMCS > CS. Furthermore, antioxidant activity of all samples increased gradually in a concentration-dependent manner. The in vivo result indicates that oral CS and its derivatives samples result in a decrease in lipid peroxides (LPO) and free fatty acids (FFA) levels in serum with an increase in superoxide dismutase (SOD) activity. Especially for the HPCS and HACC groups, the LPO, FFA, and SOD activity in serum was different significantly in comparison with the high-fat controlgroup (HF) (p < 0.05). These results indicate that chitosan and its derivatives can be used as good antioxidants, and the antioxidant activity might be related to the molecular structure of chitosan derivatives.

13.
Artículo en Inglés | MEDLINE | ID: mdl-38512300

RESUMEN

Background: Long noncoding RNAs (lncRNAs), as emerging regulators of a wide variety of biological processes via diverse mechanisms, have been demonstrated to be of increasing importance in biology. Genome-wide association studies of tumor samples have identified several lncRNAs as either oncogenes or tumor suppressors in various types of cancers. In recent years, the importance of lncRNAs, especially in endometrioid cancer (EEC), has become increasingly well understood. The lncRNA Forkhead box P4 antisense RNA 1 (FOXP4-AS1) has been reported to fulfill roles in several types of cancers; however, the main biological function and associated underlying molecular mechanism of FOXP4-AS1 in EEC have yet to be fully elucidated. Materials and Methods: The present study therefore aimed to investigate how RNA FOXP4-AS1 may participate in the development and progression of endometrioid carcinoma tissues. To meet this aim, in the present study, the expression level of FOXP4-AS1 was investigated in endometrioid carcinoma tissues and matching nearby normal endometrial tissues collected from patients receiving surgery at the hospital, and a series of molecular biological assays were performed to investigate the effect of FOXP4-AS1 on cell proliferation, cell migration, and cell invasion, and so on. Results: An increased concentration of FOXP4-AS1 was identified in endometrioid carcinoma samples and cell lines compared with the corresponding controls, and this lncRNA was found to be positively correlated with advanced FIGO stages in patients with endometrial cancer. Furthermore, knocking down endogenous FOXP4-AS1 led to a significant reduction in the colony formation number and a significant inhibition of cell proliferation, cell migration, and cell invasion in endometrioid carcinoma cells. Moreover, dual-specificity phosphatase 5 (DUSP5), which is lowly expressed in endometrioid carcinoma tissues cells and negatively modulated by FOXP4-AS1, was identified as the downstream target molecule of FOXP4-AS1. Subsequently, the mechanistic experiments confirmed that, through binding to enhancer of zeste homolog 2 (EZH2; one of the catalytic subunits of polycomb repressive complex 2 [PRC2]), FOXP4-AS1 could epigenetically suppress the expression of DUSP5. Finally, the oncogenic function of the FOXP4-AS1/EZH2/DUSP5 axis in endometrioid carcinoma was confirmed via rescue assays. Conclusions: The findings of the present study have highlighted how FOXP4-AS1 fulfills an oncogenic role in endometrioid carcinoma, and targeting FOXP4-AS1 and its pathway may provide new biomarkers for patients with endometrioid carcinoma.

14.
Sci Rep ; 14(1): 13794, 2024 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-38877066

RESUMEN

Mayo Imaging Classification (MIC) for predicting future kidney growth in autosomal dominant polycystic kidney disease (ADPKD) patients is calculated from a single MRI/CT scan assuming exponential kidney volume growth and height-adjusted total kidney volume at birth to be 150 mL/m. However, when multiple scans are available, how this information should be combined to improve prediction accuracy is unclear. Herein, we studied ADPKD subjects ( n = 36 ) with 8+ years imaging follow-up (mean = 11 years) to establish ground truth kidney growth trajectory. MIC annual kidney growth rate predictions were compared to ground truth as well as 1- and 2-parameter least squares fitting. The annualized mean absolute error in MIC for predicting total kidney volume growth rate was 2.1 % ± 2 % compared to 1.1 % ± 1 % ( p = 0.002 ) for a 2-parameter fit to the same exponential growth curve used for MIC when 4 measurements were available or 1.4 % ± 1 % ( p = 0.01 ) with 3 measurements averaging together with MIC. On univariate analysis, male sex ( p = 0.05 ) and PKD2 mutation ( p = 0.04 ) were associated with poorer MIC performance. In ADPKD patients with 3 or more CT/MRI scans, 2-parameter least squares fitting predicted kidney volume growth rate better than MIC, especially in males and with PKD2 mutations where MIC was less accurate.


Asunto(s)
Riñón , Imagen por Resonancia Magnética , Riñón Poliquístico Autosómico Dominante , Humanos , Riñón Poliquístico Autosómico Dominante/diagnóstico por imagen , Riñón Poliquístico Autosómico Dominante/patología , Riñón Poliquístico Autosómico Dominante/fisiopatología , Masculino , Femenino , Riñón/diagnóstico por imagen , Riñón/patología , Análisis de los Mínimos Cuadrados , Adulto , Tamaño de los Órganos , Imagen por Resonancia Magnética/métodos , Persona de Mediana Edad , Tomografía Computarizada por Rayos X/métodos
15.
Food Chem ; 457: 140124, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38908239

RESUMEN

Phenolics in bound form extensively exist in cereal dietary fiber, especially insoluble fiber, while their release profile in gastrointestinal tract and contribution to the potential positive effects of dietary fiber in modulating gut microbiota still needs to be disclosed. In this work, the composition of bound phenolics (BPs) in triticale insoluble dietary fiber (TIDF) was studied, and in vitro gastrointestinal digestion as well as colonic fermentation were performed to investigate BPs liberation and their role in regulating intestinal flora of TIDF. It turned out that most BPs were unaccessible in digestion but partly released continuously during fermentation. 16 s rRNA sequencing demonstrated that TIDF possessed prebiotic effects by promoting anti-inflammatory while inhibiting proinflammatory bacteria alongside boosting SCFAs production and antioxidative BPs contributed a lot to these effects. Results indicated that TIDF held capabilities to regulate intestinal flora and BPs were important functional components to the health benefits of cereal dietary fiber.

16.
Cell Prolif ; : e13691, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38886174

RESUMEN

The regulatory mechanisms involved in embryonic development are complex and yet remain unclear. SCP4 represents a novel nucleus-resident phosphatase identified in our previous study. The primary aim of this study was to elucidate the function of SCP4 in the progress of cartilage development and endochondral osteogenesis. SCP4-/- and SCP4Col2ER mice were constructed to assess differences in bone formation using whole skeleton staining. ABH/OG staining was used to compare chondrocyte differentiation and cartilage development. Relevant biological functions were analysed using RNA-sequencing and GO enrichment, further validated by immunohistochemical staining, Co-IP and Western Blot. Global SCP4 knockout led to abnormal embryonic development in SCP4-/- mice, along with delayed endochondral osteogenesis. In parallel, chondrocyte-specific removal of SCP4 yielded more severe embryonic deformities in SCP4Col2ER mice, including limb shortening, reduced chondrocyte number in the growth plate, disorganisation and cell enlargement. Moreover, RNA-sequencing analysis showed an association between SCP4 and chondrocyte apoptosis. Notably, Tunnel-positive cells were indeed increased in the growth plates of SCP4Col2ER mice. The deficiency of SCP4 up-regulated the expression levels of pro-apoptotic proteins both in vivo and in vitro. Additionally, phosphorylation of FoxO3a (pFoxO3a), a substrate of SCP4, was heightened in chondrocytes of SCP4Col2ER mice growth plate, and the direct interaction between SCP4 and pFoxO3a was further validated in chondrocytes. Our findings underscore the critical role of SCP4 in regulating cartilage development and endochondral osteogenesis during embryonic development partially via inhibition of chondrocytes apoptosis regulated by FoxO3a dephosphorylation.

17.
Medicine (Baltimore) ; 102(40): e35406, 2023 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-37800830

RESUMEN

BACKGROUND: Endometriosis is a chronic inflammatory, benign disorder that often co-occurs with adenomyosis and/or leiomyoma. The overall incidence of endometriosis in reproductive period women was nearly 10%. However, the exact mechanisms of endometriosis-associated pathogenesis are still unknown. METHODS: In this study, we aimed to investigate whether Frizzled-7 (FZD7) would effectively promote the development of endometriosis. The microarray-based data analysis was performed to screen endometriosis-related differentially expressed genes. This process uncovered specific hub genes, and the nexus of vital genes and ferroptosis-related genes were pinpointed. Then, we collected human endometrial and endometriotic tissues from patients with endometriosis of the ovary (n = 39) and control patients without endometriosis (n = 10, who underwent hysterectomy for uterine fibroids) to compare the expression of FZD7. RESULTS: These findings indicated that the expression of FZD7 was high compared with normal endometrium, and FZD7 may promote the progression of endometriosis. CONCLUSION: FZD7 may serve as a potential therapeutic target for endometriosis treatment.


Asunto(s)
Endometriosis , Femenino , Humanos , Biomarcadores/metabolismo , Endometriosis/genética , Endometriosis/metabolismo , Endometriosis/patología , Endometrio/metabolismo , Endometrio/patología , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Leiomioma/patología , Ovario/patología
18.
BMB Rep ; 56(7): 416, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37500559

RESUMEN

[Erratum to: BMB Reports 2023; 56(3): 184-189, PMID: 36617466, PMCID: PMC10068343] The BMB Reports would like to correct in BMB Rep. 56(3): 184-189, titled "circRNA circSnx12 confers Cisplatin chemoresistance to ovarian cancer by inhibiting ferroptosis through a miR-194-5p/SLC7A11 axis". This research has the wrong affiliation of the authors and number of affiliation. Since author's affiliation is incorrect, this information has now been corrected as follows. Kaiyun Qin1,3,#, Fenghua Zhang2,#, Hongxia Wang1, Na Wang1, Hongbing Qiu4, Xinzhuan Jia1,5, Shan Gong1 & Zhengmao Zhang1,* 1Department of Gynecology, Fourth Hospital of Hebei Medical University, Hebei Shijiazhuang 050011, 2Department of Breast & Thyroid Surgery, Hebei General Hospital, Hebei Shijiazhuang 050057, 3Department of Gynecology, Hebei General Hospital, Hebei Shijiazhuang 050057, 4Department of Gynecology, Hebei Xingtai People's Hospital, Hebei Shijiazhuang 054001, 5Department of Reproductive Medicine, Fourth Hospital of Hebei Medical University, Hebei Shijiazhuang 050011, China The author apologizes for any inconvenience or confusion this error may cause. Author information has been modified in the original PDF version.

19.
BMB Rep ; 56(2): 184-189, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36617466

RESUMEN

Ovarian cancer (OC) is the most common gynecological malignancy worldwide, and chemoresistance occurs in most patients, resulting in treatment failure. A better understanding of the molecular processes underlying drug resistance is crucial for development of efficient therapies to improve OC patient outcomes. Circular RNAs (circRNAs) and ferroptosis play crucial roles in tumorigenesis and resistance to chemotherapy. However, little is known about the role(s) of circRNAs in regulating ferroptosis in OC. To gain insights into cisplatin resistance in OC, we studied the ferroptosis-associated circRNA circSnx12. We evaluated circSnx12 expression in OC cell lines and tissues that were susceptible or resistant to cisplatin using quantitative real-time PCR. We also conducted in vitro and in vivo assays examining the function and mechanism of lnc-LBCSs. Knockdown of circSnx12 rendered cisplatin-resistant OC cells more sensitive to cisplatin in vitro and in vivo by activating ferroptosis, which was at least partially abolished by downregulation of miR-194-5p. Molecular mechanics studies indicate that circSnx12 can be a molecular sponge of miR-194-5p, which targets SLC7A11. According to our findings, circSnx12 ameliorates cisplatin resistance by blocking ferroptosis via a miR-194-5p/SLC7A11 pathway. CircARNT2 may thus serve as an effective therapeutic target for overcoming cisplatin resistance in OC. [BMB Reports 2023; 56(3): 184-189].


Asunto(s)
Ferroptosis , MicroARNs , Neoplasias Ováricas , Humanos , Femenino , Cisplatino/farmacología , Cisplatino/uso terapéutico , ARN Circular/genética , MicroARNs/genética , MicroARNs/metabolismo , Resistencia a Antineoplásicos/genética , Ferroptosis/genética , Línea Celular Tumoral , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Sistema de Transporte de Aminoácidos y+/genética
20.
Open Med (Wars) ; 18(1): 20230729, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37333452

RESUMEN

The miR-141-3p has been reported to participate in regulating autophagy and tumor-stroma interactions in ovarian cancer (OC). We aim to investigate whether miR-141-3p accelerates the progression of OC and its effect on macrophage 2 polarization by targeting the Kelch-like ECH-associated protein1-Nuclear factor E2-related factor2 (Keap1-Nrf2) pathway. SKOV3 and A2780 cells were transfected with miR-141-3p inhibitor and negative control to confirm the regulation of miR-141-3p on OC development. Moreover, the growth of tumors in xenograft nude mice treated by cells transfected with miR-141-3p inhibitor was established to further testify the role of miR-141-3p in OC. The expression of miR-141-3p was higher in OC tissue compared with non-cancerous tissue. Downregulation of miR-141-3p inhibited the proliferation, migration, and invasion of ovarian cells. Furthermore, miR-141-3p inhibition also suppressed M2-like macrophage polarization and in vivo OC progression. Inhibition of miR-141-3p significantly enhanced the expression of Keap1, the target gene of miR-141-3p, and thus downregulated Nrf2, while activation of Nrf2 reversed the reduction in M2 polarization by miR-141-3p inhibitor. Collectively, miR-141-3p contributes to tumor progression, migration, and M2 polarization of OC by activating the Keap1-Nrf2 pathway. Inhibition of miR-141-3p attenuates the malignant biological behavior of ovarian cells by inactivating the Keap1-Nrf2 pathway.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA