Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Cell Physiol ; 239(4): e31189, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38219074

RESUMEN

Joubert syndrome (JBTS) is a systematic developmental disorder mainly characterized by a pathognomonic mid-hindbrain malformation. All known JBTS-associated genes encode proteins involved in the function of antenna-like cellular organelle, primary cilium, which plays essential roles in cellular signal transduction and development. Here, we identified four unreported variants in ARL13B in two patients with the classical features of JBTS. ARL13B is a member of the Ras GTPase family and functions in ciliogenesis and cilia-related signaling. The two missense variants in ARL13B harbored the substitutions of amino acids at evolutionarily conserved positions. Using model cell lines, we found that the accumulations of the missense variants in cilia were impaired and the variants showed attenuated functions in ciliogenesis or the trafficking of INPP5E. Overall, these findings expanded the ARL13B pathogenetic variant spectrum of JBTS.


Asunto(s)
Factores de Ribosilacion-ADP , Anomalías Múltiples , Cerebelo , Anomalías del Ojo , Enfermedades Renales Quísticas , Retina , Humanos , Anomalías Múltiples/genética , Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/metabolismo , Cerebelo/anomalías , Cilios/genética , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Monoéster Fosfórico Hidrolasas/metabolismo , Retina/metabolismo , Retina/anomalías , Masculino , Femenino , Lactante
2.
J Nanobiotechnology ; 22(1): 299, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38812031

RESUMEN

BACKGROUND: Discrepancies in the utilization of reactive oxygen species (ROS) between cancer cells and their normal counterparts constitute a pivotal juncture for the precise treatment of cancer, delineating a noteworthy trajectory in the field of targeted therapies. This phenomenon is particularly conspicuous in the domain of nano-drug precision treatment. Despite substantial strides in employing nanoparticles to disrupt ROS for cancer therapy, current strategies continue to grapple with challenges pertaining to efficacy and specificity. One of the primary hurdles lies in the elevated levels of intracellular glutathione (GSH). Presently, predominant methods to mitigate intracellular GSH involve inhibiting its synthesis or promoting GSH efflux. However, a conspicuous gap remains in the absence of a strategy capable of directly and efficiently clearing GSH. METHODS: We initially elucidated the chemical mechanism underpinning oridonin, a diminutive pharmacological agent demonstrated to perturb reactive oxygen species, through its covalent interaction with glutathione. Subsequently, we employed the incorporation of maleimide-liposomes, renowned for their capacity to disrupt the ROS delivery system, to ameliorate the drug's water solubility and pharmacokinetics, thereby enhancing its ROS-disruptive efficacy. In a pursuit to further refine the targeting for acute myeloid leukemia (AML), we harnessed the maleic imide and thiol reaction mechanism, facilitating the coupling of Toll-like receptor 2 (TLR2) peptides to the liposomes' surface via maleic imide. This strategic approach offers a novel method for the precise removal of GSH, and its enhancement endeavors are directed towards fortifying the precision and efficacy of the drug's impact on AML targets. RESULTS: We demonstrated that this peptide-liposome-small molecule machinery targets AML and consequently induces cell apoptosis both in vitro and in vivo through three disparate mechanisms: (I) Oridonin, as a Michael acceptor molecule, inhibits GSH function through covalent bonding, triggering an initial imbalance of oxidative stress. (II) Maleimide further induces GSH exhaustion, aggravating redox imbalance as a complementary augment with oridonin. (III) Peptide targets TLR2, enhances the directivity and enrichment of oridonin within AML cells. CONCLUSION: The rationally designed nanocomplex provides a ROS drug enhancement and targeted delivery platform, representing a potential solution by disrupting redox balance for AML therapy.


Asunto(s)
Diterpenos de Tipo Kaurano , Glutatión , Leucemia Mieloide Aguda , Liposomas , Especies Reactivas de Oxígeno , Diterpenos de Tipo Kaurano/química , Diterpenos de Tipo Kaurano/farmacología , Glutatión/metabolismo , Glutatión/química , Liposomas/química , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Humanos , Especies Reactivas de Oxígeno/metabolismo , Animales , Ratones , Línea Celular Tumoral , Receptor Toll-Like 2/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Apoptosis/efectos de los fármacos
3.
Exp Cell Res ; 411(1): 113000, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34958764

RESUMEN

The accumulation of activated myofibroblastic pancreatic stellate cells (MF-PSCs) induces pancreatic cancer desmoplasia. These MF-PSCs are derived from quiescent pancreatic stellate cells (Q-PSCs). MF-PSCs in pancreatic cancer tend to glycolysis. However, increased glycolysis alone could not be sufficient for the increased metabolic demands of MF-PSCs. Yap and Myc signaling activation is involved in pancreatic cancer metabolism. Since elucidating the metabolic processes of MF-PSCs may be a promising strategy to suppress pancreatic cancer desmoplasia, we explored whether glutaminolysis meets the bioenergetic and biosynthetic demands of Q-PSCs converted into MF-PSCs and whether this is mediated by Yap signaling to Myc. In this study, we found that during the transdifferentiation of Q-PSCs into MF-PSCs, glutaminolysis regulatory genes were upregulated, and suppression of glutaminolysis inhibited transdifferentiation. Disrupting glutaminolysis in MF-PSCs inhibited cell growth, mitochondrial respiration, and fibrogenesis, while treatment of MF-PSCs with DKG (a glutaminolysis metabolite) reversed these activities. The expression of glutaminase (GLS1), a rate-limiting enzyme in glutaminolysis, was upregulated by Yap overexpression. Yap upregulates Myc to regulate the expression of GLS1 in MF-PSCs. Yap and Myc inhibitors disrupted glutaminolysis and inhibited myofibroblastic activities in PSCs. Thus, Yap-Myc signaling controls glutaminolysis to activate PSCs and might be a therapeutic target for pancreatic cancer desmoplasia.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Transdiferenciación Celular , Glutamina/metabolismo , Glucólisis , Células Estrelladas Pancreáticas/citología , Células Estrelladas Pancreáticas/fisiología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Ciclo Celular/genética , Proliferación Celular , Humanos , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal , Factores de Transcripción/genética
4.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 40(1): 57-61, 2023 Jan 10.
Artículo en Zh | MEDLINE | ID: mdl-36585002

RESUMEN

OBJECTIVE: To explore the genetic basis for a child with mental retardation. METHODS: Whole exome sequencing was carried out for the child. Candidate variant was screened based on his clinical features and verified by Sanger sequencing. RESULTS: The child was found to harbor a c.995_1002delAGACAAAA(p.Asp332AlafsTer84) frameshift variant in the SYNGAP1 gene. Bioinformatic analysis suggested it to be pathogenic. The same variant was not detected in either parent. CONCLUSION: The c.995_1002delAGACAAAA(p.Asp332AlafsTer84) frameshift variant of the SYNGAP1 gene probably underlay the mental retardation in this child. Above finding has expanded the spectrum of SYNGAP1 gene variants and provided a basis for the diagnosis and treatment for this child.


Asunto(s)
Discapacidad Intelectual , Niño , Humanos , Discapacidad Intelectual/genética , Mutación del Sistema de Lectura , Secuenciación de Nucleótidos de Alto Rendimiento , Biología Computacional , Heterocigoto , Mutación , Proteínas Activadoras de ras GTPasa/genética
5.
Carcinogenesis ; 43(12): 1162-1175, 2022 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-36194598

RESUMEN

The Notch1 (Notch1 receptor) and yes-associated protein 1 (YAP1) signaling can regulate breast cancer metastasis. This study aimed at investigating whether and how these two signal pathways crosstalk to promote breast cancer lung metastasis. Here, we show that YAP1 expression was positively correlated with Notch1 in breast cancer according to bioinformatics and experimental validation. Mechanistically, YAP1 with TEA domain transcription factors (TEADs) enhanced Jagged1(JAG1)-Notch1 signaling. Meanwhile, Notch1 promoted YAP1 stability in breast cancer cells by inhibiting the ß-TrCP-mediated degradation, thereby, forming a YAP1- JAG1/Notch1 positive feedback loop in breast cancer. Furthermore, YAP1 enhanced the mammosphere formation and stemness of MDA-MB-231 cells by attenuating the inhibition of the BMP4-SMAD1/5 signaling. In vivo, the YAP1- JAG1/Notch1 positive feedback loop promoted the lung colonization of MDA-MB-231 cells. Our data for the first time indicate that the YAP1-Notch1 positive feedback loop promotes lung metastasis of breast cancer by modulating self-renewal and inhibiting the BMP4-SMAD1/5 signaling.


