Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 275
Filtrar
1.
Sci Adv ; 10(27): eadj4433, 2024 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-38959322

RESUMEN

Memory processes rely on a molecular signaling system that balances the interplay between positive and negative modulators. Recent research has focused on identifying memory-regulating genes and their mechanisms. Phospholipase C beta 1 (PLCß1), highly expressed in the hippocampus, reportedly serves as a convergence point for signal transduction through G protein-coupled receptors. However, the detailed role of PLCß1 in memory function has not been elucidated. Here, we demonstrate that PLCß1 in the dentate gyrus functions as a memory suppressor. We reveal that mice lacking PLCß1 in the dentate gyrus exhibit a heightened fear response and impaired memory extinction, and this excessive fear response is repressed by upregulation of PLCß1 through its overexpression or activation using a newly developed optogenetic system. Last, our results demonstrate that PLCß1 overexpression partially inhibits exaggerated fear response caused by traumatic experience. Together, PLCß1 is crucial in regulating contextual fear memory formation and potentially enhancing the resilience to trauma-related conditions.


Asunto(s)
Giro Dentado , Miedo , Memoria , Neuronas , Fosfolipasa C beta , Animales , Fosfolipasa C beta/metabolismo , Fosfolipasa C beta/genética , Miedo/fisiología , Giro Dentado/metabolismo , Giro Dentado/fisiología , Memoria/fisiología , Ratones , Neuronas/metabolismo , Neuronas/fisiología , Ratones Noqueados , Masculino , Optogenética , Ratones Endogámicos C57BL
2.
Cells ; 13(11)2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38891118

RESUMEN

Crohn's disease is a chronic, debilitating, inflammatory bowel disease. Here, we report a critical role of phospholipase C-ß3 (PLC-ß3) in intestinal homeostasis. In PLC-ß3-deficient mice, exposure to oral dextran sodium sulfate induced lethality and severe inflammation in the small intestine. The lethality was due to PLC-ß3 deficiency in multiple non-hematopoietic cell types. PLC-ß3 deficiency resulted in reduced Wnt/ß-catenin signaling, which is essential for homeostasis and the regeneration of the intestinal epithelium. PLC-ß3 regulated the Wnt/ß-catenin pathway in small intestinal epithelial cells (IECs) at transcriptional, epigenetic, and, potentially, protein-protein interaction levels. PLC-ß3-deficient IECs were unable to respond to stimulation by R-spondin 1, an enhancer of Wnt/ß-catenin signaling. Reduced expression of PLC-ß3 and its signature genes was found in biopsies of patients with ileal Crohn's disease. PLC-ß regulation of Wnt signaling was evolutionally conserved in Drosophila. Our data indicate that a reduction in PLC-ß3-mediated Wnt/ß-catenin signaling contributes to the pathogenesis of ileal Crohn's disease.


Asunto(s)
Enfermedad de Crohn , Fosfolipasa C beta , Vía de Señalización Wnt , Enfermedad de Crohn/patología , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/genética , Fosfolipasa C beta/metabolismo , Fosfolipasa C beta/genética , Animales , Humanos , Ratones , beta Catenina/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Íleon/patología , Íleon/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados
3.
Thorac Cancer ; 15(19): 1477-1489, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38778543

RESUMEN

BACKGROUND: Lung cancer is the most common malignant tumor. In the present study, we identified a long non-coding RNA (lncRNA) AC100826.1 (simplify to Lnc1), which was highly expressed in non-small cell lung cancer (NSCLC) tissues compared with the paracancerous tissues. We also observed the critical role of Lnc1 in regulating the metastasis ability of NSCLC cells. METHODS: RNA sequencing was performed to detect differential expression levels of lncRNAs in NSCLC tissues and its paracancerous tissues. Effects of Lnc1 on cell proliferation, invasion, and migration were determined by CCK-8, transwell and scratch assays. The xenograft experiment confirmed the effect of Lnc1 on NSCLC cells proliferation and migration abilities in vivo. RT-qPCR and western blots were performed to determine the expression levels of mRNAs and proteins. RESULTS: The expression level of Lnc1 was related to multiple pathological results, knockdown of Lnc1 can inhibit the proliferation and metastasis abilities of NSCLC cells. silencing phospholipase C, ß1(PLCB1) can reverse the promoting effects of overexpression Lnc1 on NSCLC cells proliferation and migration abilities. In addition, the Rap1 signaling pathway was implicated in the regulation of Lnc1 in NSCLC metastasis. CONCLUSION: Our results suggest that Lnc1 regulated the metastatic ability of NSCLC cells through targeting the PLCB1/Rap1 signal pathway.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Proliferación Celular , Neoplasias Pulmonares , Fosfolipasa C beta , ARN Largo no Codificante , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Humanos , ARN Largo no Codificante/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Ratones , Animales , Fosfolipasa C beta/metabolismo , Fosfolipasa C beta/genética , Movimiento Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Femenino , Masculino , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto , Línea Celular Tumoral
4.
Int J Mol Sci ; 25(9)2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38731807

RESUMEN

Fat tissue-a vital energy storage organ-is intricately regulated by various factors, including circular RNA, which plays a significant role in modulating fat development and lipid metabolism. Therefore, this study aims to clarify the regulatory mechanism of sheep adipocyte proliferation and differentiation by investigating the involvement of circTIAM1, miR-485-3p, and its target gene PLCB1. Through previous sequencing data, circTIAM1 was identified in sheep adipocytes, with its circularization mechanism elucidated, confirming its cytoplasmic localization. Experimental evidence from RNase R treatment and transcription inhibitors highlighted that circTIAM1 is more stable than linear RNA. Additionally, circTIAM1 promoted sheep adipocyte proliferation and differentiation. Furthermore, bioinformatic analysis demonstrated a robust interaction between miR-485-3p and circTIAM1. Further experiments revealed that miR-485-3p inhibits fat cell proliferation and differentiation by inhibiting PLCB1, with circTIAM1 alleviating the inhibitory effect via competitive binding. In summary, our findings elucidate the mechanism through which circTIAM1 regulates Guangling Large-Tailed sheep adipocyte proliferation and differentiation via the miR-485-3p-PLCB1 pathway, offering a novel perspective for further exploring fat metabolism regulation.


Asunto(s)
Adipocitos , Diferenciación Celular , MicroARNs , Fosfolipasa C beta , ARN Circular , Animales , Adipocitos/metabolismo , Adipocitos/citología , Diferenciación Celular/genética , Proliferación Celular/genética , MicroARNs/genética , MicroARNs/metabolismo , Fosfolipasa C beta/metabolismo , Fosfolipasa C beta/genética , ARN Circular/genética , ARN Circular/metabolismo , Ovinos , Transducción de Señal
5.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167207, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38701954

RESUMEN

PURPOSE: In this study, we identified and diagnosed a novel inherited condition called Dyschromatosis, Ichthyosis, Deafness, and Atopic Disease (DIDA) syndrome. We present a series of studies to clarify the pathogenic variants and specific mechanism. METHODS: Exome sequencing and Sanger sequencing was conducted in affected and unaffected family members. A variety of human and cell studies were performed to explore the pathogenic process of keratosis. RESULTS: Our finding indicated that DIDA syndrome was caused by compound heterozygous variants in the oxysterol-binding protein-related protein 2 (OSBPL2) gene. Furthermore, our findings revealed a direct interaction between OSBPL2 and Phosphoinositide phospholipase C-beta-3 (PLCB3), a key player in hyperkeratosis. OSBPL2 effectively inhibits the ubiquitylation of PLCB3, thereby stabilizing PLCB3. Conversely, OSBPL2 variants lead to enhanced ubiquitination and subsequent degradation of PLCB3, leading to epidermal hyperkeratosis, characterized by aberrant proliferation and delayed terminal differentiation of keratinocytes. CONCLUSIONS: Our study not only unveiled the association between OSBPL2 variants and the newly identified DIDA syndrome but also shed light on the underlying mechanism.


Asunto(s)
Sordera , Ictiosis , Linaje , Fosfolipasa C beta , Humanos , Sordera/genética , Sordera/patología , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Femenino , Masculino , Ictiosis/genética , Ictiosis/patología , Ictiosis/metabolismo , Heterocigoto , Ubiquitinación , Queratinocitos/metabolismo , Queratinocitos/patología , Secuenciación del Exoma , Adulto , Síndrome , Células HEK293 , Receptores de Esteroides
6.
Cancer Lett ; 588: 216746, 2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38387756

RESUMEN

Helicobacter pylori (H. pylori) infection is considered to be an important factor in gastric cancer (GC). Long noncoding RNA (lncRNA) and m6A modification are involved in the occurrence and development of GC, but the role of lncRNA m6A modification in the development of GC mediated by H. pylori is still unclear. Here, we found that H. pylori infection downregulated the expression of lnc-PLCB1 through METTL14-mediated m6A modification and IRF2-mediated transcriptional regulation. Overexpression of lnc-PLCB1 inhibited the proliferation and migration of GC cells, while downregulation of lnc-PLCB1 promoted the proliferation and migration ability of GC cells. In addition, clinical analysis showed that lnc-PLCB1 is lower in GC tissues than in normal tissues. Further study found that lnc-PLCB1 reduced the protein stability of its binding protein DEAD-box helicase 21 (DDX21) and then downregulated the expression of CCND1 and Slug, thereby playing tumour suppressing role in the occurrence and development of GC. In conclusion, the METTL14/lnc-PLCB1/DDX21 axis plays an important role in H. pylori-mediated GC, and lnc-PLCB1 can be used as a new target for GC treatment.


Asunto(s)
Adenina , Infecciones por Helicobacter , Helicobacter pylori , ARN Largo no Codificante , Neoplasias Gástricas , Humanos , Helicobacter pylori/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Neoplasias Gástricas/patología , Regulación hacia Abajo , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/genética , Proliferación Celular , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo
7.
Chem Senses ; 492024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-38183495

RESUMEN

The peripheral taste system is more complex than previously thought. The novel taste-signaling proteins TRPM4 and PLCß3 appear to function in normal taste responding as part of Type II taste cell signaling or as part of a broadly responsive (BR) taste cell that can respond to some or all classes of tastants. This work begins to disentangle the roles of intracellular components found in Type II taste cells (TRPM5, TRPM4, and IP3R3) or the BR taste cells (PLCß3 and TRPM4) in driving behavioral responses to various saccharides and other sweeteners in brief-access taste tests. We found that TRPM4, TRPM5, TRPM4/5, and IP3R3 knockout (KO) mice show blunted or abolished responding to all stimuli compared with wild-type. IP3R3 KO mice did, however, lick more for glucose than fructose following extensive experience with the 2 sugars. PLCß3 KO mice were largely unresponsive to all stimuli except they showed normal concentration-dependent responding to glucose. The results show that key intracellular signaling proteins associated with Type II and BR taste cells are mutually required for taste-driven responses to a wide range of sweet and carbohydrate stimuli, except glucose. This confirms and extends a previous finding demonstrating that Type II and BR cells are both necessary for taste-driven licking to sucrose. Glucose appears to engage unique intracellular taste-signaling mechanisms, which remain to be fully elucidated.


Asunto(s)
Glucosa , Fosfolipasa C beta , Canales Catiónicos TRPM , Gusto , Animales , Ratones , Carbohidratos , Glucosa/farmacología , Glucosa/metabolismo , Ratones Noqueados , Edulcorantes/farmacología , Gusto/genética , Gusto/fisiología , Percepción del Gusto , Canales Catiónicos TRPM/genética , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo
8.
ACS Synth Biol ; 13(1): 242-258, 2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38092428

RESUMEN

Cells experience time-varying and spatially heterogeneous chemokine signals in vivo, activating cell surface proteins including G protein-coupled receptors (GPCRs). The Gαq pathway activation by GPCRs is a major signaling axis with broad physiological and pathological significance. Compared with other Gα members, GαqGTP activates many crucial effectors, including PLCß (Phospholipase Cß) and Rho GEFs (Rho guanine nucleotide exchange factors). PLCß regulates many key processes, such as hematopoiesis, synaptogenesis, and cell cycle, and is therefore implicated in terminal-debilitating diseases, including cancer, epilepsy, Huntington's Disease, and Alzheimer's Disease. However, due to a lack of genetic and pharmacological tools, examining how the dynamic regulation of PLCß signaling controls cellular physiology has been difficult. Since activated PLCß induces several abrupt cellular changes, including cell morphology, examining how the other pathways downstream of Gq-GPCRs contribute to the overall signaling has also been difficult. Here we show the engineering, validation, and application of a highly selective and efficient optogenetic inhibitor (Opto-dHTH) to completely disrupt GαqGTP-PLCß interactions reversibly in user-defined cellular-subcellular regions on optical command. Using this newly gained PLCß signaling control, our data indicate that the molecular competition between RhoGEFs and PLCß for GαqGTP determines the potency of Gq-GPCR-governed directional cell migration.


Asunto(s)
Transducción de Señal , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Transducción de Señal/fisiología
9.
Am J Med Genet A ; 191(11): 2673-2678, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37596802

RESUMEN

Auriculocondylar syndrome (ARCND) is a rare craniofacial birth defect characterized by malformations in the mandible and external ear (Question Mark Ear). Genetically, three distinct subtypes of ARCND (ARCND1, ARCND2, and ARCND3) have been identified. ARCND2 is linked to pathogenic variants in the PLCB4 gene (phospholipase C ß4). PLCB4 is a key effector of the EDN1-EDNRA pathway involved in craniofacial development via the induction, migration, and maintenance of neural crest cells. ARCND2 is typically inherited in an autosomal dominant pattern, with recessive inheritance pattern being rare. In this study, we report the first homozygous missense variant (NM_000933.4: c.2050G>A: p.(Gly684Arg)) in the PLCB4 gene causing ARCND in a 3-year-old patient with a severe clinical phenotype of the syndrome. The patient presented with typical craniofacial ARCND features, in addition to intestinal transit defect, macropenis, and hearing loss. These findings further delineate the phenotypic spectrum of ARCND associated with autosomal recessive PLCB4 loss of function variants. Notably, our results provide further evidence that these variants can result in a more severe and diverse manifestations of the syndrome. Clinicians should consider the rare features of this condition for better management of patients.


Asunto(s)
Enfermedades del Oído , Humanos , Preescolar , Mutación , Enfermedades del Oído/diagnóstico , Enfermedades del Oído/genética , Enfermedades del Oído/patología , Fenotipo , Oído Externo , Linaje , Fosfolipasa C beta/genética
10.
Biochem Genet ; 61(6): 2618-2632, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37208557

RESUMEN

Phospholipase C Beta 1 (PLCB1) regulates the abundance of PI(4,5)P2 in the plasma membrane and is implicated in various kinds of cancers. This study aimed to investigate the role and underlying mechanisms of PLCB1 in gastric cancer. Herein, it was found that PLCB1 mRNA and protein were highly expressed in gastric cancer, and high levels of PLCB1 were correlated with poor outcomes of patients with gastric cancer via the GEPIA database. Moreover, our results revealed that PLCB1 depletion inhibited gastric cancer cell proliferation, migration, and invasion. Meanwhile, PLCB1 overexpression resulted in an inverse result. Furthermore, PLCB1 mediated actin cytoskeleton rearrangement and activated the RhoA/LIMK/Cofilin pathway. Besides, PLCB1 promoted the Epithelial-Mesenchymal transition process via activating ATK signaling. In conclusion, PLCB1 promoted gastric cancer cell migratory and invasive abilities via regulating actin cytoskeleton rearrangement and Epithelial-Mesenchymal transition process. These findings imply that targeting PLCB1 may be a potential strategy to improve the prognosis of gastric cancer patients.


Asunto(s)
Citoesqueleto de Actina , Transición Epitelial-Mesenquimal , Fosfolipasa C beta , Neoplasias Gástricas , Humanos , Movimiento Celular , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Fosfolipasa C beta/genética , Invasividad Neoplásica , Masculino , Femenino , Persona de Mediana Edad , Línea Celular Tumoral , Pronóstico
11.
Methods Enzymol ; 682: 17-52, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36948701

RESUMEN

Phospholipase C (PLC) enzymes convert the membrane phospholipid phosphatidylinositol-4,5-bisphosphate (PIP2) into inositol-1,4,5-triphosphate (IP3) and diacylglycerol (DAG). IP3 and DAG regulate numerous downstream pathways, eliciting diverse and profound cellular changes and physiological responses. In the six PLC subfamilies in higher eukaryotes, PLCß is intensively studied due to its prominent role in regulating crucial cellular events underlying many processes including cardiovascular and neuronal signaling, and associated pathological conditions. In addition to GαqGTP, Gßγ generated upon G protein heterotrimer dissociation also regulates PLCß activity. Here, we not only review how Gßγ directly activates PLCß, and also extensively modulates Gαq-mediated PLCß activity, but also provide a structure-function overview of PLC family members. Given that Gαq and PLCß are oncogenes, and Gßγ shows unique cell-tissue-organ specific expression profiles, Gγ subtype-dependent signaling efficacies, and distinct subcellular activities, this review proposes that Gßγ is a major regulator of Gαq-dependent and independent PLCß signaling.


Asunto(s)
Proteínas de Unión al GTP , Transducción de Señal , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Proteínas de Unión al GTP/metabolismo , Fosfolípidos
12.
Sci Rep ; 13(1): 4366, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36927770

RESUMEN

Oxaliplatin (OXA) is the first-line chemotherapy drug for metastatic colorectal cancer (mCRC), and the emergence of drug resistance is a major clinical challenge. Although there have been numerous studies on OXA resistance, but its underlying molecular mechanisms are still unclear. This study aims to identify key regulatory genes and pathways associated with OXA resistance. The Gene Expression Omnibus (GEO) GSE42387 dataset containing gene expression profiles of parental and OXA-resistant LoVo cells was applied to explore potential targets. GEO2R, STRING, CytoNCA (a plug-in of Cytoscape), and DAVID were used to analyze differentially expressed genes (DEGs), protein-protein interactions (PPIs), hub genes in PPIs, and gene ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. R2 online platform was used to run a survival analysis of validated hub genes enriched in KEGG pathways. The ENCORI database predicted microRNAs for candidate genes. A survival analysis of those genes was performed, and validated using the OncoLnc database. In addition, the 'clusterProfiler' package in R was used to perform gene set enrichment analysis (GSEA). We identified 395 DEGs, among which 155 were upregulated and 240 were downregulated. In total, 95 DEGs were screened as hub genes after constructing the PPI networks. Twelve GO terms and three KEGG pathways (steroid hormone biosynthesis, malaria, and pathways in cancer) were identified as being significant in the enrichment analysis of hub genes. Twenty-one hub genes enriched in KEGG pathways were defined as key genes. Among them AKT3, phospholipase C Beta 4 (PLCB4), and TGFB1 were identified as OXA-resistance genes through the survival analysis. High expressions of AKT3 and TGFB1 were each associated with a poor prognosis, and lower expression of PLCB4 was correlated with worse survival. Further, high levels of hsa-miR-1271-5p, which potentially targets PLCB4, were associated with poor overall survival in patients with CRC. Finally, we found that PLCB4 low expression was associated with MAPK signaling pathway and VEGF signaling pathway in CRC. Our results demonstrated that hsa-miR-1271-5p/PLCB4 in the pathway in cancer could be a new potential therapeutic target for mCRC with OXA resistance.


Asunto(s)
Neoplasias Colorrectales , MicroARNs , Humanos , Oxaliplatino/farmacología , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Redes Reguladoras de Genes , Biología Computacional/métodos
13.
J Biomed Sci ; 29(1): 105, 2022 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-36517806

RESUMEN

BACKGROUND: In endothelial cells, phospholipase C (PLC) ß1-activated Ca2+ is a crucial second messenger for the signaling pathways governing angiogenesis. PLCß1 is inactivated by complexing with an intracellular protein called translin-associated factor X (TRAX). This study demonstrates specific interactions between Globo H ceramide (GHCer) and TRAX, which highlight a new angiogenic control through PLCß1 activation. METHODS: Globo-series glycosphingolipids (GSLs), including GHCer and stage-specific embryonic antigen-3 ceramide (SSEA3Cer), were analyzed using enzyme-linked immunosorbent assay (ELISA) and Biacore for their binding with TRAX. Angiogenic activities of GSLs in human umbilical vein endothelial cells (HUVECs) were evaluated. Molecular dynamics (MD) simulation was used to study conformations of GSLs and their molecular interactions with TRAX. Fluorescence resonance energy transfer (FRET) analysis of HUVECs by confocal microscopy was used to validate the release of PLCß1 from TRAX. Furthermore, the in vivo angiogenic activity of extracellular vesicles (EVs) containing GHCer was confirmed using subcutaneous Matrigel plug assay in mice. RESULTS: The results of ELISA and Biacore analysis showed a stable complex between recombinant TRAX and synthetic GHCer with KD of 40.9 nM. In contrast, SSEA3Cer lacking a fucose residue of GHCer at the terminal showed ~ 1000-fold decrease in the binding affinity. These results were consistent with their angiogenic activities in HUVECs. The MD simulation indicated that TRAX interacted with the glycan moiety of GHCer at amino acid Q223, Q219, L142, S141, and E216. At equilibrium the stable complex maintained 4.6 ± 1.3 H-bonds. TRAX containing double mutations with Q223A and Q219A lost its ability to interact with GHCer in both MD simulation and Biacore assays. Removal of the terminal fucose from GHCer to become SSEA3Cer resulted in decreased H-bonding to 1.2 ± 1.0 by the MD simulation. Such specific H-bonding was due to the conformational alteration in the whole glycan which was affected by the presence or absence of the fucose moiety. In addition, ELISA, Biacore, and in-cell FRET assays confirmed the competition between GHCer and PLCß1 for binding to TRAX. Furthermore, the Matrigel plug assay showed robust vessel formation in the plug containing tumor-secreted EVs or synthetic GHCer, but not in the plug with SSEA3Cer. The FRET analysis also indicated the disruption of colocalization of TRAX and PLCß1 in cells by GHCer derived from EVs. CONCLUSIONS: Overall, the fucose residue in GHCer dictated the glycan conformation for its complexing with TRAX to release TRAX-sequestered PLCß1, leading to Ca2+ mobilization in endothelial cells and enhancing angiogenesis in tumor microenvironments.


Asunto(s)
Proteínas de Unión al ADN , Fucosa , Células Endoteliales de la Vena Umbilical Humana , Animales , Humanos , Ratones , Ceramidas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fucosa/genética , Fucosa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo
14.
Cells ; 11(22)2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36428956

RESUMEN

Reduction of insulin/insulin-like growth factor 1 (IGF1) signaling (IIS) promotes longevity across species. In the nematode Caenorhabditis elegans, ablation of germline stem cells (GSCs) and activity changes of the conserved signaling mediators unc-43/CaMKII (calcium/calmodulin-dependent kinase type II) and egl-8/PLCß (phospholipase Cß) also increase lifespan. Like IIS, these pathways depend on the conserved transcription factor daf-16/FOXO for lifespan extension, but how they functionally interact is unknown. Here, we show that altered unc-43/egl-8 activity further increases the lifespan of long-lived GSC-deficient worms, but not of worms that are long-lived due to a strong reduction-of-function mutation in the insulin/IGF1-like receptor daf-2. Additionally, we provide evidence for unc-43 and, to a lesser extent, egl-8 modulating the expression of certain collagen genes, which were reported to be dispensable for longevity of these particular daf-2 mutant worms, but not for other forms of longevity. Together, these results provide new insights into the conditions and potential mechanisms by which CaMKII- and PLCß-signals modulate C. elegans lifespan.


Asunto(s)
Proteínas de Caenorhabditis elegans , Insulinas , Animales , Caenorhabditis elegans/metabolismo , Longevidad/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Factores de Transcripción Forkhead/metabolismo , Células Germinativas/metabolismo , Mutación/genética , Insulinas/genética , Insulinas/metabolismo
15.
J Cell Sci ; 135(18)2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36125065

RESUMEN

Phospholipase Cß (PLCß) is the main effector of the Gq family of heterotrimeric G proteins that transduces signals from hormones and neurotransmitters into Ca2+ signals. While PLCß is critical for Ca2+ responses, recent studies have suggested that PLCß has additional roles independent of its lipase activity. These novel functions are carried out by a cytosolic population of PLCß that binds and inhibits the component 3 promoter of RNA-induced silencing complex (C3PO) to impact cytosolic RNA populations. Additionally, cytosolic PLCß binds to stress granule proteins, keeping them dispersed and thus inhibiting stress granule formation. Upon activation of the Gα subunit of Gq (Gαq), cytosolic PLCß relocalizes to the membrane, releasing C3PO and stress granule proteins, which in turn promotes activation of C3PO and RNA processing, as well as sequestration of specific transcripts into newly formed stress granules. As highlighted in this Cell Science at a Glance and the accompanying poster, the link between Gαq signaling, increased intracellular Ca2+ and changes in RNA processing impacts neuronal cell differentiation and may also affect neuronal development and dysfunction.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Proteínas de Unión al GTP Heterotriméricas , Calcio/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Hormonas , Lipasa/metabolismo , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , ARN , Complejo Silenciador Inducido por ARN/genética , Complejo Silenciador Inducido por ARN/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 42(10): 1229-1241, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35861069

RESUMEN

BACKGROUND: Regulation of vascular permeability is critical to maintaining tissue metabolic homeostasis. VEGF (vascular endothelial growth factor) is a key stimulus of vascular permeability in acute and chronic diseases including ischemia reperfusion injury, sepsis, and cancer. Identification of novel regulators of vascular permeability would allow for the development of effective targeted therapeutics for patients with unmet medical need. METHODS: In vitro and in vivo models of VEGFA-induced vascular permeability, pathological permeability, quantitation of intracellular calcium release and cell entry, and phosphatidylinositol 4,5-bisphosphate levels were evaluated with and without modulation of PLC (phospholipase C) ß2. RESULTS: Global knock-out of PLCß2 in mice resulted in blockade of VEGFA-induced vascular permeability in vivo and transendothelial permeability in primary lung endothelial cells. Further work in an immortalized human microvascular cell line modulated with stable knockdown of PLCß2 recapitulated the observations in the mouse model and primary cell assays. Additionally, loss of PLCß2 limited both intracellular release and extracellular entry of calcium following VEGF stimulation as well as reduced basal and VEGFA-stimulated levels of phosphatidylinositol 4,5-bisphosphate compared to control cells. Finally, loss of PLCß2 in both a hyperoxia-induced lung permeability model and a cardiac ischemia:reperfusion model resulted in improved animal outcomes when compared with wild-type controls. CONCLUSIONS: The results implicate PLCß2 as a key positive regulator of VEGF-induced vascular permeability through regulation of both calcium flux and phosphatidylinositol 4,5-bisphosphate levels at the cellular level. Targeting of PLCß2 in a therapeutic setting may provide a novel approach to regulating vascular permeability in patients.


Asunto(s)
Permeabilidad Capilar , Fosfatidilinositol 4,5-Difosfato , Fosfolipasa C beta , Mucosa Respiratoria , Factor A de Crecimiento Endotelial Vascular , Animales , Calcio/metabolismo , Permeabilidad Capilar/genética , Permeabilidad Capilar/fisiología , Células Endoteliales/metabolismo , Humanos , Pulmón/metabolismo , Ratones , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Fosfolipasa C beta/fisiología , Mucosa Respiratoria/metabolismo
17.
Mol Cell Biochem ; 477(8): 2133-2147, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35608718

RESUMEN

To explore the role of the miRNA-1297/phospholipase Cß1 (PLCß1) axis in intestinal barrier injury. Abnormally expressed miR-1297 and its target gene PLCß1 as well as their transcriptome sequencing were confirmed by bioinformatics analysis. Next, the intestinal barrier injury was induced by lipopolysaccharide (LPS) in the CCCHIE-2 cells. Subsequently, the impacts of miR-1297 and PLCß1 on the transcriptome were estimated. QRT-PCR and Western blotting were conducted to detect the relative mRNA and protein expressions, respectively. The cell viability and permeability were analyzed by MTT assay and fluorescent yellow detection. miR-1297 was significantly upregulated in patients with human immunodeficiency virus/acquired immunodeficiency syndrome and targeted PLCß1. Moreover, overexpressed PLCß1 was mainly enriched in the transforming growth factor-beta signaling pathway, while the knockdown of miR-1297 was focused on the arginine biosynthesis pathway. The overexpression of miR-1297 could reduce the PLCß1 expression and inhibit the viability of CCCHIE-2 cells injured by LPS, while the effect of the downregulation of miR-1297 was on the opposite. Western blotting and cell fluorescence localization experiments revealed that the inhibition of miR-1297 increased the expressions of PLCß1 and ZO-1. In addition, the upregulation of miR-1297 strengthened the permeability in cells injured by LPS, as did the knockdown of PLCß1. miR-1297 could restrain the repair of intestinal barrier injury via negatively regulating PLCß1 and its tight junction downstream protein ZO-1 in CCC-HIE-2 cells injured by LPS, which indicated that PLCß1 and miR-1297 might be important targets for the repair of intestinal barrier injury.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida , MicroARNs , Regulación hacia Abajo , Humanos , Lipopolisacáridos/farmacología , MicroARNs/metabolismo , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo
18.
Sci Rep ; 12(1): 8758, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35610292

RESUMEN

The Gαq/phospholipase Cß1 (PLCß1) signaling system mediates calcium responses from hormones and neurotransmitters. While PLCß1 functions on the plasma membrane, there is an atypical cytosolic population that binds Argonaute 2 (Ago2) and other proteins associated with stress granules preventing their aggregation. Activation of Gαq relocalizes cytosolic PLCß1 to the membrane, releasing bound proteins, promoting the formation of stress granules. Here, we have characterized Ago2 stress granules associated with Gαq activation in differentiated PC12 cells, which have a robust Gαq/PLCß1 signaling system. Characterization of Ago2-associated stress granules shows shifts in protein composition when cells are stimulated with a Gαq agonist, or subjected to heat shock or osmotic stress, consistent with the idea that different stresses result in unique stress granules. Purified Ago2 stress granules from control cells do not contain RNA, while those from heat shock contain many different mRNAs and miRs. Surprisingly, Ago2 particles from cells where Gαq was stimulated show only two transcripts, chromogranin B, which is involved in secretory function, and ATP synthase 5f1b, which is required for ATP synthesis. RT-PCR, western blotting and other studies support the idea that Gαq-activation protects these transcripts. Taken together, these studies show a novel pathway where Gαq/PLCß regulates the translation of specific proteins.


Asunto(s)
Proteínas de Unión al GTP , Transducción de Señal , Adenosina Trifosfato , Animales , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Proteínas de Unión al GTP/metabolismo , Células PC12 , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Ratas , Transducción de Señal/fisiología
19.
Cell Mol Life Sci ; 79(4): 195, 2022 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-35303162

RESUMEN

Glioblastoma represents the most lethal brain tumor in adults. Several studies have shown the key role of phospholipase C ß1 (PLCß1) in the regulation of many mechanisms within the central nervous system suggesting PLCß1 as a novel signature gene in the molecular classification of high-grade gliomas. This study aims to determine the pathological impact of PLCß1 in glioblastoma, confirming that PLCß1 gene expression correlates with glioma's grade, and it is lower in 50 glioblastoma samples compared to 20 healthy individuals. PLCß1 silencing in cell lines and primary astrocytes, leads to increased cell migration and invasion, with the increment of mesenchymal transcription factors and markers, as Slug and N-Cadherin and metalloproteinases. Cell proliferation, through increased Ki-67 expression, and the main survival pathways, as ß-catenin, ERK1/2 and Stat3 pathways, are also affected by PLCß1 silencing. These data suggest a potential role of PLCß1 in maintaining a normal or less aggressive glioma phenotype.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Proliferación Celular/genética , Glioblastoma/patología , Glioma/patología , Humanos , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo
20.
Dis Model Mech ; 15(4)2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35284927

RESUMEN

Auriculocondylar syndrome 2 (ARCND2) is a rare autosomal dominant craniofacial malformation syndrome linked to multiple genetic variants in the coding sequence of phospholipase C ß4 (PLCB4). PLCB4 is a direct signaling effector of the endothelin receptor type A (EDNRA)-Gq/11 pathway, which establishes the identity of neural crest cells (NCCs) that form lower jaw and middle ear structures. However, the functional consequences of PLCB4 variants on EDNRA signaling is not known. Here, we show, using multiple signaling reporter assays, that known PLCB4 variants resulting from missense mutations exert a dominant-negative interference over EDNRA signaling. In addition, using CRISPR/Cas9, we find that F0 mouse embryos modeling one PLCB4 variant have facial defects recapitulating those observed in hypomorphic Ednra mouse models, including a bone that we identify as an atavistic change in the posterior palate/oral cavity. Remarkably, we have identified a similar osseous phenotype in a child with ARCND2. Our results identify the disease mechanism of ARCND2, demonstrate that the PLCB4 variants cause craniofacial differences and illustrate how minor changes in signaling within NCCs may have driven evolutionary changes in jaw structure and function. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Enfermedades del Oído , Animales , Oído/anomalías , Enfermedades del Oído/genética , Humanos , Ratones , Cresta Neural , Fenotipo , Fosfolipasa C beta/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA