Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 836
Filtrar
1.
Anat Histol Embryol ; 53(3): e13044, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38695121

RESUMEN

The vitamin D receptor (VDR) signalling has been implicated in vertebrate limb or fin formation. However, the involvement of VDR signalling in the early stages of limb/fin development remains to be elucidated. In this study, the role of VDR signalling in pectoral fin development was investigated in zebrafish embryos. Knockdown of vdr induced the severe impairment of pectoral fin development. The zebrafish larvae lacking vdr exhibited reduced pectoral fins with no skeletal elements. In situ hybridization revealed depletion of vdr downregulated fibroblast growth factor 24 (fgf24), a marker of early pectoral fin bud mesenchyme, in the presumptive fin field even before fin buds were visible. Moreover, a perturbed expression pattern of bone morphogenetic protein 4 (bmp4), a marker of the pectoral fin fold, was observed in the developing fin buds of zebrafish embryos that lost the vdr function. These findings suggest that VDR signalling is crucial in the early stages of fin development, potentially influencing the process by regulating other signalling molecules such as Fgf24 and Bmp4.


Asunto(s)
Aletas de Animales , Proteína Morfogenética Ósea 4 , Factores de Crecimiento de Fibroblastos , Receptores de Calcitriol , Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/genética , Pez Cebra/embriología , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Aletas de Animales/embriología , Aletas de Animales/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/genética , Técnicas de Silenciamiento del Gen , Transducción de Señal , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ
2.
Endocrinology ; 165(6)2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38679470

RESUMEN

CONTEXT: Recurrent spontaneous abortion (RSA) is defined as the loss of 2 or more consecutive intrauterine pregnancies with the same sexual partner in the first trimester. Despite its significance, the etiology and underlying mechanisms of RSA remain elusive. Defective decidualization is proposed as one of the potential causes of RSA, with abnormal decidualization leading to disturbances in trophoblast invasion function. OBJECTIVE: To assess the role of bone morphogenetic protein 4 (BMP4) in decidualization and RSA. METHODS: Decidual samples were collected from both RSA patients and healthy controls to assess BMP4 expression. In vitro cell experiments utilized the hESC cell line to investigate the impact of BMP4 on decidualization and associated aging, as well as its role in the maternal-fetal interface communication. Subsequently, a spontaneous abortion mouse model was established to evaluate embryo resorption rates and BMP4 expression levels. RESULTS: Our study identified a significant downregulation of BMP4 expression in the decidua of RSA patients compared to the normal control group. In vitro, BMP4 knockdown resulted in inadequate decidualization and inhibited associated aging processes. Mechanistically, BMP4 was implicated in the regulation of FOXO1 expression, thereby influencing decidualization and aging. Furthermore, loss of BMP4 hindered trophoblast migration and invasion via FOXO1 modulation. Additionally, BMP4 downregulation was observed in RSA mice. CONCLUSION: Our findings highlighted the downregulation of BMP4 in both RSA patients and mice. BMP4 in human endometrial stromal cells was shown to modulate decidualization by regulating FOXO1 expression. Loss of BMP4 may contribute to the pathogenesis of RSA, suggesting potential avenues for abortion prevention strategies.


Asunto(s)
Aborto Habitual , Proteína Morfogenética Ósea 4 , Decidua , Endometrio , Proteína Forkhead Box O1 , Células del Estroma , Femenino , Humanos , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Células del Estroma/metabolismo , Animales , Ratones , Decidua/metabolismo , Embarazo , Endometrio/metabolismo , Endometrio/citología , Aborto Habitual/metabolismo , Aborto Habitual/genética , Adulto , Trofoblastos/metabolismo , Estudios de Casos y Controles
3.
Cell Commun Signal ; 22(1): 248, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689334

RESUMEN

BACKGROUND: Bone morphogenetic protein 4 (BMP4) is a potent inhibitor of breast cancer metastasis. However, a tumor-promoting effect of BMP4 is reported in other tumor types, especially when SMAD4 is inactive. METHODS: To assess the requirement for SMAD4 in BMP4-mediated suppression of metastasis, we knocked down SMAD4 in two different breast tumors and enforced SMAD4 expression in a third line with endogenous SMAD4 deletion. In addition, we assessed the requirement for SMAD4 in tumor cell-specific BMP signalling by expression of a constitutively active BMP receptor. Delineation of genes regulated by BMP4 in the presence or absence of SMAD4 was assessed by RNA sequencing and a BMP4-induced gene, MYO1F was assessed for its role in metastasis. Genes regulated by BMP4 and/or SMAD4 were assessed in a publicly available database of gene expression profiles of breast cancer patients. RESULTS: In the absence of SMAD4, BMP4 promotes primary tumor growth that is accompanied by increased expression of genes associated with DNA replication, cell cycle, and MYC signalling pathways. Despite increased primary tumor growth, BMP4 suppresses metastasis in the absence of tumor cell expression of SMAD4. Consistent with the anti-metastatic activity of BMP4, enforced signalling through the constitutively active receptor in SMAD4 positive tumors that lacked BMP4 expression still suppressed metastasis, but in the absence of SMAD4, the suppression of metastasis was largely prevented. Thus BMP4 is required for suppression of metastasis regardless of tumor SMAD4 status. The BMP4 upregulated gene, MYO1F, was shown to be a potent suppressor of breast cancer metastasis. Gene signature upregulated by BMP4 in the absence of SMAD4 was associated with poor prognosis in breast cancer patients, whereas gene signature upregulated by BMP4 in the presence of SMAD4 was associated with improved prognosis. CONCLUSIONS: BMP4 expression is required for suppression of metastasis regardless of the SMAD4 status of the tumor cells. Since BMP4 is a secreted protein, we conclude that it can act both in an autocrine manner in SMAD4-expressing tumor cells and in a paracrine manner on stromal cells to suppress metastasis. Deletion of SMAD4 from tumor cells does not prevent BMP4 from suppressing metastasis via a paracrine mechanism.


Asunto(s)
Proteína Morfogenética Ósea 4 , Neoplasias de la Mama , Metástasis de la Neoplasia , Transducción de Señal , Proteína Smad4 , Proteína Smad4/genética , Proteína Smad4/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Humanos , Animales , Femenino , Línea Celular Tumoral , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Ratones , Proliferación Celular/genética
4.
J Mol Med (Berl) ; 102(5): 693-707, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38492027

RESUMEN

Physical therapy is extensively employed in clinical settings. Nevertheless, the absence of suitable animal models has resulted in an incomplete understanding of the in vivo mechanisms and cellular distribution that respond to physical stimuli. The objective of this research was to create a mouse model capable of indicating the cells affected by physical stimuli. In this study, we successfully established a mouse line based on the heat shock protein 70 (Hsp70) promoter, wherein the expression of CreERT2 can be induced by physical stimuli. Following stimulation of the mouse tail, ear, or cultured calvarias with heat shock (generated by heating, ultrasound, or laser), a distinct Cre-mediated excision was observed in cells stimulated by these physical factors with minimal occurrence of leaky reporter expression. The application of heat shock to Hsp70-CreERT2; FGFR2-P253R double transgenic mice or Hsp70-CreERT2 mice infected with AAV-BMP4 at calvarias induced the activation of Cre-dependent mutant FGFR2-P253R or BMP4 respectively, thereby facilitating the premature closure of cranial sutures or the repair of calvarial defects. This novel mouse line holds significant potential for investigating the underlying mechanisms of physical therapy, tissue repair and regeneration, lineage tracing, and targeted modulation of gene expression of cells in local tissue stimulated by physical factor at the interested time points. KEY MESSAGES: In the study, an Hsp70-CreERT2 transgenic mouse was generated for heat shock-induced gene modulation. Heat shock, ultrasound, and laser stimulation effectively activated Cre expression in Hsp70-CreERT2; reporter mice, which leads to deletion of floxed DNA sequence in the tail, ear, and cultured calvaria tissues of mice. Local laser stimuli on cultured calvarias effectively induce Fgfr2-P253R expression in Hsp70-mTmG-Fgfr2-P253R mice and result in accelerated premature closure of cranial suture. Heat shock activated AAV9-FLEX-BMP4 expression and subsequently promoted the repair of calvarial defect of Hsp70-CreERT2; Rosa26-mTmG mice.


Asunto(s)
Proteína Morfogenética Ósea 4 , Proteínas HSP70 de Choque Térmico , Ratones Transgénicos , Regiones Promotoras Genéticas , Animales , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Ratones , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/genética , Respuesta al Choque Térmico/genética , Cráneo/metabolismo , Regulación de la Expresión Génica , Integrasas/metabolismo , Integrasas/genética
5.
Mol Cells ; 47(4): 100058, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38522664

RESUMEN

A comprehensive regulatory network of transcription factors controls the dorsoventral patterning of the body axis in developing vertebrate embryos. Bone morphogenetic protein signaling is essential for activating the Ventx family of homeodomain transcription factors, which regulates embryonic patterning and germ layer identity during Xenopus gastrulation. Although Ventx1.1 and Ventx2.1 of the Xenopus Ventx family have been extensively investigated, Ventx3.2 remains largely understudied. Therefore, this study aimed to investigate the transcriptional regulation of ventx3.2 during the embryonic development of Xenopus. We used goosecoid (Gsc) genome-wide chromatin immunoprecipitation-sequencing data to isolate and replicate the promoter region of ventx3.2. Serial deletion and site-directed mutagenesis were used to identify the cis-acting elements for Gsc and caudal type homeobox 1 (Cdx1) within the ventx3.2 promoter. Cdx1 and Gsc differentially regulated ventx3.2 transcription in this study. Additionally, positive cis-acting and negative response elements were observed for Cdx1 and Gsc, respectively, within the 5' flanking region of the ventx3.2 promoter. This result was corroborated by mapping the active Cdx1 response element (CRE) and Gsc response element (GRE). Moreover, a point mutation within the CRE and GRE completely abolished the activator and repressive activities of Cdx1 and Gsc, respectively. Furthermore, the chromatin immunoprecipitation-polymerase chain reaction confirmed the direct binding of Cdx1 and Gsc to the CRE and GRE, respectively. Inhibition of Cdx1 and Gsc activities at their respective functional regions, namely, the ventral marginal zone and dorsal marginal zone, reversed their effects on ventx3.2 transcription. These results indicate that Cdx1 and Gsc modulate ventx3.2 transcription in the ventral marginal zone and dorsal marginal zone by directly binding to the promoter region during Xenopus gastrulation.


Asunto(s)
Gástrula , Proteínas de Homeodominio , Regiones Promotoras Genéticas , Proteínas de Xenopus , Animales , Regiones Promotoras Genéticas/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Gástrula/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/genética , Xenopus laevis/genética , Xenopus laevis/metabolismo , Unión Proteica , Transcripción Genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteína Goosecoide/genética , Proteína Goosecoide/metabolismo
6.
Int J Biol Macromol ; 265(Pt 1): 130649, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38453121

RESUMEN

Bone Morphogenetic Protein 4 (BMP4) is crucial for bone and cartilage tissue regeneration, essential in medical tissue engineering, cosmetology, and aerospace. However, its cost and degradation susceptibility pose significant clinical challenges. To enhance its osteogenic activity while reducing dosage and administration frequency, we developed a novel long-acting BMP4 delivery system using poly(3-hydroxybutyrate-co-3-hydroxyvalerate-co-3-hydroxyhexanoate) (PBVHx) nanoparticles with soybean lecithin-modified BMP4 (sBP-NPs). These nanoparticles promote directed osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) through sustained BMP4 release. sBP-NPs exhibited uniform size (100-200 nm) and surface charges, with higher BMP4 entrapment efficiency (82.63 %) compared to controls. After an initial burst release within 24 h, sBP-NPs achieved 80 % cumulative BMP4 release within 20 days, maintaining levels better than control BP-NPs with unmodified BMP4. Co-incubation and nanoparticle uptake experiments confirmed excellent biocompatibility of sBP-NPs, promoting hBMSC differentiation towards osteogenic lineage with increased expression of type I collagen, calcium deposition, and ALP activity (> 20,000 U/g protein) compared to controls. Moreover, hBMSCs treated with sBP-NPs exhibited heightened expression of osteogenic genetic markers, surpassing control groups. Hence, this innovative strategy of sustained BMP4 release from sBP-NPs holds potential to revolutionize bone regeneration in minimally invasive surgery, medical cosmetology or space environments.


Asunto(s)
Células Madre Mesenquimatosas , Nanopartículas , Humanos , Osteogénesis/genética , Proteína Morfogenética Ósea 4/genética , Preparaciones de Acción Retardada/farmacología , Diferenciación Celular , Células de la Médula Ósea/metabolismo , Células Cultivadas
7.
Neoplasia ; 49: 100972, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38237535

RESUMEN

Papillary thyroid cancer (PTC) is the most prevalent endocrine cancer worldwide. Approximately 30 % of PTC patients will progress into the advanced or metastatic stage and have a relatively poor prognosis. It is well known that epithelial-mesenchymal transition (EMT) plays a pivotal role in thyroid cancer metastasis, resistance to therapy, and recurrence. Clarifying the molecular mechanisms of EMT in PTC progression will help develop the targeted therapy of PTC. The aberrant expression of some transcription factors (TFs) participated in many pathological processes of cancers including EMT. In this study, by performing bioinformatics analysis, adipocyte enhancer-binding protein 1 (AEBP1) was screened as a pivotal TF that promoted EMT and tumor progression in PTC. In vitro experiments indicated that knockout of AEBP1 can inhibit the growth and invasion of PTC cells and reduce the expression of EMT markers including N-cadherin, TWIST1, and ZEB2. In the xenograft model, knockout of AEBP1 inhibited the growth and lung metastasis of PTC cells. By performing RNA-sequencing, dual-luciferase reporter assay, and chromatin immunoprecipitation assay, Bone morphogenetic protein 4 (BMP4) was identified as a downstream target of AEBP1. Over-expression of BMP4 can rescue the inhibitory effects of AEBP1 knockout on the growth, invasion, and EMT phenotype of PTC cells. In conclusion, these findings demonstrated that AEBP1 plays a critical role in PTC progression by regulating BMP4 expression and the AEBP1-BMP4 axis may present novel therapeutic targets for PTC treatment.


Asunto(s)
MicroARNs , Neoplasias de la Tiroides , Humanos , Cáncer Papilar Tiroideo/metabolismo , MicroARNs/genética , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Transición Epitelial-Mesenquimal/genética , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica , Carboxipeptidasas/genética , Carboxipeptidasas/metabolismo , Proteínas Represoras/genética
8.
J Photochem Photobiol B ; 250: 112828, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38101122

RESUMEN

Rheumatoid arthritis (RA) is caused by inflammatory response of joints with cartilage and damage of synovium and bone erosion. In our previous studies, it has showed that irradiation of 630 nm LED reduce inflammation of synovial fibroblasts and cartilage and bone destruction in RA. However, the key genes and mechanism in ameliorating RA by irradiation of 630 nm LED remains unknown. In this study, human fibroblast-like synoviocytes (FLS) cell line MH7A and primary human RA-FLSs were treated with TNF-α and 630 nm LED irradiation with the different energy density. The mRNA sequencing was performed to screen the differentially expressed genes (DEGs). In all datasets, 10 DEGs were identified through screening. The protein interaction network analysis showed that 8 out of the 10 DEGs interacted with each other including IL-6, CXCL2, CXCL3, MAF, PGF, IL-1RL1, RRAD and BMP4. This study focused on BMP4, which is identified as important morphogens in regulating the development and homeostasis. CCK-8 assay results showed that 630 nm LED irradiation did not affect the cell viability. The qPCR and ELISA results showed that TNF-α stimulation inhibited BMP4 mRNA and protein level and irradiation of 630 nm LED increased the BMP4 mRNA and protein level in MH7A cells. In CIA and transgenic hTNF-α mice models, H&E staining showed that irradiation of 630 nm LED decreased the histological scores assessed from inflammation and bone erosion, while BMP4 expression level was up-regulated after 630 nm LED irradiation. Pearson correlation analysis shown that BMP4 protein expression was negatively correlated with the histological score of CIA mice and transgenic hTNF-α mice. These results indicated that BMP4 increased by irradiation of 630 nm LED was associated with the amelioration of RA, which suggested that BMP4 may be a potential targeting gene for photobiomodulation.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Proteína Morfogenética Ósea 4 , Luz , Animales , Humanos , Ratones , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Artritis Reumatoide/terapia , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/fisiología , Proliferación Celular , Células Cultivadas , Fibroblastos/metabolismo , Inflamación/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Membrana Sinovial/metabolismo , Membrana Sinovial/patología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
9.
Int J Mol Sci ; 24(21)2023 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-37958648

RESUMEN

The enteric nervous system (ENS) is principally derived from vagal neural crest cells that migrate caudally along the entire length of the gastrointestinal tract, giving rise to neurons and glial cells in two ganglionated plexuses. Incomplete migration of enteric neural crest-derived cells (ENCDC) leads to Hirschsprung disease, a congenital disorder characterized by the absence of enteric ganglia along variable lengths of the colorectum. Our previous work strongly supported the essential role of the avian ceca, present at the junction of the midgut and hindgut, in hindgut ENS development, since ablation of the cecal buds led to incomplete ENCDC colonization of the hindgut. In situ hybridization shows bone morphogenetic protein-4 (BMP4) is highly expressed in the cecal mesenchyme, leading us to hypothesize that cecal BMP4 is required for hindgut ENS development. To test this, we modulated BMP4 activity using embryonic intestinal organ culture techniques and retroviral infection. We show that overexpression or inhibition of BMP4 in the ceca disrupts hindgut ENS development, with GDNF playing an important regulatory role. Our results suggest that these two important signaling pathways are required for normal ENCDC migration and enteric ganglion formation in the developing hindgut ENS.


Asunto(s)
Neoplasias Colorrectales , Sistema Nervioso Entérico , Humanos , Transducción de Señal/fisiología , Diferenciación Celular/fisiología , Sistema Nervioso Entérico/metabolismo , Movimiento Celular/fisiología , Neoplasias Colorrectales/metabolismo , Cresta Neural/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo
10.
J Steroid Biochem Mol Biol ; 235: 106410, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37858799

RESUMEN

Polycystic ovary syndrome (PCOS) is a complex reproductive endocrine disease characterized by ovulation dysfunction with multiple etiologies and manifestations, and it is widely believed that the disorders of hyper-androgen and glucose metabolism play a key role in its progression. There has been evidence that bone morphogenetic protein 4 (BMP4) is essential for the regulation of granulosa cells, but whether it regulates metabolism level of granulosa cells under hyperandrogenic environment remains unclear. In this study, Gene Expression Omnibus, clinical data and serum of PCOS patient were collected to detect androgen and BMP4 levels. KGN cells exposed to androgens as a model for simulating PCOS granulosa cells. Lactate/pyruvate kits, and Extracellular Acidification Rate and Oxygen Consumption Rate assay were performed to detect glycolysis and autophagy levels of granulosa cells. Lentivirus infection was used to investigate the effects of BMP4 on granulosa cells. RNA-seq were performed to explore the special mechanism. We found that BMP4 was increased in PCOS patients with hyper-androgen and granulosa cells with dihydrotestosterone treatment. Mechanically, on the one hand, hyperandrogenemia can up-regulate BMP4 secretion and induce glycolysis and autophagy levels. On the other hand, we found that hyperandrogenic-induced YAP1 upregulation may mediate BMP4 to increase glycolysis level and decrease autophagy, which plays a protective role in granulosa cells to ensure subsequent energy utilization and mitochondrial function. Overall, we innovated on the protective effect of BMP4 on glycolysis and autophagy disorders induced by excessive androgen in granulosa cells. Our study will provide guidance for future understanding of PCOS from a metabolic perspective and for exploring treatment options.


Asunto(s)
Proteína Morfogenética Ósea 4 , Síndrome del Ovario Poliquístico , Femenino , Humanos , Andrógenos/farmacología , Andrógenos/metabolismo , Autofagia , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Glucosa/metabolismo , Células de la Granulosa/metabolismo , Síndrome del Ovario Poliquístico/metabolismo
11.
Vascul Pharmacol ; 153: 107236, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37774964

RESUMEN

Intracranial aneurysms (IAs) are characterized by abnormal dilatation of the cerebral vessels. Vascular smooth muscle cells (VSMCs) are implicated in maintaining vascular homeostasis. Disordered VSMCs are one of the most common causes for occurrence and development of IAs. The bone morphogenetic protein 4 (BMP4) signalling pathway is involved in regulating cell proliferation, apoptosis, and differentiation. This study aimed to investigate the effects of BMP4 on VSMCs and its underlying mechanisms. BMP4 was upregulated in the VSMCs of IAs and caused apoptosis of VSMCs through Smad1/5 phosphorylation. In addition, BMP4 overexpression significantly promoted the proliferation and migration of VSMCs and induced a phenotypic transformation from contractile to inflammatory. Our findings facilitate further understanding of the occurrence and development of IAs and provide a potential therapeutic target.


Asunto(s)
Aneurisma Intracraneal , Músculo Liso Vascular , Humanos , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/farmacología , Músculo Liso Vascular/metabolismo , Aneurisma Intracraneal/metabolismo , Transducción de Señal , Proliferación Celular , Miocitos del Músculo Liso/metabolismo , Células Cultivadas
12.
Int J Mol Sci ; 24(16)2023 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-37628872

RESUMEN

The cytosine-phosphate-guanine (CpG) island methylator phenotype (CIMP) represents one of the pathways involved in the development of colorectal cancer, characterized by genome-wide hypermethylation. To identify samples exhibiting hypermethylation, we used unsupervised hierarchical clustering on genome-wide methylation data. This clustering analysis revealed the presence of four distinct subtypes within the tumor samples, namely, CIMP-H, CIMP-L, cluster 3, and cluster 4. These subtypes demonstrated varying levels of methylation, categorized as high, intermediate, and very low. To gain further insights, we mapped significant probes from all clusters to Ensembl Regulatory build 89, with a specific focus on those located within promoter regions or bound regions. By intersecting the methylated promoter and bound regions across all methylation subtypes, we identified a total of 253 genes exhibiting aberrant methylation patterns in the promoter regions across all four subtypes of colorectal cancer. Among these genes, our comprehensive genome-wide analysis highlights bone morphogenic protein 4 (BMP4) as the most prominent candidate. This significant finding was derived through the utilization of various bioinformatics tools, emphasizing the potential role of BMP4 in colorectal cancer development and progression.


Asunto(s)
Neoplasias Colorrectales , Humanos , Metilación , Proteína Morfogenética Ósea 4/genética , Análisis por Conglomerados , Regiones Promotoras Genéticas , Neoplasias Colorrectales/genética
13.
Anim Sci J ; 94(1): e13866, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37632404

RESUMEN

This study investigated the expression dynamics of bone morphogenetic protein 4 (BMP4) and its receptors (BMPR1A, BMPR1B, and BMPR2) in bovine endometrium and examined the physiological function and regulatory mechanism of BMP4 expression. The messenger RNA (mRNA) expression of BMP4 and its receptors was detected in bovine endometrium of both ipsilateral (corpus luteum [CL]-side) and contralateral (non-CL-side) uterine horns during the estrous cycle and early pregnancy. BMP4 protein levels were higher in the endometrial tissues obtained from those cows in early pregnancy than in the estrous cycle. Immunohistochemical analysis showed that BMP4 and its receptors were localized in endometrial epithelial cells. The addition of BMP4 to cultured endometrial epithelial cells did not affect caspase-3/-8 mRNA expression, whereas it significantly inhibited cell proliferation. Both prostaglandin (PG) E2 and PGF2α concentrations in the culture supernatant were decreased when stimulated by BMP4. Furthermore, BMP4 mRNA expression was increased by stimulation with tumor necrosis factor-α (TNF) and interferon-γ (IFNG). In conclusion, BMP4 is produced in bovine endometrial epithelial cells and may contribute to the regulation of cell proliferation and suppression of PG secretion through autocrine or paracrine mechanisms. BMP4 expression in the bovine endometrium may be regulated by TNF and IFNG.


Asunto(s)
Dinoprostona , Endometrio , Embarazo , Femenino , Bovinos , Animales , Proteína Morfogenética Ósea 4/genética , Endometrio/metabolismo , Dinoprostona/metabolismo , Dinoprost/metabolismo , ARN Mensajero/metabolismo
14.
J Mol Neurosci ; 73(9-10): 713-723, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37632651

RESUMEN

Most neurodegenerative diseases are exacerbated by aging, with symptoms often worsening over time. Programmed cell death (PCD) is a controlled cell suicide mechanism that is essential for the stability, growth, and homeostasis of organisms. Understanding the effects of aging at the level of systems biology could lead to new therapeutic approaches for a broad spectrum of neurodegenerative diseases. In the absence of comprehensive functional studies on the relationship between PCD and aging of the prefrontal cortex, this study provides prefrontal brain biomarkers of aging associated with PCD that could open the way for improved therapeutic techniques for age-related neurodegenerative diseases. To this end, publicly available transcriptome data were subjected to bioinformatic analyses such as differential gene expression, functional enrichment, and the weighted gene coexpression network analysis (WGCNA). The diagnostic utility of the biomarkers was tested using a logistic regression-based prediction model. Three genes, namely BMP4, SGSH, and SLC11A2, were found to be aging biomarkers associated with PCD. Finally, a multifactorial regulatory network with interacting proteins, transcription factors (TFs), competing endogenous RNAs (ceRNAs), and microRNAs (miRNAs) was constructed around these biomarkers. The elements of this multifactorial regulatory network were mainly enriched in BMP signaling. Further exploration of these three biomarkers and their regulatory elements would enable the development of 3PM (predictive, preventive, and personalized) medicine for the treatment of age-related neurodegenerative diseases.


Asunto(s)
MicroARNs , Enfermedades Neurodegenerativas , ARN Largo no Codificante , Humanos , Apoptosis/genética , MicroARNs/metabolismo , Envejecimiento/genética , Biomarcadores , Redes Reguladoras de Genes , ARN Largo no Codificante/metabolismo , Proteína Morfogenética Ósea 4/genética
15.
Exp Cell Res ; 427(1): 113585, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37030332

RESUMEN

Cervical cancer is the fourth most common malignancy tumor worldwide with high incidence and mortality. Accumulating evidence indicated that through an m6A-dependent or m6A-independent mechanism, fat mass and obesity associated gene (FTO) exhibits the tumor-promoting and suppressive roles of FTO involved in various cancers, including cervical cancer. This study aims to verify the biological function and potential mechanisms of FTO in cervical cancer cell proliferation, colony formation, migration, and invasion in vitro as well as tumor growth in vivo. Herein, we confirmed that knockdown of FTO inhibits cell proliferation, colony formation, migration, and invasion of cervical cancer cells in vitro via cell counting kit-8 (CCK8) assay, colony formation assay, and transwell migration and invasion assay. The demethylase activity of FTO is required for cell proliferation, colony formation, migration, and invasion of cervical cancer cells in vitro. RNA sequencing, online database analysis, and western blotting revealed that FTO regulated the BMP4/Hippo/YAP1/TAZ pathway. In addition, FTO upregulates the expression of BMP4 in an m6A-dependent manner and binds to the N-terminal of BMP4 to form a dimer at the C-terminal in cervical cancer cells through protein-protein interaction. We further discovered that BMP4 treatment promoted cell proliferation, colony formation, migration, and invasion of cervical cancer cells, and rescue experiments validated that BMP4 treatment reversed the inhibition of FTO knockdown on the Hippo/YAP1/TAZ pathway and the progression of cervical cancer cells in vitro. Notably, the knockdown of FTO significantly suppressed xenograft tumor growth and the protein level of BMP4 in vivo. Collectively, our results demonstrate that the FTO promotes cervical cancer progression in vitro and in vivo via the regulation of the BMP4/Hippo/YAP1/TAZ pathway, suggesting that FTO acts as an oncogenic molecule and the FTO/BMP4 Hippo/YAP1/TAZ axis may serve as valuable targets for cervical cancer treatment.


Asunto(s)
Neoplasias del Cuello Uterino , Femenino , Humanos , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Proteína Morfogenética Ósea 4/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Vía de Señalización Hippo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
16.
EMBO J ; 42(10): e112196, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-36994549

RESUMEN

Blood vessels can play dual roles in tissue growth by transporting gases and nutrients and by regulating tissue stem cell activity via signaling. Correlative evidence implicates skin endothelial cells (ECs) as signaling niches of hair follicle stem cells (HFSCs), but functional demonstration from gene depletion of signaling molecules in ECs is missing to date. Here, we show that depletion of the vasculature-factor Alk1 increases BMP4 secretion from ECs, which delays HFSC activation. Furthermore, while previous evidence suggests a lymphatic vessel role in adult HFSC activation possibly through tissue drainage, a blood vessel role has not yet been addressed. Genetic perturbation of the ALK1-BMP4 axis in all ECs or the lymphatic ECs specifically unveils inhibition of HFSC activation by blood vessels. Our work suggests a broader relevance of blood vessels, adding adult HFSCs to the EC functional repertoire as signaling niches for the adult stem cells.


Asunto(s)
Receptores de Activinas Tipo II , Células Madre Adultas , Proteína Morfogenética Ósea 4 , Folículo Piloso , Animales , Ratones , Células Endoteliales , Transducción de Señal , Células Madre , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo
17.
Curr Mol Med ; 23(4): 324-331, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36883260

RESUMEN

Bone morphogenetic protein 4 (BMP4) is a multifunctional secretory protein that belongs to the transforming growth factor ß superfamily. BMPs transduce their signaling to the cytoplasm by binding to membrane receptors of the serine/threonine kinase family, including BMP type I and type II receptors. BMP4 participates in various biological processes, such as embryonic development, epithelial-mesenchymal transition, and maintenance of tissue homeostasis. The interaction between BMP4 and the corresponding endogenous antagonists plays a key role in the precise regulation of BMP4 signaling. In this paper, we review the pathogenesis of BMP4-related lung diseases and the foundation on which BMP4 endogenous antagonists have been developed as potential targets.


Asunto(s)
Proteína Morfogenética Ósea 4 , Enfermedades Pulmonares , Femenino , Humanos , Embarazo , Proteína Morfogenética Ósea 4/antagonistas & inhibidores , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/fisiología , Enfermedades Pulmonares/tratamiento farmacológico , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/metabolismo , Proteínas Serina-Treonina Quinasas
18.
J Pediatr Surg ; 58(7): 1317-1321, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36906487

RESUMEN

OBJECTIVE: To study the influence of gene methylation in the Shh/Bmp4 signaling pathway on the enteric nervous system in the rectum of rat embryos with anorectal malformations (ARMs). METHODS: Pregnant Sprague Dawley rats were divided into three groups; two groups treated with either ethylene thiourea (ETU induce ARM) or ETU+5-azacitidine (5-azaC inhibit DNA methylation) and a normal control group. The levels of DNA methyltransferases (DNMT1, DNMT3a, DNMT3b), the methylation status of the Shh gene promoter region and the expression of the key components were detected by PCR, immunohistochemistry and western blotting. RESULTS: The expression of DNMTs in the rectal tissue of the ETU and ETU+5-azaC groups was higher than that of the control. The expression of DNMT1, DNMT3a and methylation level of the Shh gene promoter in the ETU group was higher than in the ETU+5-azaC group (P < 0.01). The methylation level of the Shh gene promoter was higher in the ETU+5-azaC group than in the control. The Shh and Bmp4 expression in the ETU and ETU+5-azaC groups were lower than in the control, and their expression in the ETU group was also lower than in the ETU+5-azaC group. CONCLUSION: The methylation status of genes in the rectum of the ARM rat model may be changed by intervention. The low methylation level of the Shh gene may promote the expression of key Shh/Bmp4 signaling pathway components.


Asunto(s)
Malformaciones Anorrectales , Recto , Embarazo , Femenino , Ratas , Animales , Recto/anomalías , Malformaciones Anorrectales/genética , Ratas Sprague-Dawley , Canal Anal/anomalías , Metilación de ADN , Transducción de Señal , Sistema Nervioso/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo
19.
Dev Dyn ; 252(6): 761-769, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36825302

RESUMEN

BACKGROUND: The Bone morphogenetic protein 4 (BMP4) precursor protein is cleaved at two sites to generate an active ligand and inactive prodomain. The ligand and prodomain form a noncovalent complex following the first cleavage, but dissociate after the second cleavage. Transient formation of this complex is essential to generate a stable ligand. Fibrillins (FBNs) bind to the prodomains of BMPs, and can regulate the activity of some ligands. Whether FBNs regulate BMP4 activity is unknown. RESULTS: Mice heterozygous for a null allele of Bmp4 showed incompletely penetrant kidney defects and females showed increased mortality between postnatal day 6 and 8. Removal of one copy of Fbn1 did not rescue or enhance kidney defects or lethality. The lungs of Fbn1+/- females had enlarged airspaces that were unchanged in Bmp4+/- ;Fbn1+/- mice. Additionally, removal of one or both alleles of Fbn1 had no effect on steady state levels of BMP4 ligand or on BMP activity in postnatal lungs. CONCLUSIONS: These findings do not support the hypothesis that FBN1 plays a role in promoting BMP4 ligand stability or signaling, nor do they support the alternative hypothesis that FBN1 sequesters BMP4 in a latent form, as is the case for other BMP family members.


Asunto(s)
Proteínas Morfogenéticas Óseas , Riñón , Femenino , Ratones , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Ligandos , Proteínas Morfogenéticas Óseas/metabolismo , Alelos , Riñón/metabolismo , Proteína Morfogenética Ósea 7 , Proteína Morfogenética Ósea 2
20.
Cell Mol Life Sci ; 80(3): 58, 2023 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-36746787

RESUMEN

The exposure to an unhealthy environment in utero can lead to the occurrence of cardiovascular diseases in the offspring. Glucocorticoids (GC) are essential for normal development and maturation of fetal organs and is a first-line treatment for pregnant women affected by autoimmune diseases. However, excess prenatal GC exposure might program the development of fetal organs and cause a number of chronic diseases in later life. Our previous studies indicated that cardiac functions were significantly compromised in rat offspring prenatally exposed to the synthetic glucocorticoid dexamethasone (DEX), only after ischemia-reperfusion. In the present study, we further observed that DNA hypermethylation of bone morphogenetic protein 4 (Bmp4) promoter in cardiomyocytes caused by prenatal DEX exposure substantially dampened the binding activity of transcription factor HIF-1α induced by cardiac ischemia. Therefore, prenatal DEX exposure inhibits the induction of BMP4 upon I/R and attenuates the protective effects of BMP4 in cardiomyocytes, which eventually manifests as malfunction of the adult heart. Moreover, we employed two cardiac-specific Bmp4 knock-in mouse models and found that in vivo BMP4 overexpression could rescue the cardiac dysfunction caused by prenatal GC exposure. In depth mechanistic research revealed that BMP4 protects the cardiomyocytes from mitophagy and apoptosis by attenuating mitochondrial PGC-1α expression in a p-Smad and Parkin-dependent manner. These findings suggest that prenatal GC exposure increases the susceptibility of the offspring's heart to a "second strike" after birth, due to the failure of hypoxia-induced HIF-1α transactivation of the hypermethylated Bmp4 promoter in cardiomyocytes. Pretreatment with the DNA methylation inhibitor, 5-Aza-2'-deoxycytidine, could be a potential therapeutic method for this programming effect of GC exposure during pregnancy on neonatal cardiac dysfunction.


Asunto(s)
Glucocorticoides , Cardiopatías , Animales , Femenino , Humanos , Ratones , Embarazo , Ratas , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/farmacología , Decitabina/metabolismo , Decitabina/farmacología , Metilación de ADN , Glucocorticoides/metabolismo , Cardiopatías/metabolismo , Miocitos Cardíacos/metabolismo , Estrés Oxidativo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA