Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50.578
Filtrar
Más filtros











Intervalo de año de publicación
1.
Mol Biol Rep ; 51(1): 712, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38824221

RESUMEN

INTRODUCTION: Coronary artery disease (CAD) in young adults can have devastating consequences. The cardiac developmental gene MEIS1 plays important roles in vascular networks and heart development. This gene effects on the regeneration capacity of the heart. Considering role of MEIS1 in cardiac tissue development and the progression of myocardial infarction this study investigated the expression levels of the MEIS1, HIRA, and Myocardin genes in premature CAD patients compared to healthy subjects and evaluated the relationships between these genes and possible inflammatory factors. METHODS AND RESULTS: The study conducted a case-control design involving 35 CAD patients and 35 healthy individuals. Peripheral blood mononuclear cells (PBMCs) were collected, and gene expression analysis was performed using real-time PCR. Compared with control group, the number of PBMCs in the CAD group exhibited greater MEIS1 and HIRA gene expression, with fold changes of 2.45 and 3.6. The expression of MEIS1 exhibited a negative correlation with IL-10 (r= -0.312) expression and positive correlation with Interleukin (IL)-6 (r = 0.415) and tumor necrosis factor (TNF)-α (r = 0.534) gene expression. Moreover, there was an inverse correlation between the gene expression of HIRA and that of IL-10 (r= -0.326), and a positive correlation was revealed between the expression of this gene and that of the IL-6 (r = 0.453) and TNF-α (r = 0.572) genes. CONCLUSION: This research demonstrated a disparity in expression levels of MEIS1, HIRA, and Myocardin, between CAD and healthy subjects. The results showed that, MEIS1 and HIRA play significant roles in regulating the synthesis of proinflammatory cytokines, namely, TNF-α and IL-6.


Asunto(s)
Enfermedad de la Arteria Coronaria , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas Nucleares , Transactivadores , Humanos , Enfermedad de la Arteria Coronaria/genética , Femenino , Masculino , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Estudios de Casos y Controles , Adulto , Persona de Mediana Edad , Interleucina-6/genética , Interleucina-6/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Leucocitos Mononucleares/metabolismo , Interleucina-10/genética , Regulación de la Expresión Génica/genética , Expresión Génica/genética
2.
Mol Biol Rep ; 51(1): 707, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38824255

RESUMEN

BACKGROUND: Non-coding RNAs (ncRNAs) have a crucial impact on diverse cellular processes, influencing the progression of breast cancer (BC). The objective of this study was to identify novel ncRNAs in BC with potential effects on patient survival and disease progression. METHODS: We utilized the cancer genome atlas data to identify ncRNAs associated with BC pathogenesis. We explored the association between these ncRNA expressions and survival rates. A risk model was developed using candidate ncRNA expression and beta coefficients obtained from a multivariate Cox regression analysis. Co-expression networks were constructed to determine potential relationships between these ncRNAs and molecular pathways. For validation, we employed BC samples and the RT-qPCR method. RESULTS: Our findings revealed a noteworthy increase in the expression of AC093850.2 and CHCHD2P9 in BC, which was correlated with a poor prognosis. In contrast, ADAMTS9-AS1 and ZNF204P displayed significant downregulation and were associated with a favorable prognosis. The risk model, incorporating these four ncRNAs, robustly predicted patient survival. The co-expression network showed an effective association between levels of AC093850.2, CHCHD2P9, ADAMTS9-AS1, and ZNF204P and genes involved in pathways like metastasis, angiogenesis, metabolism, and DNA repair. The RT-qPCR results verified notable alterations in the expression of CHCHD2P9 and ZNF204P in BC samples. Pan-cancer analyses revealed alterations in the expression of these two ncRNAs across various cancer types. CONCLUSION: This study presents a groundbreaking discovery, highlighting the substantial dysregulation of CHCHD2P9 and ZNF204P in BC and other cancers, with implications for patient survival.


Asunto(s)
Neoplasias de la Mama , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/mortalidad , Femenino , Pronóstico , Regulación Neoplásica de la Expresión Génica/genética , Biomarcadores de Tumor/genética , Persona de Mediana Edad , ARN no Traducido/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Redes Reguladoras de Genes , Perfilación de la Expresión Génica/métodos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Zhonghua Bing Li Xue Za Zhi ; 53(6): 546-551, 2024 Jun 08.
Artículo en Chino | MEDLINE | ID: mdl-38825898

RESUMEN

Objective: To investigate the clinical and pathological characteristics of primary mucinous gland lesions of the fallopian tubes. Methods: The clinical data, pathomorphological characteristics and immunophenotype of 14 cases of primary mucinous gland lesions of the fallopian tube diagnosed at Obstetrics and Gynecology Hospital of Fudan University from 2015 to 2023 were analyzed retrospectively. In addition, a comprehensive review of relevant literature was conducted. Results: The age of 14 patients ranged from 53 to 83 years, with an average of 65 years. Among them, 13 cases exhibited unilateral involvement while one case showed bilateral presentation. Nine cases were mucinous metaplasia of the fallopian tube, four cases were invasive mucinous adenocarcinoma and one case was mucinous carcinoma in situ. Morphologically, mucinous metaplasia of the fallopian tube was focal, with or without inflammation. The cells of mucinous adenocarcinoma or mucinous carcinoma in situ exhibited characteristics indicative of gastrointestinal differentiation. Immunohistochemical analysis revealed diffuse positive expression of CK7, and negative expression of SATB2. CDX2 demonstrated positive staining in two cases. One case exhibited diffuse and strongly positive mutant expression of p53, whereas the remaining cases displayed wild-type expression. MUC6 showed diffuse or focally positive staining in mucinous gland lesions characterized by gastric differentiation. Some cases of mucinous adenocarcinoma of fallopian tube were subject to AB-PAS staining, resulting in red to purple cytoplasmic staining. Conclusions: Primary mucinous lesions of the fallopian tube are exceedingly uncommon. All cases of mucinous adenocarcinoma of fallopian tubes in this study exhibit the morphology and immunohistochemical characteristics of gastrointestinal differentiation. Mucinous metaplasia of the fallopian tube is a benign lesion of incidental finding, which is closely related to inflammation or gastric differentiation. Mucinous lesions of cervix, ovary and digestive tract are excluded in all patients, confirming the independent existence of mucinous lesions within fallopian tubes.


Asunto(s)
Adenocarcinoma Mucinoso , Neoplasias de las Trompas Uterinas , Trompas Uterinas , Metaplasia , Proteína p53 Supresora de Tumor , Humanos , Femenino , Neoplasias de las Trompas Uterinas/patología , Neoplasias de las Trompas Uterinas/metabolismo , Adenocarcinoma Mucinoso/patología , Adenocarcinoma Mucinoso/metabolismo , Anciano , Persona de Mediana Edad , Estudios Retrospectivos , Trompas Uterinas/patología , Anciano de 80 o más Años , Proteína p53 Supresora de Tumor/metabolismo , Metaplasia/patología , Queratina-7/metabolismo , Factor de Transcripción CDX2/metabolismo , Factor de Transcripción CDX2/genética , Mucina 6/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Carcinoma in Situ/patología , Inmunohistoquímica
6.
Artículo en Inglés | MEDLINE | ID: mdl-38830699

RESUMEN

Transcription factors play crucial roles in cancer, and oncogenic counterparts of cellular transcription factors are present in a number of tumor viruses. It was studies in the early 1980s that first showed tumor viruses could encode nuclear as well as cytoplasmic oncoproteins. Subsequent work provided detailed insight into their mechanisms of action, as well as potential therapeutic avenues. In this excerpt from his forthcoming book on the history of cancer research, Joe Lipsick looks back at early work on nuclear oncogenes, including the discovery of MYC, MYB, FOS and JUN, Rel/NF-κB, and nuclear receptors such as the retinoic acid receptor and thyroid hormone receptor.


Asunto(s)
Neoplasias , Factores de Transcripción , Humanos , Factores de Transcripción/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Historia del Siglo XX , Oncogenes , Historia del Siglo XXI , Animales
7.
Sci Rep ; 14(1): 12704, 2024 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-38830996

RESUMEN

To explore the role of YAP, a key effector of the Hippo pathway, in temporomandibular joint (TMJ) ankylosis. The temporal and spatial expression of YAP was detected via immunohistochemistry and multiplex immunohistochemistry on postoperative Days 1, 4, 7, 9, 11, 14 and 28 in a sheep model. Isolated mesenchymal stem cells (MSCs) from samples of the Day 14. The relative mRNA expression of YAP was examined before and after the osteogenic induction of MSCs. A YAP-silenced MSC model was constructed, and the effect of YAP knockdown on MSC function was examined. YAP is expressed in the nucleus of the key sites that determine the ankylosis formation, indicating that YAP is activated in a physiological state. The expression of YAP increased gradually over time. Moreover, the number of cells coexpressing of RUNX2 and YAP-with the osteogenic active zone labelled by RUNX2-tended to increase after Day 9. After the osteogenic induction of MSCs, the expression of YAP increased. After silencing YAP, the osteogenic, proliferative and migratory abilities of the MSCs were inhibited. YAP is involved in the early development of TMJ bony ankylosis. Inhibition of YAP using shRNA might be a promising way to prevent or treat TMJ ankylosis.


Asunto(s)
Anquilosis , Células Madre Mesenquimatosas , Osteogénesis , Trastornos de la Articulación Temporomandibular , Animales , Células Madre Mesenquimatosas/metabolismo , Trastornos de la Articulación Temporomandibular/metabolismo , Trastornos de la Articulación Temporomandibular/patología , Trastornos de la Articulación Temporomandibular/genética , Anquilosis/metabolismo , Anquilosis/patología , Anquilosis/genética , Proteínas Señalizadoras YAP/metabolismo , Articulación Temporomandibular/metabolismo , Articulación Temporomandibular/patología , Ovinos , Proliferación Celular , Modelos Animales de Enfermedad , Diferenciación Celular , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Movimiento Celular , Factores de Transcripción/metabolismo , Factores de Transcripción/genética
8.
BMC Biol ; 22(1): 132, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38835016

RESUMEN

BACKGROUND: ARID1A, a subunit of the SWI/SNF chromatin remodeling complex, is thought to play a significant role both in tumor suppression and tumor initiation, which is highly dependent upon context. Previous studies have suggested that ARID1A deficiency may contribute to cancer development. The specific mechanisms of whether ARID1A loss affects tumorigenesis by RNA editing remain unclear. RESULTS: Our findings indicate that the deficiency of ARID1A leads to an increase in RNA editing levels and alterations in RNA editing categories mediated by adenosine deaminases acting on RNA 1 (ADAR1). ADAR1 edits the CDK13 gene at two previously unidentified sites, namely Q113R and K117R. Given the crucial role of CDK13 as a cyclin-dependent kinase, we further observed that ADAR1 deficiency results in changes in the cell cycle. Importantly, the sensitivity of ARID1A-deficient tumor cells to SR-4835, a CDK12/CDK13 inhibitor, suggests a promising therapeutic approach for individuals with ARID1A-mutant tumors. Knockdown of ADAR1 restored the sensitivity of ARID1A deficient cells to SR-4835 treatment. CONCLUSIONS: ARID1A deficiency promotes RNA editing of CDK13 by regulating ADAR1.


Asunto(s)
Adenosina Desaminasa , Quinasas Ciclina-Dependientes , Proteínas de Unión al ADN , Edición de ARN , Proteínas de Unión al ARN , Factores de Transcripción , Adenosina Desaminasa/metabolismo , Adenosina Desaminasa/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Humanos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Quinasas Ciclina-Dependientes/metabolismo , Quinasas Ciclina-Dependientes/genética , Línea Celular Tumoral , Proteína Quinasa CDC2
9.
Folia Biol (Praha) ; 70(1): 53-61, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38830123

RESUMEN

Psoriasis is a chronic non-contagious autoimmune disease. Gallic acid is a natural compound with potential health benefits, including antioxidant, anticancer, antiviral and antibacterial properties. Nevertheless, the influence of gallic acid on psoriasis has not been fully determined. This investigation aimed to discover the effect of gallic acid on psoriasis. Thirty-one pairs of psoriatic skin tissues and healthy adult human skin tissues were collected. Human keratinocytes (HaCaT cells) were transfected with interleukin 17A (IL-17A) to create the psoriatic keratinocyte model. The content of bromodomain-containing protein 4 (BRD4) microRNA was assessed using qRT-PCR testing. The content of BRD4 was detected by Western blotting. Cell migration was evaluated by conducting a wound healing assay. Cell proliferation was determined using an EdU assay. Apoptosis was detected by the TUNEL assay. The contents of interferon gamma (IFN-γ), IL-6, IL-8 and IL-17 were detected by ELISA. BRD4 was up-regulated in psoriatic skin tissues and in the IL-17A group compared to the healthy adult human skin tissues and the control group. Silencing BRD4 inhibited cell migration, proliferation and inflammatory response but induced apoptosis in IL-17A-treated HaCaT cells. Conversely, BRD4 over-expression promoted cell migration, proliferation and inflammatory response but suppressed apoptosis in IL-17A-treated HaCaT cells. Gallic acid repressed cell migration, proliferation and inflammatory response but indu-ced apoptosis in HaCaT cells transfected with IL-17A by down-regulating BRD4. Gallic acid represses cell migration, proliferation and inflammatory response but induces apoptosis in IL-17A-transfected HaCaT cells by down-regulating BRD4.


Asunto(s)
Apoptosis , Proteínas de Ciclo Celular , Movimiento Celular , Proliferación Celular , Ácido Gálico , Inflamación , Queratinocitos , Psoriasis , Factores de Transcripción , Humanos , Psoriasis/metabolismo , Psoriasis/patología , Psoriasis/tratamiento farmacológico , Factores de Transcripción/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Ácido Gálico/farmacología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Apoptosis/efectos de los fármacos , Inflamación/patología , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Interleucina-17/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Adulto , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Masculino , Células HaCaT , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Línea Celular , Proteínas que Contienen Bromodominio
10.
Cell Stem Cell ; 31(5): 754-771.e6, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38701759

RESUMEN

Development of embryonic stem cells (ESCs) into neurons requires intricate regulation of transcription, splicing, and translation, but how these processes interconnect is not understood. We found that polypyrimidine tract binding protein 1 (PTBP1) controls splicing of DPF2, a subunit of BRG1/BRM-associated factor (BAF) chromatin remodeling complexes. Dpf2 exon 7 splicing is inhibited by PTBP1 to produce the DPF2-S isoform early in development. During neuronal differentiation, loss of PTBP1 allows exon 7 inclusion and DPF2-L expression. Different cellular phenotypes and gene expression programs were induced by these alternative DPF2 isoforms. We identified chromatin binding sites enriched for each DPF2 isoform, as well as sites bound by both. In ESC, DPF2-S preferential sites were bound by pluripotency factors. In neuronal progenitors, DPF2-S sites were bound by nuclear factor I (NFI), while DPF2-L sites were bound by CCCTC-binding factor (CTCF). DPF2-S sites exhibited enhancer modifications, while DPF2-L sites showed promoter modifications. Thus, alternative splicing redirects BAF complex targeting to impact chromatin organization during neuronal development.


Asunto(s)
Empalme Alternativo , Diferenciación Celular , Cromatina , Ribonucleoproteínas Nucleares Heterogéneas , Neuronas , Proteína de Unión al Tracto de Polipirimidina , Factores de Transcripción , Empalme Alternativo/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , Animales , Diferenciación Celular/genética , Cromatina/metabolismo , Ratones , Neuronas/metabolismo , Neuronas/citología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Transcripción Genética , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/citología , Exones/genética , Humanos , Autorrenovación de las Células/genética
11.
PLoS One ; 19(5): e0296003, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38787854

RESUMEN

Maintenance of the intestinal epithelium requires constant self-renewal and regeneration. Tight regulation of proliferation and differentiation of intestinal stem cells within the crypt region is critical to maintaining homeostasis. The transcriptional co-factors ß-catenin and YAP are required for proliferation during normal homeostasis as well as intestinal regeneration after injury: aberrant signaling activity results in over proliferation and tumorigenesis. Although both YAP and ß-catenin activity are controlled along canonical pathways, it is becoming increasingly clear that non-canonical regulation of these transcriptional regulators plays a role in fine tuning their activity. We have shown previously that MAMDC4 (Endotubin, AEGP), an integral membrane protein present in endosomes, regulates both YAP and ß-catenin activity in kidney epithelial cells and in the developing intestinal epithelium. Here we show that MAMDC4 interacts with members of the signalosome and mediates cross-talk between YAP and ß-catenin. Interestingly, this cross-talk occurs through a non-canonical pathway involving interactions between AMOT:YAP and AMOT:ß-catenin.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Endosomas , Factores de Transcripción , Vía de Señalización Wnt , beta Catenina , Humanos , beta Catenina/metabolismo , Endosomas/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Animales , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Señalizadoras YAP/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Ratones , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Células HEK293 , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Unión Proteica
12.
Cell Syst ; 15(5): 462-474.e5, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38754366

RESUMEN

Single-cell expression dynamics, from differentiation trajectories or RNA velocity, have the potential to reveal causal links between transcription factors (TFs) and their target genes in gene regulatory networks (GRNs). However, existing methods either overlook these expression dynamics or necessitate that cells be ordered along a linear pseudotemporal axis, which is incompatible with branching trajectories. We introduce Velorama, an approach to causal GRN inference that represents single-cell differentiation dynamics as a directed acyclic graph of cells, constructed from pseudotime or RNA velocity measurements. Additionally, Velorama enables the estimation of the speed at which TFs influence target genes. Applying Velorama, we uncover evidence that the speed of a TF's interactions is tied to its regulatory function. For human corticogenesis, we find that slow TFs are linked to gliomas, while fast TFs are associated with neuropsychiatric diseases. We expect Velorama to become a critical part of the RNA velocity toolkit for investigating the causal drivers of differentiation and disease.


Asunto(s)
Diferenciación Celular , Redes Reguladoras de Genes , ARN , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Redes Reguladoras de Genes/genética , Diferenciación Celular/genética , ARN/genética , ARN/metabolismo , Análisis de la Célula Individual/métodos , Regulación de la Expresión Génica/genética
13.
Nat Commun ; 15(1): 3873, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719882

RESUMEN

Human glial progenitor cells (hGPCs) exhibit diminished expansion competence with age, as well as after recurrent demyelination. Using RNA-sequencing to compare the gene expression of fetal and adult hGPCs, we identify age-related changes in transcription consistent with the repression of genes enabling mitotic expansion, concurrent with the onset of aging-associated transcriptional programs. Adult hGPCs develop a repressive transcription factor network centered on MYC, and regulated by ZNF274, MAX, IKZF3, and E2F6. Individual over-expression of these factors in iPSC-derived hGPCs lead to a loss of proliferative gene expression and an induction of mitotic senescence, replicating the transcriptional changes incurred during glial aging. miRNA profiling identifies the appearance of an adult-selective miRNA signature, imposing further constraints on the expansion competence of aged GPCs. hGPC aging is thus associated with acquisition of a MYC-repressive environment, suggesting that suppression of these repressors of glial expansion may permit the rejuvenation of aged hGPCs.


Asunto(s)
Envejecimiento , MicroARNs , Neuroglía , Factores de Transcripción , Humanos , Neuroglía/metabolismo , Neuroglía/citología , Envejecimiento/genética , Envejecimiento/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , MicroARNs/genética , MicroARNs/metabolismo , Senescencia Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre/metabolismo , Células Madre/citología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Adulto , Redes Reguladoras de Genes , Proliferación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Perfilación de la Expresión Génica
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 765-772, 2024 Apr 20.
Artículo en Chino | MEDLINE | ID: mdl-38708511

RESUMEN

OBJECTIVE: To investigate the expression level of Kruppel-like transcription factor family member KLF11 in intestinal mucosal tissues of Crohn's disease (CD) and its regulatory effect on intestinal inflammation in CD-like colitis. METHODS: We examined KLF11 expression levels in diseased and normal colon mucosal tissues from 12 CD patients and 12 patients with colorectal cancer using immunofluorescence staining. KLF11 expression was also detected in the colon mucosal tissues of a mouse model of 2, 4, 6-trinitrobenesulfonic acid (TNBS)-induced colitis. A recombinant adenoviral vector was used to upregulate KLF11 expression in the mouse models and the changes in intestinal inflammation was observed. A Caco-2 cell model with stable KLF11 overexpression was constructed by lentiviral infection. The effect of KLF11 overexpression on expressions of JAK2/STAT3 signaling pathway proteins was investigated using immunoblotting in both the mouse and cell models. The mouse models were treated with coumermycin A1, a JAK2/STAT3 signaling pathway agonist, and the changes in intestinal inflammatory responses were observed. RESULTS: The expression level of KLF11 was significantly lowered in both the clinical specimens of diseased colon mucosal tissues and the colon tissues of mice with TNBS-induced colitis (P < 0.05). Adenovirus-mediated upregulation of KLF11 significantly improved intestinal inflammation and reduced the expression levels of inflammatory factors in the intestinal mucosa of the colitis mouse models (P < 0.05). Overexpression of KLF11 significantly inhibited the expression levels of p-JAK2 and p-STAT3 in intestinal mucosal tissues of the mouse models and in Caco-2 cells (P < 0.05). Treatment with coumermycin A1 obviously inhibited the effect of KLF11 upregulation for improving colitis and significantly increased the expression levels of inflammatory factors in the intestinal mucosa of the mouse models (P < 0.05). CONCLUSION: KLF11 is downregulated in the intestinal mucosa in CD, and upregulation of KLF11 can improve intestinal inflammation and reduce the production of inflammatory factors probably by inhibiting the JAK2/STAT3 signaling pathway.


Asunto(s)
Colitis , Mucosa Intestinal , Janus Quinasa 2 , Proteínas Represoras , Transducción de Señal , Animales , Humanos , Ratones , Proteínas Reguladoras de la Apoptosis , Células CACO-2 , Colitis/inducido químicamente , Colitis/metabolismo , Enfermedad de Crohn/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Janus Quinasa 2/metabolismo , Factor de Transcripción STAT3/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Ácido Trinitrobencenosulfónico , Regulación hacia Arriba
15.
Kidney Int ; 105(6): 1157-1159, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38777398

RESUMEN

Chen et al. identify dysregulation of the transcriptional activator Yes-associated protein in the podocytes of diabetic mouse and human kidneys. Podocyte Yes-associated protein deficiency led to downregulation of the key transcription factor Wilms' tumor 1, and worsened podocyte injury in a mouse model of diabetic kidney injury. Yes-associated protein may therefore play a critical role in diabetic podocyte injury via regulation of Wilms' tumor 1 expression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Nefropatías Diabéticas , Podocitos , Factores de Transcripción , Proteínas WT1 , Proteínas Señalizadoras YAP , Podocitos/metabolismo , Podocitos/patología , Animales , Humanos , Proteínas Señalizadoras YAP/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas WT1/metabolismo , Proteínas WT1/genética , Ratones , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/etiología , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética
16.
Sci Rep ; 14(1): 11688, 2024 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-38778150

RESUMEN

Prostate cancer lineage plasticity is a key driver in the transition to neuroendocrine prostate cancer (NEPC), and the RTK/RAS signaling pathway is a well-established cancer pathway. Nevertheless, the comprehensive link between the RTK/RAS signaling pathway and lineage plasticity has received limited investigation. In particular, the intricate regulatory network governing the interplay between RTK/RAS and lineage plasticity remains largely unexplored. The multi-omics data were clustered with the coefficient of argument and neighbor joining algorithm. Subsequently, the clustered results were analyzed utilizing the GSEA, gene sets related to stemness, multi-lineage state datasets, and canonical cancer pathway gene sets. Finally, a comprehensive exploration of the data based on the ssGSEA, WGCNA, GSEA, VIPER, prostate cancer scRNA-seq data, and the GPSAdb database was conducted. Among the six modules in the clustering results, there are 300 overlapping genes, including 3 previously unreported prostate cancer genes that were validated to be upregulated in prostate cancer through RT-qPCR. Function Module 6 shows a positive correlation with prostate cancer cell stemness, multi-lineage states, and the RTK/RAS signaling pathway. Additionally, the 19 leading-edge genes of the RTK/RAS signaling pathway promote prostate cancer lineage plasticity through a complex network of transcriptional regulation and copy number variations. In the transcriptional regulation network, TP63 and FOXO1 act as suppressors of prostate cancer lineage plasticity, whereas RORC exerts a promoting effect. This study provides a comprehensive perspective on the role of the RTK/RAS pathway in prostate cancer lineage plasticity and offers new clues for the treatment of NEPC.


Asunto(s)
Minería de Datos , Neoplasias de la Próstata , Transducción de Señal , Factores de Transcripción , Masculino , Humanos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo , Variaciones en el Número de Copia de ADN , Regulación Neoplásica de la Expresión Génica , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Redes Reguladoras de Genes , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Linaje de la Célula/genética
17.
Int J Biol Macromol ; 270(Pt 2): 132519, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38768919

RESUMEN

The Lrp/AsnC family of transcriptional regulators is commonly found in prokaryotes and is associated with the regulation of amino acid metabolism. However, it remains unclear how the L-cysteine-responsive Lrp/AsnC family regulator perceives and responds to L-cysteine. Here, we try to elucidate the molecular mechanism of the L-cysteine-responsive transcriptional regulator. Through 5'RACE and EMSA, we discovered a 15 bp incompletely complementary pair palindromic sequence essential for DecR binding, which differed slightly from the binding sequence of other Lrp/AsnC transcription regulators. Using alanine scanning, we identified the L-cysteine binding site on DecR and found that different Lrp/AsnC regulators adjust their binding pocket's side-chain residues to accommodate their specific effector. MD simulations were then conducted to explore how ligand binding influences the allosteric behavior of the protein. PCA and in silico docking revealed that ligand binding induced perturbations in the linker region, triggering conformational alterations and leading to the relocalization of the DNA-binding domains, enabling the embedding of the DNA-binding region of DecR into the DNA molecule, thereby enhancing DNA-binding affinity. Our findings can broaden the understanding of the recognition and regulatory mechanisms of the Lrp/AsnC-type transcription factors, providing a theoretical basis for further investigating the molecular mechanisms of other transcription factors.


Asunto(s)
Proteínas Bacterianas , Cisteína , Unión Proteica , Cisteína/química , Cisteína/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Sitios de Unión , Factores de Transcripción/metabolismo , Factores de Transcripción/química , Factores de Transcripción/genética , Simulación de Dinámica Molecular , Simulación del Acoplamiento Molecular , Proteína Reguladora de Respuesta a la Leucina/metabolismo , Proteína Reguladora de Respuesta a la Leucina/química , Proteína Reguladora de Respuesta a la Leucina/genética
18.
Biomed Pharmacother ; 175: 116690, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38718519

RESUMEN

Acute pancreatitis (AP) is one of the most common gastrointestinal tract diseases with significant morbidity and mortality. Current treatments remain unspecific and supportive due to the severity and clinical course of AP, which can fluctuate rapidly and unpredictably. Mitochondria, cellular power plant to produce energy, are involved in a variety of physiological or pathological activities in human body. There is a growing evidence indicating that mitochondria damage-associated molecular patterns (mtDAMPs) play an important role in pathogenesis and progression of AP. With the pro-inflammatory properties, released mtDAMPs may damage pancreatic cells by binding with receptors, activating downstream molecules and releasing inflammatory factors. This review focuses on the possible interaction between AP and mtDAMPs, which include cytochrome c (Cyt c), mitochondrial transcription factor A (TFAM), mitochondrial DNA (mtDNA), cardiolipin (CL), adenosine triphosphate (ATP) and succinate, with focus on experimental research and potential therapeutic targets in clinical practice. Preventing or diminishing the release of mtDAMPs or targeting the mtDAMPs receptors might have a role in AP progression.


Asunto(s)
Mitocondrias , Pancreatitis , Humanos , Pancreatitis/metabolismo , Pancreatitis/patología , Pancreatitis/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Animales , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Enfermedad Aguda , Alarminas/metabolismo , Adenosina Trifosfato/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética
19.
FASEB J ; 38(9): e23633, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38690712

RESUMEN

Recent reports suggest that the Hippo signaling pathway regulates testis development, though its exact roles in Sertoli cell differentiation remain unknown. Here, we examined the functions of the main Hippo pathway kinases, large tumor suppressor homolog kinases 1 and 2 (Lats1 and Lats2) in developing mouse Sertoli cells. Conditional inactivation of Lats1/2 in Sertoli cells resulted in the disorganization and overgrowth of the testis cords, the induction of a testicular inflammatory response and germ cell apoptosis. Stimulated by retinoic acid 8 (STRA8) expression in germ cells additionally suggested that germ cells may have been preparing to enter meiosis prior to their loss. Gene expression analyses of the developing testes of conditional knockout animals further suggested impaired Sertoli cell differentiation, epithelial-to-mesenchymal transition, and the induction of a specific set of genes associated with Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ)-mediated integrin signaling. Finally, the involvement of YAP/TAZ in Sertoli cell differentiation was confirmed by concomitantly inactivating Yap/Taz in Lats1/2 conditional knockout model, which resulted in a partial rescue of the testicular phenotypic changes. Taken together, these results identify Hippo signaling as a crucial pathway for Sertoli cell development and provide novel insight into Sertoli cell fate maintenance.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Diferenciación Celular , Proteínas Serina-Treonina Quinasas , Células de Sertoli , Proteínas Supresoras de Tumor , Proteínas Señalizadoras YAP , Animales , Células de Sertoli/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Masculino , Ratones , Proteínas Señalizadoras YAP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Diferenciación Celular/fisiología , Ratones Noqueados , Transducción de Señal , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Testículo/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Aciltransferasas/genética , Aciltransferasas/metabolismo , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/metabolismo , Transactivadores/metabolismo , Transactivadores/genética
20.
Nat Commun ; 15(1): 3771, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704377

RESUMEN

Ovarian metastasis is one of the major causes of treatment failure in patients with gastric cancer (GC). However, the genomic characteristics of ovarian metastasis in GC remain poorly understood. In this study, we enroll 74 GC patients with ovarian metastasis, with 64 having matched primary and metastatic samples. Here, we show a characterization of the mutation landscape of this disease, alongside an investigation into the molecular heterogeneity and pathway mutation enrichments between synchronous and metachronous metastasis. We classify patients into distinct clonal evolution patterns based on the distribution of mutations in paired samples. Notably, the parallel evolution group exhibits the most favorable prognosis. Additionally, by analyzing the differential response to chemotherapy, we identify potential biomarkers, including SALL4, CCDC105, and CLDN18, for predicting the efficacy of paclitaxel treatment. Furthermore, we validate that CLDN18 fusion mutations improve tumor response to paclitaxel treatment in GC with ovarian metastasis in vitro and vivo.


Asunto(s)
Biomarcadores de Tumor , Mutación , Neoplasias Ováricas , Paclitaxel , Neoplasias Gástricas , Paclitaxel/uso terapéutico , Humanos , Neoplasias Gástricas/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología , Femenino , Neoplasias Ováricas/genética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Biomarcadores de Tumor/genética , Claudinas/genética , Claudinas/metabolismo , Evolución Molecular , Animales , Persona de Mediana Edad , Pronóstico , Línea Celular Tumoral , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Anciano , Antineoplásicos Fitogénicos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA