Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.954
Filtrar
1.
Elife ; 122024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39137024

RESUMEN

Hepatic factors secreted by the liver promote homeostasis and are pivotal for maintaining the liver-gut axis. Bile acid metabolism is one such example wherein, bile acid synthesis occurs in the liver and its biotransformation happens in the intestine. Dysfunctional interactions between the liver and the intestine stimulate varied pathological outcomes through its bidirectional portal communication. Indeed, aberrant bile acid metabolism has been reported in inflammatory bowel disease (IBD). However, the molecular mechanisms underlying these crosstalks that perpetuate intestinal permeability and inflammation remain obscure. Here, we identify a novel hepatic gene program regulated by Rela and Stat3 that accentuates the inflammation in an acute experimental colitis model. Hepatocyte-specific ablation of Rela and Stat3 reduces the levels of primary bile acids in both the liver and the gut and shows a restricted colitogenic phenotype. On supplementation of chenodeoxycholic acid (CDCA), knock-out mice exhibit enhanced colitis-induced alterations. This study provides persuasive evidence for the development of multi-organ strategies for treating IBD and identifies a hepatocyte-specific Rela-Stat3 network as a promising therapeutic target.


Asunto(s)
Ácidos y Sales Biliares , Colitis , Modelos Animales de Enfermedad , Hepatocitos , Ratones Noqueados , Factor de Transcripción STAT3 , Factor de Transcripción ReIA , Animales , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/genética , Colitis/patología , Hepatocitos/metabolismo , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , Ratones , Ácidos y Sales Biliares/metabolismo , Regulación de la Expresión Génica , Hígado/metabolismo , Hígado/patología , Ratones Endogámicos C57BL
2.
Hum Exp Toxicol ; 43: 9603271241279166, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39190898

RESUMEN

Bromodomain and WD-repeat domain-containing protein 3 (BRWD3) exhibits high expression in lung adenocarcinoma (LUAD) tissues and cells; however, its function in arsenic-induced toxicological responses remains unclear. This study aimed to investigate BRWD3 expression in response to arsenic-induced conditions and its impact on the proliferation and apoptosis of LUAD cell line SPC-A1 upon BRWD3 knockdown. The results revealed a decrease in BRWD3 expression in SPC-A1 cells treated with sodium arsenite (NaAsO2), but not sodium arsenite's metabolites. BRWD3 knockdown suppressed cell proliferation and induced apoptosis in SPC-A1 cells. Western blot analysis revealed that BRWD3 knockdown resulted in the upregulation of p53, phospho-p53-Ser392, and its downstream factors including MDM2, Bak, and Bax. Additionally, we observed the downregulation of p65, phospho-p65-Ser276, phospho-p65-Ser536, and its downstream factors, including IκBα, BIRC3, XIAP and CIAP1. Moreover, polymerase chain reaction analysis showed that BRWD3 knockdown also resulted in the downregulation of proliferation-related genes and upregulation of apoptosis-related genes. In conclusion, BRWD3 mediated proliferation and apoptosis via the p53 and p65 pathways in response to arsenic exposure, suggesting potential implications for LUAD treatment through BRWD3 downregulation by arsenic.


Asunto(s)
Adenocarcinoma del Pulmón , Apoptosis , Proliferación Celular , Regulación hacia Abajo , Neoplasias Pulmonares , Proteína p53 Supresora de Tumor , Humanos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Línea Celular Tumoral , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/patología , Regulación hacia Abajo/efectos de los fármacos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/tratamiento farmacológico , Arsénico/toxicidad , Compuestos de Sodio/toxicidad , Arsenitos/toxicidad , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética
3.
Int J Mol Sci ; 25(15)2024 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-39125811

RESUMEN

Advanced glycated end products (AGEs) are cytotoxic compounds that are mainly increased in diabetes mellitus (DM), kidney failure, inflammation, and in response to the ingestion of AGE-rich diets. AGEs can also impair glycemic homeostasis by decreasing the expression of the Slc2a4 (solute carrier family 2 member 4) gene and its GLUT4 (solute carrier family 2, facilitated glucose transporter member 4) protein in muscle. However, the mechanisms underlying AGE's effect on adipocytes have not been demonstrated yet. This study investigated the effects of AGEs upon Slc2a4/GLUT4 expression in 3T3-L1 adipocytes, as well as the potential role of NFKB (nuclear factor NF-kappa-B) activity in the effects observed. Adipocytes were cultured in the presence of control albumin (CA) or advanced glycated albumin (GA) at concentrations of 0.4, 3.6, and 5.4 mg/mL for 24 h or 72 h. Slc2a4, Rela, and Nfkb1mRNAs were measured by RT-qPCR, GLUT4, IKKA/B, and p50/p65 NFKB subunits using Western blotting, and p50/p65 binding into the Slc2a4 promoter was analyzed by chromatin immunoprecipitation (ChIP) assay. GA at 0.4 mg/mL increased Slc2a4/GLUT4 expression after 24 h and 72 h (from 50% to 100%), but at 5.4 mg/mL, Slc2a4/GLUT4 expression decreased at 72 h (by 50%). Rela and Nfkb1 expression increased after 24 h at all concentrations, but this effect was not observed at 72 h. Furthermore, 5.4 mg/mL of GA increased the p50/p65 nuclear content and binding into Slc2a4 at 72 h. In summary, this study reveals AGE-induced and NFKB-mediated repression of Slc2a4/GLUT4 expression. This can compromise the adipocyte glucose utilization, contributing not only to the worsening of glycemic control in DM subjects but also the impairment of glycemic homeostasis in non-DM subjects under the high intake of AGE-rich foods.


Asunto(s)
Células 3T3-L1 , Adipocitos , Transportador de Glucosa de Tipo 4 , Productos Finales de Glicación Avanzada , Factor de Transcripción ReIA , Animales , Transportador de Glucosa de Tipo 4/metabolismo , Transportador de Glucosa de Tipo 4/genética , Ratones , Productos Finales de Glicación Avanzada/metabolismo , Productos Finales de Glicación Avanzada/farmacología , Adipocitos/metabolismo , Adipocitos/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , FN-kappa B/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regiones Promotoras Genéticas , Subunidad p50 de NF-kappa B/metabolismo , Subunidad p50 de NF-kappa B/genética
4.
Mol Carcinog ; 63(9): 1827-1841, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39016636

RESUMEN

LAIR1, a receptor found on immune cells, is capable of binding to collagen and is involved in immune-related diseases. However, the precise contribution of LAIR1 expressed on hepatocellular carcinoma (HCC) cells to tumor microenvironment is still unclear. In our study, bioinformatics analysis and immunofluorescence were employed to study the correlation between LAIR1 levels and clinical indicators. Transwell and scratch tests were used to evaluate how LAIR1 affected the migration and invasion of HCC cells. The chemotactic capacity and alternative activation of macrophages were investigated using RT-qPCR, transwell, and immunofluorescence. To investigate the molecular mechanisms, transcriptome sequencing analysis, Western blot, nucleus/cytoplasm fractionation, ELISA, and cytokine microarray were employed. We revealed a significant correlation between the presence of LAIR1 and an unfavorable outcome in HCC. We indicated that LAIR1 promoted migration and invasion of HCC cells through the AKT-IKKß-p65 axis. Additionally, the alternative activation and infiltration of tumor-associated macrophages induced by LAIR1 were reliant on the upregulation of IL6 and CCL5 within this axis, respectively. In conclusion, blocking LAIR1 was found to be an effective approach in combating the cancerous advancement of HCC.


Asunto(s)
Carcinoma Hepatocelular , Movimiento Celular , Neoplasias Hepáticas , Proteínas Proto-Oncogénicas c-akt , Receptores Inmunológicos , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/genética , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Quinasa I-kappa B/metabolismo , Quinasa I-kappa B/genética , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , Línea Celular Tumoral , Microambiente Tumoral , Regulación Neoplásica de la Expresión Génica , Transducción de Señal , Macrófagos/metabolismo , Macrófagos/patología , Proliferación Celular , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/patología , Macrófagos Asociados a Tumores/inmunología , Invasividad Neoplásica
5.
J Clin Invest ; 134(14)2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-39007267

RESUMEN

Emerging evidence has linked the dysregulation of N6-methyladenosine (m6A) modification to inflammation and inflammatory diseases, but the underlying mechanism still needs investigation. Here, we found that high levels of m6A modification in a variety of hyperinflammatory states are p65-dependent because Wilms tumor 1-associated protein (WTAP), a key component of the "writer" complex, is transcriptionally regulated by p65, and its overexpression can lead to increased levels of m6A modification. Mechanistically, upregulated WTAP is more prone to phase separation to facilitate the aggregation of the writer complex to nuclear speckles and the deposition of m6A marks on transcriptionally active inflammatory transcripts, thereby accelerating the proinflammatory response. Further, a myeloid deficiency in WTAP attenuates the severity of LPS-induced sepsis and DSS-induced IBD. Thus, the proinflammatory effect of WTAP is a general risk-increasing mechanism, and interrupting the assembly of the m6A writer complex to reduce the global m6A levels by targeting the phase separation of WTAP may be a potential and promising therapeutic strategy for alleviating hyperinflammation.


Asunto(s)
Adenosina , Proteínas de Ciclo Celular , Inflamación , Factores de Empalme de ARN , Animales , Humanos , Ratones , Adenosina/metabolismo , Adenosina/análogos & derivados , Modelos Animales de Enfermedad , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos , Ratones Noqueados , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Sepsis/metabolismo , Sepsis/genética , Sepsis/patología , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética
6.
Genes Dev ; 38(11-12): 528-535, 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-38960718

RESUMEN

As part of the efforts to understand nuclear IκB function in NF-κB-dependent gene expression, we report an X-ray crystal structure of the IκBζ ankyrin repeat domain in complex with the dimerization domain of the NF-κB p50 homodimer. IκBζ possesses an N-terminal α helix that conveys domain folding stability. Affinity and specificity of the complex depend on a small portion of p50 at the nuclear localization signal. The model suggests that only one p50 subunit supports binding with IκBζ, and biochemical experiments confirm that IκBζ associates with DNA-bound NF-κB p50:RelA heterodimers. Comparisons of IκBζ:p50 and p50:κB DNA complex crystallographic models indicate that structural rearrangement is necessary for ternary complex formation of IκBζ and p50 with DNA.


Asunto(s)
Modelos Moleculares , Subunidad p50 de NF-kappa B , Unión Proteica , Multimerización de Proteína , Humanos , Secuencia de Aminoácidos , Núcleo Celular/metabolismo , Cristalografía por Rayos X , ADN/metabolismo , ADN/química , Proteínas I-kappa B/metabolismo , Proteínas I-kappa B/química , Proteínas I-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , Subunidad p50 de NF-kappa B/química , Subunidad p50 de NF-kappa B/genética , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/química , Factor de Transcripción ReIA/genética
7.
Sci Signal ; 17(843): eadk0231, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38954637

RESUMEN

The Hippo pathway is generally understood to inhibit tumor growth by phosphorylating the transcriptional cofactor YAP to sequester it to the cytoplasm and reduce the formation of YAP-TEAD transcriptional complexes. Aberrant activation of YAP occurs in various cancers. However, we found a tumor-suppressive function of YAP in clear cell renal cell carcinoma (ccRCC). Using cell cultures, xenografts, and patient-derived explant models, we found that the inhibition of upstream Hippo-pathway kinases MST1 and MST2 or expression of a constitutively active YAP mutant impeded ccRCC proliferation and decreased gene expression mediated by the transcription factor NF-κB. Mechanistically, the NF-κB subunit p65 bound to the transcriptional cofactor TEAD to facilitate NF-κB-target gene expression that promoted cell proliferation. However, by competing for TEAD, YAP disrupted its interaction with NF-κB and prompted the dissociation of p65 from target gene promoters, thereby inhibiting NF-κB transcriptional programs. This cross-talk between the Hippo and NF-κB pathways in ccRCC suggests that targeting the Hippo-YAP axis in an atypical manner-that is, by activating YAP-may be a strategy for slowing tumor growth in patients.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Carcinoma de Células Renales , Proliferación Celular , Neoplasias Renales , Proteínas Serina-Treonina Quinasas , Factores de Transcripción , Proteínas Señalizadoras YAP , Humanos , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Neoplasias Renales/metabolismo , Neoplasias Renales/genética , Neoplasias Renales/patología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas Señalizadoras YAP/metabolismo , Proteínas Señalizadoras YAP/genética , Animales , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , Ratones , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Vía de Señalización Hippo , Transducción de Señal , Factores de Transcripción de Dominio TEA/metabolismo , FN-kappa B/metabolismo , FN-kappa B/genética , Ratones Desnudos , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Serina-Treonina Quinasa 3
8.
J Agric Food Chem ; 72(31): 17444-17454, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39074384

RESUMEN

Acute kidney injury (AKI), if not well controlled, may progress to chronic kidney disease (CKD). Diosgenin is a natural phytosteroid sapogenin from plants. This study aimed to investigate the mechanistic effects of diosgenin on AKI and AKI related development of CKD. The mouse model of ischemia/reperfusion (I/R)-induced AKI was used, and its progressive changes were followed. Human renal proximal tubular epithelial cells were used, and hypoxia stimulation was applied to mimic the in vivo I/R. Diosgenin, given after renal injury, preserved kidney function, as evidenced by a reduction in serum levels of BUN, creatinine, and UACR in both acute and chronic phases of AKI. Diosgenin alleviated I/R-induced tubular injury and prevented macrophage infiltration and renal fibrosis in AKI mice. Furthermore, diosgenin also mitigated the development of CKD from AKI with reduced renal expression of inflammatory, fibrotic, and epithelial-mesenchymal transition markers. In human renal tubular epithelial cells, diosgenin downregulated the hypoxia-induced oxidative stress and cellular damages that were dependent on the NOX4/p65 signaling pathways. Taken together, diosgenin treatment reduced I/R-induced AKI and ameliorated the progression to CKD from AKI probably by modifying the NOX4/p65 signaling pathways.


Asunto(s)
Lesión Renal Aguda , Diosgenina , Ratones Endogámicos C57BL , NADPH Oxidasa 4 , Insuficiencia Renal Crónica , Transducción de Señal , Diosgenina/farmacología , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/metabolismo , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/fisiopatología , Animales , Humanos , Ratones , Transducción de Señal/efectos de los fármacos , NADPH Oxidasa 4/metabolismo , NADPH Oxidasa 4/genética , Masculino , Estrés Oxidativo/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , Progresión de la Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Línea Celular
9.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167320, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38936515

RESUMEN

Postmenopausal women experience bone loss and weight gain. To date, crosstalk between estrogen receptor signals and nuclear factor-κB (NF-κB) has been reported, and estrogen depletion enhances bone resorption by osteoclasts via NF-κB activation. However, it is unclear when and in which tissues NF-κB is activated after menopause, and how NF-κB acts as a common signaling molecule for postmenopausal weight gain and bone loss. Therefore, we examined the role of NF-κB in bone and energy metabolism following menopause. NF-κB reporter mice, which can be used to measure NF-κB activation in vivo, were ovariectomized (OVX) and the luminescence intensity after OVX increased in the metaphyses of the long bones and perigonadal white adipose tissue, but not in the other tissues. OVX was performed on wild-type (WT) and p65 mutant knock-in (S534A) mice, whose mutation enhances the transcriptional activity of NF-κB. Weight gain with worsening glucose tolerance was significant in S534A mice after OVX compared with those of WT mice. The bone density of the sham group in WT or S534A mice did not change, whereas in the S534A-OVX group it significantly decreased due to the suppression of bone formation and increase in bone marrow adipocytes. Disulfiram, an anti-alcoholic drug, suppressed OVX-induced activation of NF-κB in the metaphyses of long bones and white adipose tissue (WAT), as well as weight gain and bone loss. Overall, the activation of NF-κB in the metaphyses of long bones and WAT after OVX regulates post-OVX weight gain and bone loss.


Asunto(s)
Resorción Ósea , FN-kappa B , Ovariectomía , Transducción de Señal , Aumento de Peso , Animales , Ovariectomía/efectos adversos , Femenino , Ratones , FN-kappa B/metabolismo , Resorción Ósea/metabolismo , Resorción Ósea/patología , Humanos , Densidad Ósea , Ratones Endogámicos C57BL , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética
10.
Mol Ther ; 32(8): 2549-2562, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38879753

RESUMEN

Osteoarthritis (OA) is a painful and debilitating disease affecting over 500 million people worldwide. Intraarticular injection of mesenchymal stromal cells (MSCs) shows promise for the clinical treatment of OA, but the lack of consistency in MSC preparation and application makes it difficult to further optimize MSC therapy and to properly evaluate the clinical outcomes. In this study, we used Sox9 activation and RelA inhibition, both mediated by the CRISPR-dCas9 technology simultaneously, to engineer MSCs with enhanced chondrogenic potential and downregulated inflammatory responses. We found that both Sox9 and RelA could be fine-tuned to the desired levels, which enhances the chondrogenic and immunomodulatory potentials of the cells. Intraarticular injection of modified cells significantly attenuated cartilage degradation and palliated OA pain compared with the injection of cell culture medium or unmodified cells. Mechanistically, the modified cells promoted the expression of factors beneficial to cartilage integrity, inhibited the production of catabolic enzymes in osteoarthritic joints, and suppressed immune cells. Interestingly, a substantial number of modified cells could survive in the cartilaginous tissues including articular cartilage and meniscus. Together, our results suggest that CRISPR-dCas9-based gene regulation is useful for optimizing MSC therapy for OA.


Asunto(s)
Sistemas CRISPR-Cas , Células Madre Mesenquimatosas , Osteoartritis , Factor de Transcripción SOX9 , Factor de Transcripción ReIA , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Osteoartritis/terapia , Osteoartritis/genética , Osteoartritis/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , Ratones , Humanos , Modelos Animales de Enfermedad , Cartílago Articular/metabolismo , Cartílago Articular/patología , Trasplante de Células Madre Mesenquimatosas/métodos , Condrogénesis/genética , Edición Génica , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Condrocitos/metabolismo
11.
Genes Dev ; 38(11-12): 536-553, 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-38918046

RESUMEN

The five NF-κB family members and three nuclear IκB proteins play important biological roles, but the mechanisms by which distinct members of these protein families contribute to selective gene transcription remain poorly understood, especially at a genome-wide scale. Using nascent transcript RNA-seq, we observed considerable overlap between p50-dependent and IκBζ-dependent genes in Toll-like receptor 4 (TLR4)-activated macrophages. Key immunoregulatory genes, including Il6, Il1b, Nos2, Lcn2, and Batf, are among the p50-IκBζ-codependent genes. IκBζ-bound genomic sites are occupied at earlier time points by NF-κB dimers. However, p50-IκBζ codependence does not coincide with preferential binding of either p50 or IκBζ, as RelA co-occupies hundreds of genomic sites with the two proteins. A common feature of p50-IκBζ-codependent genes is a nearby p50/RelA/IκBζ-cobound site exhibiting p50-dependent binding of both RelA and IκBζ. This and other results suggest that IκBζ acts in concert with RelA:p50 heterodimers. Notably, p50-IκBζ-codependent genes comprise a high percentage of genes exhibiting the greatest differential expression between TLR4-stimulated and tumor necrosis factor receptor (TNFR)-stimulated macrophages. Thus, our genome-centric analysis reveals a defined p50-IκBζ pathway that selectively activates a set of key immunoregulatory genes and serves as an important contributor to differential TNFR and TLR4 responses.


Asunto(s)
Regulación de la Expresión Génica , Macrófagos , Subunidad p50 de NF-kappa B , Transducción de Señal , Receptor Toll-Like 4 , Animales , Ratones , Proteínas Adaptadoras Transductoras de Señales , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Inflamación/genética , Inflamación/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , Unión Proteica , Transducción de Señal/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , Masculino
12.
Cell Mol Life Sci ; 81(1): 255, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38856747

RESUMEN

Glioblastoma multiforme (GBM) is the most common and malignant primary brain tumor; GBM's inevitable recurrence suggests that glioblastoma stem cells (GSC) allow these tumors to persist. Our previous work showed that FOSL1, transactivated by the STAT3 gene, functions as a tumorigenic gene in glioma pathogenesis and acts as a diagnostic marker and potential drug target in glioma patients. Accumulating evidence shows that STAT3 and NF-κB cooperate to promote the development and progression of various cancers. The link between STAT3 and NF-κB suggests that NF-κB can also transcriptionally regulate FOSL1 and contribute to gliomagenesis. To investigate downstream molecules of FOSL1, we analyzed the transcriptome after overexpressing FOSL1 in a PDX-L14 line characterized by deficient FOSL1 expression. We then conducted immunohistochemical staining for FOSL1 and NF-κB p65 using rabbit polyclonal anti-FOSL1 and NF-κB p65 in glioma tissue microarrays (TMA) derived from 141 glioma patients and 15 healthy individuals. Next, mutants of the human FOSL1 promoter, featuring mutations in essential binding sites for NF-κB were generated using a Q5 site-directed mutagenesis kit. Subsequently, we examined luciferase activity in glioma cells and compared it to the wild-type FOSL1 promoter. Then, we explored the mutual regulation between NF-κB signaling and FOSL1 by modulating the expression of NF-κB or FOSL1. Subsequently, we assessed the activity of FOSL1 and NF-κB. To understand the role of FOSL1 in cell growth and stemness, we conducted a CCK-8 assay and cell cycle analysis, assessing apoptosis and GSC markers, ALDH1, and CD133 under varying FOSL1 expression conditions. Transcriptome analyses of downstream molecules of FOSL1 show that NF-κB signaling pathway is regulated by FOSL1. NF-κB p65 protein expression correlates to the expression of FOSL1 in glioma patients, and both are associated with glioma grades. NF-κB is a crucial transcription factor activating the FOSL1 promoter in glioma cells. Mutual regulation between NF-κB and FOSL1 contributes to glioma tumorigenesis and stemness through promoting G1/S transition and inhibiting apoptosis. Therefore, the FOSL1 molecular pathway is functionally connected to NF-κB activation, enhances stemness, and is indicative that FOSL1 may potentially be a novel GBM drug target.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , FN-kappa B , Células Madre Neoplásicas , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fos , Animales , Humanos , Ratones , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Glioblastoma/patología , Glioblastoma/genética , Glioblastoma/metabolismo , Glioma/patología , Glioma/genética , Glioma/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Transducción de Señal , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética
13.
PLoS One ; 19(6): e0300790, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38935597

RESUMEN

Myocardial ischemia-reperfusion injury (MIRI) refers to the secondary damage to myocardial tissue that occurs when blood perfusion is rapidly restored following myocardial ischemia. This process often exacerbates the injury to myocardial fiber structure and function. The activation mechanism of angiogenesis is closely related to MIRI and plays a significant role in the occurrence and progression of ischemic injury. In this study, we utilized sequencing data from the GEO database and employed WGCNA, Mfuzz cluster analysis, and protein interaction network to identify Stat3, Rela, and Ubb as hub genes involved in MIRI-angiogenesis. Additionally, the GO and KEGG analysis of differentially expressed genes highlighted their broad participation in inflammatory responses and associated signaling pathways. Moreover, the analysis of sequencing data and hub genes revealed a notable increase in the infiltration ratio of monocytes and activated mast cells. By establishing key cell ROC curves, using independent datasets, and validating the expression of hub genes, we demonstrated their high diagnostic value. Moreover, by scrutinizing single-cell sequencing data alongside trajectory analysis, it has come to light that Stat3 and Rela exhibit predominant expression within Dendritic cells. In contrast, Ubb demonstrates expression across multiple cell types, with all three genes being expressed at distinct stages of cellular development. Lastly, leveraging the CMap database, we predicted potential small molecule compounds for the identified hub genes and validated their binding activity through molecular docking. Ultimately, our research provides valuable evidence and references for the early diagnosis and treatment of MIRI from the perspective of angiogenesis.


Asunto(s)
Biomarcadores , Daño por Reperfusión Miocárdica , Factor de Transcripción STAT3 , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Humanos , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Biomarcadores/metabolismo , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , Mapas de Interacción de Proteínas/genética , Neovascularización Patológica/genética , Perfilación de la Expresión Génica , Angiogénesis
14.
Biochem J ; 481(14): 959-980, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-38941070

RESUMEN

While IκB-kinase-ε (IKKε) induces immunomodulatory genes following viral stimuli, its up-regulation by inflammatory cytokines remains under-explored. Since airway epithelial cells respond to airborne insults and potentiate inflammation, IKKε expression was characterized in pulmonary epithelial cell lines (A549, BEAS-2B) and primary human bronchial epithelial cells grown as submersion or differentiated air-liquid interface cultures. IKKε expression was up-regulated by the pro-inflammatory cytokines, interleukin-1ß (IL-1ß) and tumour necrosis factor-α (TNFα). Thus, mechanistic interrogations in A549 cells were used to demonstrate the NF-κB dependence of cytokine-induced IKKε. Furthermore, chromatin immunoprecipitation in A549 and BEAS-2B cells revealed robust recruitment of the NF-κB subunit, p65, to one 5' and two intronic regions within the IKKε locus (IKBKE). In addition, IL-1ß and TNFα induced strong RNA polymerase 2 recruitment to the 5' region, the first intron, and the transcription start site. Stable transfection of the p65-binding regions into A549 cells revealed IL-1ß- and TNFα-inducible reporter activity that required NF-κB, but was not repressed by glucocorticoid. While critical NF-κB motifs were identified in the 5' and downstream intronic regions, the first intronic region did not contain functional NF-κB motifs. Thus, IL-1ß- and TNFα-induced IKKε expression involves three NF-κB-binding regions, containing multiple functional NF-κB motifs, and potentially other mechanisms of p65 binding through non-classical NF-κB binding motifs. By enhancing IKKε expression, IL-1ß may prime, or potentiate, responses to alternative stimuli, as modelled by IKKε phosphorylation induced by phorbol 12-myristate 13-acetate. However, since IKKε expression was only partially repressed by glucocorticoid, IKKε-dependent responses could contribute to glucocorticoid-resistant disease.


Asunto(s)
Células Epiteliales , Quinasa I-kappa B , Humanos , Quinasa I-kappa B/metabolismo , Quinasa I-kappa B/genética , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Células A549 , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , Interleucina-1beta/farmacología , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , FN-kappa B/metabolismo , FN-kappa B/genética , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/genética , Pulmón/metabolismo , Pulmón/citología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/citología , Regulación de la Expresión Génica/efectos de los fármacos
15.
Medicine (Baltimore) ; 103(26): e38737, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38941371

RESUMEN

Alterations in signaling pathways and modulation of cell metabolism are associated with the pathogenesis of cancers, including hepatocellular carcinoma (HCC). Small ubiquitin-like modifier (SUMO) proteins and NF-κB family play major roles in various cellular processes. The current study aims to determine the expression profile of SUMO and NF-κB genes in HCC tumors and investigate their association with the clinical outcome of HCC. The expression of 5 genes - SUMO1, SUMO2, SUMO3, NF-κB p65, and NF-κB p50 - was quantified in tumor and adjacent non-tumor tissues of 58 HBV-related HCC patients by real-time quantitative PCR and was analyzed for the possible association with clinical parameters of HCC. The expression of SUMO2 was significantly higher in HCC tumor tissues compared to the adjacent non-tumor tissues (P = .01), while no significant difference in SUMO1, SUMO3, NF-κB p65, and NF-κB p50 expression was observed between HCC tumor and non-tumor tissues (P > .05). In HCC tissues, a strong correlation was observed between the expression of SUMO2 and NF-κB p50, between SUMO3 and NF-κB p50, between SUMO3 and NF-κB p65 (Spearman rho = 0.83; 0.82; 0.772 respectively; P < .001). The expression of SUMO1, SUMO2, SUMO3, NF-κB p65, and NF-κB p50 was decreased in grade 3 compared to grades 1 and 2 in HCC tumors according to the World Health Organization grades system. Our results highlighted that the SUMO2 gene is upregulated in tumor tissues of patients with HCC, and is related to the development of HCC, thus it may be associated with the pathogenesis of HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina , Humanos , Carcinoma Hepatocelular/virología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virología , Neoplasias Hepáticas/metabolismo , Masculino , Femenino , Persona de Mediana Edad , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , FN-kappa B/metabolismo , Adulto , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , Virus de la Hepatitis B/genética , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , Anciano , Regulación Neoplásica de la Expresión Génica , Ubiquitinas/genética , Ubiquitinas/metabolismo , Hepatitis B/complicaciones , Hepatitis B/genética
16.
Gene ; 927: 148649, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-38852697

RESUMEN

During the birch pollen season an enhanced incidence of virus infections is noticed, raising the question whether pollen can affect anti-viral responses independent of allergic reactions. We previously showed that birch pollen-treatment of monocyte-derived dendritic cells (moDC) enhances human cytomegalovirus (HCMV) infection. Here we addressed how in moDC the relatively weak pollen response can affect the comparably strong response to HCMV. To this end, moDC were stimulated with aqueous birch pollen extract (APE), HCMV, and APE with HCMV, and transcriptomic signatures were determined after 6 and 24 h of incubation. Infection was monitored upon exposure of moDC to GFP expressing HCMV by flow cytometric analysis of GFP expressing cells. Principle component analysis of RNA sequencing data revealed close clustering of mock and APE treated moDC, whereas HCMV as well as APE with HCMV treated moDC clustered separately after 6 and 24 h of incubation, respectively. Communally induced genes were detected in APE, HCMV and APE with HCMV treated moDC. In APE with HCMV treated moDC, the comparably weak APE induced signatures were maintained after HCMV exposure. In particular, NF-κB/RELA and PI3K/AKT/MAPK signaling were altered upon APE with HCMV exposure. Earlier, we discovered that NF-κB inhibition alleviated APE induced enhancement of HCMV infection. Here we additionally found that impairment of PI3K signaling reduced HCMV infection in HCMV and APE with HCMV treated moDC. APE treated moDC that were exposed to HCMV show a unique host gene signature, which to a large extent is regulated by NF-κB activation and PI3K/AKT/MAPK signaling.


Asunto(s)
Betula , Citomegalovirus , Células Dendríticas , Polen , Células Dendríticas/virología , Células Dendríticas/metabolismo , Células Dendríticas/inmunología , Polen/genética , Polen/inmunología , Citomegalovirus/genética , Citomegalovirus/fisiología , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Infecciones por Citomegalovirus/virología , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/inmunología , Transcriptoma , Transducción de Señal , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , Células Cultivadas
17.
Parasit Vectors ; 17(1): 239, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802961

RESUMEN

BACKGROUND: The spleen plays a critical role in the immune response against malaria parasite infection, where splenic fibroblasts (SFs) are abundantly present and contribute to immune function by secreting type I collagen (collagen I). The protein family is characterized by Plasmodium vivax tryptophan-rich antigens (PvTRAgs), comprising 40 members. PvTRAg23 has been reported to bind to human SFs (HSFs) and affect collagen I levels. Given the role of type I collagen in splenic immune function, it is important to investigate the functions of the other members within the PvTRAg protein family. METHODS: Protein structural prediction was conducted utilizing bioinformatics analysis tools and software. A total of 23 PvTRAgs were successfully expressed and purified using an Escherichia coli prokaryotic expression system, and the purified proteins were used for co-culture with HSFs. The collagen I levels and collagen-related signaling pathway protein levels were detected by immunoblotting, and the relative expression levels of inflammatory factors were determined by quantitative real-time PCR. RESULTS: In silico analysis showed that P. vivax has 40 genes encoding the TRAg family. The C-terminal region of all PvTRAgs is characterized by the presence of a domain rich in tryptophan residues. A total of 23 recombinant PvTRAgs were successfully expressed and purified. Only five PvTRAgs (PvTRAg5, PvTRAg16, PvTRAg23, PvTRAg30, and PvTRAg32) mediated the activation of the NF-κBp65 signaling pathway, which resulted in the production of inflammatory molecules and ultimately a significant reduction in collagen I levels in HSFs. CONCLUSIONS: Our research contributes to the expansion of knowledge regarding the functional role of PvTRAgs, while it also enhances our understanding of the immune evasion mechanisms utilized by parasites.


Asunto(s)
Antígenos de Protozoos , Colágeno Tipo I , Fibroblastos , Plasmodium vivax , Transducción de Señal , Bazo , Plasmodium vivax/genética , Plasmodium vivax/inmunología , Fibroblastos/parasitología , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Antígenos de Protozoos/metabolismo , Animales , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , Bazo/inmunología , Bazo/parasitología , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , Ratones , Humanos , Malaria Vivax/parasitología , Malaria Vivax/inmunología , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/inmunología , Triptófano/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Biología Computacional
18.
Sci Rep ; 14(1): 11211, 2024 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755247

RESUMEN

Lung adenocarcinoma (LUAD) is a malignancy with an abysmal survival rate. High metastasis is the leading cause of the low survival rate of LUAD. NCAPH, an oncogene, is involved in the carcinogenesis of LUAD. However, the regulation of NCAPH in LUAD remains controversial. In this work, we identified an up-regulation of NCAPH in LUAD tissues. Patients who expressed more NCAPH had shorter overall survival (OS). Furthermore, NCAPH overexpression promoted LUAD cell migration while inhibiting apoptosis. MiR-1976 and miR-133b were predicted to target NCAPH expression by searching TargetScan and linkedomics databases. Following that, we confirmed that miR-1976 suppressed NCAPH by directly targeting a 7-bp region of NCAPH 3' untranslated regions (UTR). In addition, increased expression of miR-1976 decreased the proliferation & migration and promoted apoptosis of LUAD cells, and the re-introduction of NCAPH reversed these influences. Furthermore, the xenograft and metastasis mouse models also confirmed that miR-1976 inhibited tumor growth and metastasis in vivo by targeting NCAPH. Finally, we found that MiR-1976 targeting NCAPH blocked the activation of NF-κB. In conclusion, miR-1976 inhibits NCAPH activity in LUAD and acts as a tumor suppressor. The miR-1976/NCAPH/NF-κB axis may, in the future, represent crucial diagnostic and prognostic biomarkers and promising therapeutic options.


Asunto(s)
Adenocarcinoma del Pulmón , Apoptosis , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares , MicroARNs , Factor de Transcripción ReIA , Animales , Femenino , Humanos , Masculino , Ratones , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/metabolismo , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Fenotipo , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética
19.
Bull Exp Biol Med ; 176(5): 562-566, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38724811

RESUMEN

We studied the effect of an NO donor, nitrosyl iron complex with N-ethylthiourea, on Nrf2-dependent antioxidant system activation of tumor cells in vitro. The complex increased intracellular accumulation of Nrf2 transcription factor and induced its nuclear translocation. It was shown that both heme oxygenase-1 gene and protein expression increased significantly under the influence of the complex. Nrf2 activation was accompanied by a decrease in the intracellular accumulation of proinflammatory transcription factor NF-κB p65 subunit and expression of its target genes. The cytotoxic effect of N-ethylthiourea leads to induction of Nrf2/HO-1 antioxidant response and suppression of NF-κB-dependent processes in tumor cells.


Asunto(s)
Hemo-Oxigenasa 1 , Hierro , Factor 2 Relacionado con NF-E2 , Tiourea , Humanos , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Tiourea/análogos & derivados , Tiourea/farmacología , Células HeLa , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/genética , Hierro/metabolismo , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética , Óxidos de Nitrógeno/metabolismo , Óxidos de Nitrógeno/farmacología , Antioxidantes/farmacología
20.
Biochem Biophys Res Commun ; 722: 150143, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-38795451

RESUMEN

Nuclear factor (NF)-κB signaling is not only important for the immune and inflammatory responses but also for the normal development of epithelial cells, such as those in the skin and tooth. Here, we generated epithelial cell-specific p65-deficient (p65Δepi-/-) mice to analyze the roles of NF-κB signaling in epithelial cell developent. Notably, p65Δepi-/- mice exhibited no abnormalities in their appearance compared to the control (p65flox/flox) littermates. Furthermore, no major changes were observed in the skin, hair growth, and shape and color of the incisors and molars. However, 65 % of p65Δepi-/- mice exhibited corneal thickening after 8 weeks of age, and 30 % of p65Δepi-/- mice exhibited hair growth from the mandibular incisors around 24 weeks of age. No hair growth was observed at 36 and 42 weeks of age. However, micro-computed tomography images revealed a large cavity below the mandibular incisors extending to the root of the incisor. Histological analysis revealed that the cavity was occupied by a connective tissue containing hair-like structures with many dark brown granules that disappeared after melanin bleaching, confirming the presence of hair. Although inflammatory cells were also observed near the eruption site of the incisor teeth of p65Δepi-/- mice, no major disturbance was observed in the arrangement of enamel epithelial cells. Overall, these results highlight the role of p65 in the maintenance of epithelial cell homeostasis during aging.


Asunto(s)
Senescencia Celular , Células Epiteliales , Factor de Transcripción ReIA , Animales , Ratones , Envejecimiento/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...