Asunto(s)
Neoplasias de la Mama , Neoplasias Pulmonares , Humanos , Femenino , Receptor Notch1/genética , Receptor Notch1/metabolismo , Neoplasias de la Mama/patología , Proteínas Señalizadoras YAP , Retroalimentación , Proteína Morfogenética Ósea 4/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Neoplasias Pulmonares/genética , Familia , Línea Celular Tumoral
6.
Crit Rev Food Sci Nutr ; : 1-19, 2022 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-36052610

RESUMEN

Calcium is one of the important elements for human health. Calcium deficiencies can lead to numerous diseases. Calcium chelating peptides have shown potential application in the management of calcium deficiencies. Casein phosphopeptides (CPP) are phosphoseryl-containing fragments of casein by enzymatic hydrolysis or fermentation during manufacture of milk products as well as during intestinal digestion. An increasing number of CPP with the ability to facilitate and enhance the bioavailability of calcium are being discovered and identified. In this review, 249 reported CPP derived from four types of bovine casein (αs1, αs2, ß and κ) were collected, and the amino acid sequence and phosphoserine group information were sorted out. This review outlines the current enzyme hydrolysis, detection methods, purification, structure-activity relationship and mechanism of intestinal calcium absorption in vitro and in vivo as well as application of CPP.

7.
Genet Med ; 23(6): 1041-1049, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33531668

RESUMEN

PURPOSE: Ciliopathies are a group of disorders caused by defects of the cilia. Joubert syndrome (JBTS) is a recessive and pleiotropic ciliopathy that causes cerebellar vermis hypoplasia and psychomotor delay. Although the intraflagellar transport (IFT) complex serves as a key module to maintain the ciliary structure and regulate ciliary signaling, the function of IFT in JBTS remains largely unknown. We aimed to explore the impact of IFT dysfunction in JBTS. METHODS: Exome sequencing was performed to screen for pathogenic variants in IFT genes in a JBTS cohort. Animal model and patient-derived fibroblasts were used to evaluate the pathogenic effects of the variants. RESULTS: We identified IFT74 as a JBTS-associated gene in three unrelated families. All the affected individuals carried truncated variants and shared one missense variant (p.Q179E) found only in East Asians. The expression of the human p.Q179E-IFT74 variant displayed compromised rescue effects in zebrafish ift74 morphants. Attenuated ciliogenesis; altered distribution of IFT proteins and ciliary membrane proteins, including ARL13B, INPP5E, and GPR161; and disrupted hedgehog signaling were observed in patient fibroblasts with IFT74 variants. CONCLUSION: IFT74 is identified as a JBTS-related gene. Cellular and biochemical mechanisms are also provided.


Asunto(s)
Anomalías Múltiples , Anomalías del Ojo , Enfermedades Renales Quísticas , Anomalías Múltiples/genética , Animales , Cerebelo/anomalías , Proteínas del Citoesqueleto , Anomalías del Ojo/genética , Proteínas Hedgehog , Humanos , Enfermedades Renales Quísticas/genética , Monoéster Fosfórico Hidrolasas/genética , Retina/anomalías , Pez Cebra/genética
8.
FASEB J ; 34(8): 10860-10870, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32592239

RESUMEN

The tumor microenvironment (TME) is a crucial factor in cancer progression. In breast cancer, cancer-associated fibroblasts (CAFs) and the derived stromal components have been recognized as comprising the majority of the pathological structure of the TME. In this study, we show that metformin (Met), a diabetes drug, transforms CAFs in the TME. Met disrupts tumor-stromal cross talk by preventing breast cancer cell transforming growth factor-ß (TGF-ß) signaling and the production of stromal-derived factor-1 (SDF-1) and interleukin-8 (IL-8) by CAFs. The suppression of bidirectional signaling between tumor cells and CAFs by Met is attributed to increased phospho-AMP kinase (p-AMPK) levels. By upregulating p-AMPK in CAFs, Met induces prolyl hydroxylases (PHDs), leading to the degradation of hypoxia-inducible factor-1α (HIF-1α) in CAFs. Moreover, interruption of HIF-1α-driven SDF-1 signaling in CAFs by Met leads to decreased breast cancer cell invasion. These findings suggest that Met may be used to target tumor-promoting signaling between CAFs and breast cancer cells in the TME.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metformina/farmacología , Adenilato Quinasa/metabolismo , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Quimiocina CXCL12/metabolismo , Femenino , Humanos , Interleucina-8/metabolismo , Células MCF-7 , Invasividad Neoplásica/patología , Prolil Hidroxilasas/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
9.
BMC Med Genet ; 21(1): 192, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33004012

RESUMEN

BACKGROUND: Joubert syndrome (OMIM 213300) is an autosomal recessive disorder with gene heterogeneity. Causal genes and their variants have been identified by sequencing or other technologies for Joubert syndrome subtypes. CASE PRESENTATION: A two-year-old boy was diagnosed with Joubert syndrome by global development delay and molar tooth sign of mid-brain. Whole exome sequencing was performed to detect the causative gene variants in this individual, and the candidate pathogenic variants were verified by Sanger sequencing. We identified two pathogenic variants (NM_006346.2: c.1147delC and c.1054A > G) of PIBF1 in this Joubert syndrome individual, which is consistent with the mode of autosomal recessive inheritance. CONCLUSION: In this study, we identified two novel pathogenic variants in PIBF1 in a Joubert syndrome individual using whole exome sequencing, thereby expanding the PIBF1 pathogenic variant spectrum of Joubert syndrome.


Asunto(s)
Anomalías Múltiples/genética , Cerebelo/anomalías , Secuenciación del Exoma/métodos , Anomalías del Ojo/genética , Predisposición Genética a la Enfermedad/genética , Enfermedades Renales Quísticas/genética , Mutación , Proteínas Gestacionales/genética , Retina/anomalías , Factores Supresores Inmunológicos/genética , Anomalías Múltiples/diagnóstico , Preescolar , Anomalías del Ojo/diagnóstico , Genes Recesivos , Humanos , Enfermedades Renales Quísticas/diagnóstico , Masculino
10.
Clin Exp Pharmacol Physiol ; 47(3): 449-458, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31665807

RESUMEN

Deregulation of microRNAs (miRNAs) leads to malignant growth and aggressive invasion during cancer occurrence and progression. miR-147b has emerged as one of the cancer-related miRNAs that are dysregulated in multiple cancers. Yet, the relevance of miR-147b in non-small-cell lung cancer (NSCLC) remains unclear. In the present study, we aimed to report the biological function and signalling pathways mediated by miR-147b in NSCLC. Our results demonstrate that miR-147b expression is significantly downregulated in NSCLC tissues and cell lines. Overexpression of miR-147b decreased the proliferative ability, colony-forming capability, and invasive potential of NSCLC cells. Notably, our study identified ribosomal protein S15A (RPS15A), an oncogene in NSCLC, as a target gene of miR-147b. Our results showed that miR-147b negatively modulates RPS15A expression in NSCLC cells. An inverse correlation between miR-147b and RPS15A was evidenced in NSCLC specimens. Moreover, miR-147b overexpression downregulated the activation of Wnt/ß-catenin signalling via targeting of RPS15A. Overexpression of RPS15A partially reversed the miR-147b-mediated antitumour effect in NSCLC cells. Collectively, these findings reveal that miR-147b restricts the proliferation and invasion of NSCLC cells by inhibiting RPS15A-induced Wnt/ß-catenin signalling and suggest that the miR-147b/RPS15A/Wnt/ß-catenin axis is an important regulatory mechanism for malignant progression of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proliferación Celular/fisiología , Neoplasias Pulmonares/metabolismo , MicroARNs/biosíntesis , Proteínas Ribosómicas/biosíntesis , Vía de Señalización Wnt/fisiología , Células A549 , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/prevención & control , Regulación hacia Abajo/fisiología , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/prevención & control , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control
11.
Proc Natl Acad Sci U S A ; 114(29): 7683-7688, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28674022

RESUMEN

Fibrous sheath interacting protein 1 (FSIP1), a spermatogenesis-related testicular antigen, is expressed in abundance in breast cancers, particularly in those overexpressing human epidermal growth factor receptor 2 (HER2); however, little is known about its role in regulating the growth and metastasis of breast cancer cells. We and others have shown previously that FSIP1 expression in breast cancer correlates positively with HER2-positivity, recurrence, and metastases and negatively with survival. Here, using coimmunoprecipitation and microscale thermophoresis, we find that FSIP1 binds to the intracellular domain of HER2 directly. We further show that shRNA-induced FSIP1 knockdown in SKBR3 and MCF-7 breast cancer cells inhibits proliferation, stimulates apoptosis, attenuates epithelial-mesenchymal transition, and impairs migration and invasiveness. Consistent with reduced proliferation and enhanced apoptosis, xenotransplantation of SKBR3 cells stably transfected with sh-FSIP1 into nu/nu mice results in reduced tumor volumes compared with sh-NC transplants. Furthermore, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) mapping using sh-FSIP1 gene signature yielded associations with extracellular matrix protein pathways, and a reduction in SNAI2 protein expression was confirmed on Western blot analysis. Complementarily, interrogation of the Connectivity Map using the same gene signature yielded, as top hits, chemicals known to inhibit epithelial-mesenchymal transition, including rapamycin, 17-N-allylamino-17-demethoxygeldanamycin, and LY294002. These compounds phenocopy the effects of sh-FSIP1 on SKBR3 cell viability. Thus, FSIP1 suppression limits oncogenesis and invasiveness in breast cancer cells and, considering its absence in most other tissues, including normal breast, may become a potential target for breast cancer therapy.


Asunto(s)
Apoptosis , Neoplasias de la Mama/metabolismo , Proteínas Portadoras/metabolismo , Regulación Neoplásica de la Expresión Génica , Receptor ErbB-2/metabolismo , Proteínas de Plasma Seminal/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Recurrencia Local de Neoplasia/genética , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , Factores de Transcripción/metabolismo
12.
Hum Reprod ; 32(1): 248-255, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27836978

RESUMEN

STUDY QUESTION: Does a novel homozygous NOBOX truncating variant, identified in whole exome sequencing (WES) of patients with primary ovarian insufficiency (POI), cause defective transcriptional activation of multiple oocyte-related genes? SUMMARY ANSWER: A novel homozygous truncating mutation of NOBOX was confirmed to exhibit a loss-of-function effect using well-defined molecular and functional analyses. WHAT IS KNOWN ALREADY: Several NOBOX mutations have been reported to be associated with POI but all of them are heterozygous mutations. STUDY DESIGN, SIZE, DURATION: This is a cross sectional study in 96 patients diagnosed with POI and 211 women not diagnosed with POI in China. PARTICIPANTS/MATERIALS, SETTING, METHODS: Blood samples collected from the participants were subjected to whole exome sequencing. Full-length transcript of NOBOX was cloned directly from human fetal ovary (FO). Functional analysis was performed for a NOBOX sequence variant associated with POI. MAIN RESULTS AND THE ROLE OF CHANCE: One novel homozygous truncating variant, chr7:144098161delC, in the NOBOX gene was found in a POI patient. The truncating variant showed a severe defect in transcriptional activation of GDF9 a well-known target NOBOX. Furthermore, using real-time quantitative PCR analysis, we found many oocyte-related genes were expressed at lower level in truncating variant cells than in control cells. In addition, we found that the truncated NOBOX lost its ability to induce the G2/M arrest.Notably, our results confirmed that the 1725 bp NOBOX transcript is expressed in human FO and is the only functional isoform in transcriptional activation assays. LIMITATIONS REASONS FOR CAUTION: Although the in vitro assays demonstrated the loss-of-function effect of truncating mutation on NOBOX transcriptional activation, further studies are needed to validate its long-term effects on folliculogenesis and POI. WIDER IMPLICATIONS OF THE FINDINGS: This is the first homozygous mutation of NOBOX associated with POI showing a loss-of-function effect using well-defined molecular and functional analyses. These results will aid both researchers and clinicians in understanding the molecular pathology of NOBOX and POI to develop diagnostic assays or therapeutic approaches. STUDY FUNDING/COMPETING INTERESTS: Research funding is provided by the Ministry of Science and Technology of China [2012CB944704; 2012CB966702], the National Natural Science Foundation of China [Grant number: 31171429] and Beijing Advanced Innovation Center for Structural Biology. The authors declare no conflict of interest.


Asunto(s)
Proteínas de Homeodominio/genética , Mutación , Insuficiencia Ovárica Primaria/genética , Factores de Transcripción/genética , Activación Transcripcional/genética , Adulto , China , Estudios Transversales , Femenino , Hormona Folículo Estimulante/sangre , Estudios de Asociación Genética , Humanos , Insuficiencia Ovárica Primaria/sangre , Adulto Joven
14.
Reprod Biomed Online ; 32(4): 388-93, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26896429

RESUMEN

Homeobox A10 (HOXA10) and empty spiracles homeobox 2 (EMX2) are two transcription factors necessary for female Müllerian duct differentiation and development. They are thought to play important roles in embryo implantation in mice and humans. The EMX2 gene is a known direct target of HOXA10 in the reproductive tract. Human TENM1 is directly regulated by EMX2 and is expressed during embryonic pattern formation and morphogenesis. This study aimed to investigate expression patterns of HOXA10, EMX2 and TENM1 in the mid-secretory endometrium of infertile patients with a Müllerian duct anomaly causing a partially septate uterus. Thirteen mid-secretory endometrial tissue samples were collected from women with partially septate uteri and 12 from women with normal uteri as controls. Expression levels of HOXA10, EMX2 and TENM1 mRNA and protein in the mid-secretory endometrium of infertile patients and controls were measured by quantitative reverse transcription polymerase chain reaction and western blotting. Compared with controls, mRNA and protein expression levels of HOXA10 decreased significantly (P < 0.01), whereas EMX2 and TENM1 increased dramatically in patients with Müllerian duct anomaly (P < 0.001). Changes in HOXA10, EMX2, and TENM1 expression levels might act in infertile women with Müllerian duct anomaly to cause a partially septate uterus.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Infertilidad/metabolismo , Fase Luteínica , Conductos Paramesonéfricos/anomalías , Proteínas del Tejido Nervioso/metabolismo , Tenascina/metabolismo , Factores de Transcripción/metabolismo , Western Blotting , Femenino , Proteínas Homeobox A10 , Proteínas de Homeodominio/genética , Humanos , Infertilidad/genética , Conductos Paramesonéfricos/embriología , Proteínas del Tejido Nervioso/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tenascina/genética , Factores de Transcripción/genética
15.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 47(5): 786-789, 2016 Sep.
Artículo en Zh | MEDLINE | ID: mdl-28598100

RESUMEN

OBJECTIVES: To construct a cDNA phage expression library for human esophageal cancer cells. METHODS: After the total RNA were obtained from esophageal cancer cells, the mRNA were separated with magnetic beads adsorption method, and the single-strand and double-strand cDNA were synthesized through reverse transcription. With the undesirable cDNA fragments removed, the remaining cDNA (linked withEcoR1 aptamer and phosphorylated its 5'end) combined with the carrier of T7 Select10-3b. The recombinant phage were packaged in vitro for preliminary cDNA library. PCR was used to identify the size of inserted cDNA. RESULTS: The constructed original cDNA phage expression library for human esophageal cancer cells was consisted of 2.01×106 pfu/mL bacteriophages with a recombination rate of 100%. The length of the inserted cDNA fragments were range from 300 bp to 1 500 bp. CONCLUSIONS: The cDNA phage expression library of human esophageal cell is successfully constructed to meet the currently recognized standards, and can be well used to screen cDNA-cloned genes of human esophageal cancer antigens by serological analysis of recombinantly expressed cDNA clone (SEREX).


Asunto(s)
ADN Complementario/genética , Neoplasias Esofágicas/genética , Biblioteca de Genes , Bacteriófagos , Humanos , Reacción en Cadena de la Polimerasa
16.
Mol Cancer ; 14: 28, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25645291

RESUMEN

BACKGROUND: The epithelial-mesenchymal transition (EMT) is crucial for the invasion and metastasis of breast cancer. However, how Notch signaling regulates the EMT process and invasion in breast cancer remains largely unknown. METHODS: The impact of Notch1 silencing by specific shRNAs on the EMT and invasion of human breast cancer MCF-7 and MDA-MB-231 cells as well as xenografts was tested by western blot, real-time polymerase chain reaction (RT-PCR), immunofluorescence, transwell, and immunohistochemistry assays. The effect of Slug silencing or upregulation on the EMT and invasion of breast cancer cells was analyzed, and the effect of Notch1 signaling on Slug expression was determined by the luciferase reporter assay. RESULTS: The Notch1 intracellular domain (N1ICD) and Jagged1 were expressed in breast cancer cells. Notch1 silencing reversed the spontaneous EMT process and inhibited the migration and invasion of breast cancer cells and the growth of xenograft breast cancers. The expression of N1ICD was upregulated significantly by Jagged1-mediated Notch signaling activation. Moreover, Jagged1-mediated Notch signaling promoted the EMT process, migration, and invasion of breast cancer cells, which were abrogated by Notch silencing. Furthermore, the N1ICD positively regulated the Slug expression by inducing Slug promoter activation. Importantly, the knockdown of Slug weakened the invasion ability of breast cancer cells and reversed the Jagged1-induced EMT process with significantly decreased expression of vimentin and increased expression of E-cadherin. In addition, Slug overexpression restored the Notch1 knockdown-suppressed EMT process. CONCLUSIONS: Our novel data indicate that Notch signaling positively regulates the EMT, invasion, and growth of breast cancer cells by inducing Slug expression. The Notch1-Slug signaling axis may represent a potential therapeutic target for breast cancer therapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Transición Epitelial-Mesenquimal , Receptor Notch1/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Neoplasias de la Mama/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Invasividad Neoplásica , Regiones Promotoras Genéticas , Receptor Notch1/genética , Proteínas Serrate-Jagged , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Activación Transcripcional
17.
Tumour Biol ; 36(4): 2601-5, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25424702

RESUMEN

Small proline-rich repeat protein 1A (SPRR1A) is a marker for terminal squamous cell differentiation. Previous studies showed that SPRR1A expression increases in squamous cell carcinoma of the skin, but decreases in esophageal squamous cell carcinoma. This study focuses on the expression of SPRR1A protein in breast cancers (BCs) in China. A total of 111 patients with histologically confirmed BC, who underwent radical surgery between January 2006 and September 2007 in China Medical University, were enrolled. The relationship between SPRR1A expression and clinicopathological factors as well as BC prognoses was also determined. Overall, SPRR1A expression was detected in more than half of the BC specimens by immunohistochemistry (56/111, 53.8%), but there was no significant difference between age groups (≥50 vs. <50 years) in terms of SPRR1A expression (P = 0.915), as well as no differences between SPRR1A expression and the clinical stage (0-I vs. II-III) or nodal status (P = 0.234 and 0.632, respectively). Moreover, human epidermal growth factor receptor 2 overexpression was not correlated with SPRR1A expression, whereas Ki67 was associated with SPRR1A expression (P = 0.155 and 0.028, respectively). Interestingly, SPRR1A expression was significantly associated with progesterone receptor-positive (P = 0.010) rather than estrogen receptor-positive (0.778) BCs. The 5-year survival rate in patients did not differ with the presence or absence of SPRR1A expression (P = 0.753), whereas the combination of SPRR1A expression, progesterone receptor status, and menopausal status allowed identification of a subgroup of BC patients with a good long-term prognosis. Thus, the SPRR1A status might play an important role in the prognosis of postmenopausal breast carcinoma patients, especially that of progesterone receptor-positive subgroups.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Neoplasias de la Mama/genética , Proteínas Ricas en Prolina del Estrato Córneo/biosíntesis , Receptores de Progesterona/genética , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Neoplasias de la Mama/patología , China , Proteínas Ricas en Prolina del Estrato Córneo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Persona de Mediana Edad , Pronóstico , Análisis de Supervivencia
18.
Tumour Biol ; 36(10): 8167-75, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25990455

RESUMEN

Antimicrobial peptides (AMPs) with Asn-Gly-Arg (NGR) motif have potent cytotoxicity, preferably against tumor cells due to their binding to CD13 on tumor cells. However, the importance of αvß3 expression for antitumor activity of AMPs containing NGR has not been clarified. This study was aimed at designing a new AMP containing NGR and testing their biological activity against different types of tumor cells with varying CD13 and αvß3 expression. We first synthesized the new AMP containing NGR motif (CK21), which effectively entered into CD13+ HT-1080, but less into CD13- αvß3+ MDA-MB-435 and further less into stable αvß3-silencing MDA-MB-435 cells. Furthermore, CK21 displayed dose-dependent antiproliferation against these tumor cells and induced cell cycling arrest at G2/M phases and apoptosis of these tumor cells. In addition, CK21 inhibited the invasion of these tumor cells in vitro and inhibited the growth of implanted tumor cells in vivo. Particularly, the antitumor effect of CK21 in CD13+ HT-1080 was stronger than that of CD13- αvß3+ MDA-MB-435 and much stronger than that of stable αvß3-silencing MDA-MB-435. Our data indicated that the new AMPs containing NGR had potent antitumor activity against CD13+ or αvß3+ tumor cells, preferably against CD13+ tumor cells, possibly through binding to CD13 or αvß3 on tumor cells.


Asunto(s)
Antibacterianos/farmacología , Antígenos CD13/metabolismo , Proliferación Celular/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Oligopéptidos/farmacología , Fragmentos de Péptidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Movimiento Celular/efectos de los fármacos , Citometría de Flujo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Int J Biol Sci ; 20(2): 446-463, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38169627

RESUMEN

Triple-negative breast cancer (TNBC) has long been considered a major clinical challenge due to its aggressive behavior and poor prognosis. Cancer stem cells (CSCs) are known as the main cells responsible for tumor origination, progression, recurrence and metastasis. Here, we report that M2-type tumor-associated macrophages (TAMs) contribute to cancer stemness in TNBC cells via the secretion of VEGFA. Reciprocally, elevated VEGFA expression by TAM-educated TNBC cells acts as a regulator of macrophage polarization, therefore constitute a feed-back loop between TNBC cells and TAMs. Mechanistically, VEGFA facilitates the CSC phenotype via the NRP-1 receptor and downstream GAPVD1/Wnt/ß-catenin signaling pathway in TNBC cells. Our study underscores the crosstalk between TNBC cells and TAMs mediated by VEGFA and further clarifies the role and underlying mechanisms of the VEGFA/NRP-1/GAPVD1 axis in regulating cancer stemness. We also document an immunosuppressive function of VEGFA in the tumor microenvironment (TME). Therefore, the present study indicates crosstalk between TNBC cells and TAMs induced by VEGFA and provides a potential implication for the combination of immunotherapy and VEGFA-targeted agents in TNBC therapy.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Línea Celular Tumoral , Macrófagos/metabolismo , Antineoplásicos/farmacología , Vía de Señalización Wnt , Microambiente Tumoral/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
J Agric Food Chem ; 72(18): 10355-10365, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38620073

RESUMEN

The genus Bifidobacterium has been widely used in functional foods for health promotion due to its beneficial effects on human health, especially in the gastrointestinal tract (GIT). In this study, we characterize the anti-inflammatory potential of the probiotic strain Bifidobacterium pseudocatenulatum G7, isolated from a healthy male adult. G7 secretion inhibited inflammatory response in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Moreover, oral administration of bacteria G7 alleviated the severity of colonic inflammation in dextran sulfate sodium (DSS)-treated colitis mice, which was evidenced by a decreased disease activity index (DAI) and enhanced structural integrity of the colon. The 16S rRNA gene sequencing result illustrated that the G7 alleviated DSS-induced gut microbiota dysbiosis, accompanied by the modulated bile acids and short-chain fatty acid (SCFA) levels. Overall, our results demonstrated the potential anti-inflammatory effects of Bifidobacterium pseudocatenulatum G7 on both in vitro and in vivo models, which provided a solid foundation for further development of a novel anti-inflammatory probiotic.


Asunto(s)
Antiinflamatorios , Bifidobacterium pseudocatenulatum , Colitis , Microbioma Gastrointestinal , Probióticos , Probióticos/administración & dosificación , Probióticos/farmacología , Ratones , Animales , Células RAW 264.7 , Masculino , Antiinflamatorios/administración & dosificación , Humanos , Colitis/microbiología , Colitis/terapia , Colitis/inducido químicamente , Bifidobacterium pseudocatenulatum/genética , Bifidobacterium pseudocatenulatum/química , Ratones Endogámicos C57BL , Macrófagos/inmunología , Ácidos Grasos Volátiles/metabolismo , Colon/microbiología , Colon/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA