Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 240
Filtrar
1.
Sci Rep ; 14(1): 15142, 2024 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956267

RESUMEN

Multiple myeloma (MM) is an incurable hematological malignancy with poor survival. Accumulating evidence reveals that lactylation modification plays a vital role in tumorigenesis. However, research on lactylation-related genes (LRGs) in predicting the prognosis of MM remains limited. Differentially expressed LRGs (DELRGs) between MM and normal samples were investigated from the Gene Expression Omnibus database. Univariate Cox regression and LASSO Cox regression analysis were applied to construct gene signature associated with overall survival. The signature was validated in two external datasets. A nomogram was further constructed and evaluated. Additionally, Enrichment analysis, immune analysis, and drug chemosensitivity analysis between the two groups were investigated. qPCR and immunofluorescence staining were performed to validate the expression and localization of PFN1. CCK-8 and flow cytometry were performed to validate biological function. A total of 9 LRGs (TRIM28, PPIA, SOD1, RRP1B, IARS2, RB1, PFN1, PRCC, and FABP5) were selected to establish the prognostic signature. Kaplan-Meier survival curves showed that high-risk group patients had a remarkably worse prognosis in the training and validation cohorts. A nomogram was constructed based on LRGs signature and clinical characteristics, and showed excellent predictive power by calibration curve and C-index. Moreover, biological pathways, immunologic status, as well as sensitivity to chemotherapy drugs were different between high- and low-risk groups. Additionally, the hub gene PFN1 is highly expressed in MM, knocking down PFN1 induces cell cycle arrest, suppresses cell proliferation and promotes cell apoptosis. In conclusion, our study revealed that LRGs signature is a promising biomarker for MM that can effectively early distinguish high-risk patients and predict prognosis.


Asunto(s)
Biomarcadores de Tumor , Regulación Neoplásica de la Expresión Génica , Mieloma Múltiple , Profilinas , Humanos , Mieloma Múltiple/genética , Mieloma Múltiple/mortalidad , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/patología , Pronóstico , Profilinas/genética , Profilinas/metabolismo , Biomarcadores de Tumor/genética , Masculino , Femenino , Nomogramas , Proliferación Celular/genética , Perfilación de la Expresión Génica , Estimación de Kaplan-Meier , Línea Celular Tumoral , Transcriptoma , Apoptosis/genética , Persona de Mediana Edad
2.
Biol Chem ; 405(6): 367-381, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38662449

RESUMEN

Structural and allergenic characterization of mite profilins has not been previously pursued to a similar extent as plant profilins. Here, we describe structures of profilins originating from Tyrophagus putrescentiae (registered allergen Tyr p 36.0101) and Dermatophagoides pteronyssinus (here termed Der p profilin), which are the first structures of profilins from Arachnida. Additionally, the thermal stabilities of mite and plant profilins are compared, suggesting that the high number of cysteine residues in mite profilins may play a role in their increased stability. We also examine the cross-reactivity of plant and mite profilins as well as investigate the relevance of these profilins in mite inhalant allergy. Despite their high structural similarity to other profilins, mite profilins have low sequence identity with plant and human profilins. Subsequently, these mite profilins most likely do not display cross-reactivity with plant profilins. At the same time the profilins have highly conserved poly(l-proline) and actin binding sites.


Asunto(s)
Reacciones Cruzadas , Profilinas , Animales , Reacciones Cruzadas/inmunología , Profilinas/inmunología , Profilinas/química , Profilinas/metabolismo , Humanos , Ácaros/inmunología , Ácaros/química , Secuencia de Aminoácidos , Hipersensibilidad/inmunología , Plantas/inmunología , Plantas/química , Plantas/metabolismo , Modelos Moleculares , Alérgenos/inmunología , Alérgenos/química
3.
Chembiochem ; 25(9): e202400007, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38457348

RESUMEN

The actin cytoskeleton is a biosensor of cellular stress and a potential prognosticator of human disease. In particular, aberrant cytoskeletal structures such as stress granules formed in response to energetic and oxidative stress are closely linked to ageing, cancer, cardiovascular disease, and viral infection. Whether these cytoskeletal phenomena can be harnessed for the development of biosensors for cytoskeletal dysfunction and, by extension, disease progression, remains an open question. In this work, we describe the design and development of an optogenetic iteration of profilin, an actin monomer binding protein with critical functions in cytoskeletal dynamics. We demonstrate that this optically activated profilin ('OptoProfilin') can act as an optically triggered biosensor of applied cellular stress in select immortalized cell lines. Notably, OptoProfilin is a single component biosensor, likely increasing its utility for experimentalists. While a large body of preexisting work closely links profilin activity with cellular stress and neurodegenerative disease, this, to our knowledge, is the first example of profilin as an optogenetic biosensor of stress-induced changes in the cytoskeleton.


Asunto(s)
Técnicas Biosensibles , Profilinas , Profilinas/metabolismo , Humanos , Optogenética/métodos , Estrés Fisiológico
4.
J Biol Chem ; 300(1): 105583, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38141770

RESUMEN

Membrane polyphosphoinositides (PPIs) are lipid-signaling molecules that undergo metabolic turnover and influence a diverse range of cellular functions. PPIs regulate the activity and/or spatial localization of a number of actin-binding proteins (ABPs) through direct interactions; however, it is much less clear whether ABPs could also be an integral part in regulating PPI signaling. In this study, we show that ABP profilin1 (Pfn1) is an important molecular determinant of the cellular content of PI(4,5)P2 (the most abundant PPI in cells). In growth factor (EGF) stimulation setting, Pfn1 depletion does not impact PI(4,5)P2 hydrolysis but enhances plasma membrane (PM) enrichment of PPIs that are produced downstream of activated PI3-kinase, including PI(3,4,5)P3 and PI(3,4)P2, the latter consistent with increased PM recruitment of SH2-containing inositol 5' phosphatase (SHIP2) (a key enzyme for PI(3,4)P2 biosynthesis). Although Pfn1 binds to PPIs in vitro, our data suggest that Pfn1's affinity to PPIs and PM presence in actual cells, if at all, is negligible, suggesting that Pfn1 is unlikely to directly compete with SHIP2 for binding to PM PPIs. Additionally, we provide evidence for Pfn1's interaction with SHIP2 in cells and modulation of this interaction upon EGF stimulation, raising an alternative possibility of Pfn1 binding as a potential restrictive mechanism for PM recruitment of SHIP2. In conclusion, our findings challenge the dogma of Pfn1's binding to PM by PPI interaction, uncover a previously unrecognized role of Pfn1 in PI(4,5)P2 homeostasis and provide a new mechanistic avenue of how an ABP could potentially impact PI3K signaling byproducts in cells through lipid phosphatase control.


Asunto(s)
Fosfatidilinositoles , Profilinas , Factor de Crecimiento Epidérmico/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/metabolismo , Fosfatidilinositoles/metabolismo , Humanos , Células HEK293 , Profilinas/metabolismo
5.
Anal Chem ; 95(41): 15141-15145, 2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37787459

RESUMEN

Profilin 1 (PFN1) is a cytoskeleton protein that modulates actin dynamics through binding to monomeric actin and polyproline-containing proteins. Mutations in PFN1 have been linked to the pathogenesis of familial amyotrophic lateral sclerosis (ALS). Here, we employed an unbiased proximity labeling strategy in combination with proteomic analysis for proteome-wide profiling of proteins that differentially interact with mutant and wild-type (WT) PFN1 proteins in human cells. We uncovered 11 mRNA splicing proteins that are preferentially enriched in the proximity proteomes of the two ALS-linked PFN1 variants, C71G and M114T, over that of wild-type PFN1. We validated the preferential interactions of the ALS-linked PFN1 variants with two mRNA splicing factors, hnRNPC and U2AF2, by immunoprecipitation, followed with immunoblotting. We also found that the two ALS-linked PFN1 variants promoted the exonization of Alu elements in the mRNAs of MTO1, TCFL5, WRN and POLE genes in human cells. Together, we showed that the two ALS-linked PFN1 variants interacted preferentially with mRNA splicing proteins, which elicited aberrant exonization of the Alu elements in mRNAs. Thus, our work provided pivotal insights into the perturbations of ALS-linked PFN1 variants in RNA biology and their potential contributions to ALS pathology.


Asunto(s)
Esclerosis Amiotrófica Lateral , Humanos , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Profilinas/genética , Profilinas/metabolismo , Actinas/metabolismo , Proteómica , Mutación , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética
6.
Mol Cell Probes ; 72: 101937, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37820747

RESUMEN

Doxorubicin (DOX) often causes acute or chronic cardiotoxicity during its application. LncRNA RMRP has been reported to be associated with several biological processes, such as cartilage-hair hypoplasia, but the relationship between RMRP and DOX-induced cardiotoxicity and chronic heart failure remains obscure. To test this hypothesis, GSE124401 and GSE149870 were processed for bioinformatics, and differentially expressed RMRP was then verified in the peripheral blood of 21 patients with heart failure compared with 7 controls. For in vitro validation, we used AC16 and HEK-293T cells. qPCR was used to detect the mRNA expression levels. The degree of apoptosis was detected by Western blot and TUNEL staining. Furthermore, the interaction between RMRP and PFN1 mRNA was verified by dual-luciferase reporter assays. In bioinformatics, RMRP showed significant downregulation, which was verified in clinical samples (p < 0.001) and DOX-treated AC16 models (p < 0.0001). Next, overexpression of RMRP could significantly alleviate DOX-induced apoptosis, and a potential downstream molecule of RMRP, PFN1, was also negatively associated with this change. RESCUE experiments further confirmed that PFN1 could be regulated by RMRP at both the RNA and protein levels, serving as a downstream mediator of RMRP's cardioprotective effects. This interaction was then confirmed to be a direct combination (p < 0.0001). Finally, we found that overexpression of RMRP could inhibit the expression of p53 and its phosphorylation level by suppressing PFN1. In summary, RMRP could exert cardioprotective effects via the PFN1/p53 axis, holding great promise for serving as a therapeutic target and potential biomarker.


Asunto(s)
Insuficiencia Cardíaca , MicroARNs , ARN Largo no Codificante , Humanos , MicroARNs/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteína p53 Supresora de Tumor/genética , Cardiotoxicidad/metabolismo , Doxorrubicina/farmacología , Apoptosis/genética , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/genética , ARN Mensajero , Profilinas/metabolismo , Profilinas/farmacología
7.
J Biol Chem ; 299(8): 105044, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37451478

RESUMEN

Overexpression of actin-binding protein profilin-1 (Pfn1) correlates with advanced disease features and adverse clinical outcome of patients with clear cell renal carcinoma, the most prevalent form of renal cancer. We previously reported that Pfn1 is predominantly overexpressed in tumor-associated vascular endothelial cells in human clear cell renal carcinoma. In this study, we combined in vivo strategies involving endothelial cell-specific depletion and overexpression of Pfn1 to demonstrate a role of vascular endothelial Pfn1 in promoting tumorigenicity and enabling progressive growth and metastasis of renal carcinoma cells in a syngeneic orthotopic mouse model of kidney cancer. We established an important role of endothelial Pfn1 in tumor angiogenesis and further identified endothelial Pfn1-dependent regulation of several pro- (VEGF, SERPINE1, CCL2) and anti-angiogenic factors (platelet factor 4) in vivo. Endothelial Pfn1 overexpression increases tumor infiltration by macrophages and concomitantly diminishes tumor infiltration by T cells including CD8+ T cells in vivo, correlating with the pattern of endothelial Pfn1-dependent changes in tumor abundance of several prominent immunomodulatory cytokines. These data were also corroborated by multiplexed quantitative immunohistochemistry and immune deconvolution analyses of RNA-seq data of clinical samples. Guided by Upstream Regulator Analysis of tumor transcriptome data, we further established endothelial Pfn1-induced Hif1α elevation and suppression of STAT1 activation. In conclusion, this study demonstrates for the first time a direct causal relationship between vascular endothelial Pfn1 dysregulation, immunosuppressive tumor microenvironment, and disease progression with mechanistic insights in kidney cancer. Our study also provides a conceptual basis for targeting Pfn1 for therapeutic benefit in kidney cancer.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Profilinas , Microambiente Tumoral , Animales , Humanos , Ratones , Carcinoma de Células Renales/genética , Células Endoteliales/metabolismo , Neoplasias Renales/genética , Profilinas/genética , Profilinas/metabolismo , Progresión de la Enfermedad
8.
Am J Pathol ; 193(8): 1059-1071, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37164274

RESUMEN

Unexplained recurrent spontaneous abortion (URSA) has been associated with the dysfunction of trophoblasts and decidual macrophages. Current evidence suggests that profilin1 (PFN1) plays an important role in many biological processes. However, little is known about whether PFN1 is related to URSA. Herein, the location of PFN1 was detected by immunohistochemistry, and the level of PFN1 was detected by quantitative real-time PCR, Western blot analysis, and immunohistochemistry. The proliferation of trophoblasts was detected by CCK8 and 5-ethynyl-2'-deoxyuridine assays, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assays were used to detect apoptosis of trophoblasts. The migration and invasion ability of trophoblasts was assessed by using the wound-healing test and transwell test. Polarization of macrophages was detected in macrophages cultured in trophoblast conditioned medium. PFN1 expression was observed in cytotrophoblasts, syncytiotrophoblasts, and extravillous trophoblasts and was decreased in the villous tissue of patients with URSA. The migration and invasion ability and cell viability of trophoblastic cell lines that underwent PFN1 knockdown significantly decreased, and apoptosis increased. Opposite findings were observed after the overexpression of PFN1 in trophoblastic cells. In addition, PFN1 could regulate trophoblast function through phosphatidylinositol 3-kinase/AKT signal transduction rather than mitogen-activated protein kinase signaling pathways. Finally, knockdown of PFN1 in trophoblasts promoted tumor necrosis factor-α secretion to induce macrophage polarization to M1 phenotype, mediated by the NF-κB signaling pathway. These findings indicate that PFN1 has a broad therapeutic potential for patients with URSA.


Asunto(s)
Aborto Espontáneo , Trofoblastos , Embarazo , Humanos , Femenino , Trofoblastos/metabolismo , Transducción de Señal/fisiología , FN-kappa B/metabolismo , Sistema de Señalización de MAP Quinasas , Aborto Espontáneo/metabolismo , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Profilinas/genética , Profilinas/metabolismo
9.
Commun Biol ; 6(1): 9, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36599901

RESUMEN

Profilin 1-encoded by PFN1-is a small actin-binding protein with a tumour suppressive role in various adenocarcinomas and pagetic osteosarcomas. However, its contribution to tumour development is not fully understood. Using fix and live cell imaging, we report that Profilin 1 inactivation results in multiple mitotic defects, manifested prominently by anaphase bridges, multipolar spindles, misaligned and lagging chromosomes, and cytokinesis failures. Accordingly, next-generation sequencing technologies highlighted that Profilin 1 knock-out cells display extensive copy-number alterations, which are associated with complex genome rearrangements and chromothripsis events in primary pagetic osteosarcomas with Profilin 1 inactivation. Mechanistically, we show that Profilin 1 is recruited to the spindle midzone at anaphase, and its deficiency reduces the supply of actin filaments to the cleavage furrow during cytokinesis. The mitotic defects are also observed in mouse embryonic fibroblasts and mesenchymal cells deriving from a newly generated knock-in mouse model harbouring a Pfn1 loss-of-function mutation. Furthermore, nuclear atypia is also detected in histological sections of mutant femurs. Thus, our results indicate that Profilin 1 has a role in regulating cell division, and its inactivation triggers mitotic defects, one of the major mechanisms through which tumour cells acquire chromosomal instability.


Asunto(s)
Fibroblastos , Inestabilidad Genómica , Profilinas , Animales , Humanos , Ratones , Anafase/genética , Citocinesis/genética , Inestabilidad Genómica/genética , Mitosis/genética , Profilinas/genética , Profilinas/metabolismo , Osteosarcoma/genética , Osteosarcoma/metabolismo
10.
Bioessays ; 45(2): e2200119, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36461738

RESUMEN

The release of AlphaFold2 (AF2), a deep-learning-aided, open-source protein structure prediction program, from DeepMind, opened a new era of molecular biology. The astonishing improvement in the accuracy of the structure predictions provides the opportunity to characterize protein systems from uncultured Asgard archaea, key organisms in evolutionary biology. Despite the accumulation in metagenomics-derived Asgard archaea eukaryotic-like protein sequences, limited structural and biochemical information have restricted the insight in their potential functions. In this review, we focus on profilin, an actin-dynamics regulating protein, which in eukaryotes, modulates actin polymerization through (1) direct actin interaction, (2) polyproline binding, and (3) phospholipid binding. We assess AF2-predicted profilin structures in their potential abilities to participate in these activities. We demonstrate that AF2 is a powerful new tool for understanding the emergence of biological functional traits in evolution.


Asunto(s)
Archaea , Profilinas , Archaea/metabolismo , Profilinas/genética , Profilinas/metabolismo , Actinas , Filogenia , Furilfuramida/metabolismo , Eucariontes/metabolismo
11.
Nat Commun ; 13(1): 6531, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36319634

RESUMEN

DNA replication forks are tightly controlled by a large protein network consisting of well-known core regulators and many accessory factors which remain functionally undefined. In this study, we report previously unknown nuclear functions of the actin-binding factor profilin-1 (PFN1) in DNA replication, which occur in a context-dependent fashion and require its binding to poly-L-proline (PLP)-containing proteins instead of actin. In unperturbed cells, PFN1 increases DNA replication initiation and accelerates fork progression by binding and stimulating the PLP-containing nucleosome remodeler SNF2H. Under replication stress, PFN1/SNF2H increases fork stalling and functionally collaborates with fork reversal enzymes to enable the over-resection of unprotected forks. In addition, PFN1 binds and functionally attenuates the PLP-containing fork protector BODL1 to increase the resection of a subset of stressed forks. Accordingly, raising nuclear PFN1 level decreases genome stability and cell survival during replication stress. Thus, PFN1 is a multi-functional regulator of DNA replication with exploitable anticancer potential.


Asunto(s)
Actinas , Profilinas , Humanos , Actinas/metabolismo , ADN Helicasas/metabolismo , Replicación del ADN , Inestabilidad Genómica , Profilinas/metabolismo , Adenosina Trifosfatasas/metabolismo , Proteínas Cromosómicas no Histona/metabolismo
12.
Oxid Med Cell Longev ; 2022: 2198923, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36160709

RESUMEN

Preeclampsia is regarded as an evolution-related disease that has only been observed in humans and our closest relatives, and the important factor contributing to its pathogenesis is endothelial dysregulation secondary to a stressed placenta. Hypoxia-inducible factor 1 subunit alpha (HIF1α), a highly conserved molecule in virtually all mammals, is regarded as a crucial regulator of the hypoxia adaptation and evolution. Persistent high expression of HIF1α in the placenta is one of the pathogenic mechanisms of preeclampsia. Therefore, human-specific molecules should link increased HIF1α to preeclampsia. We reported that urothelial cancer associated 1 (UCA1) is a potential mediator because it is a human-specific long noncoding RNA (lncRNA) that is upregulated in placental tissues and maternal serum from women with preeclampsia and is regulated by HIF1α. The cellular HIF1α-UCA1 pathway promoted the adaptation of trophoblasts to hypoxia by inducing vascular endothelial growth factor (VEGF) secretion and changes in the levels of key enzymes in glycolysis. On the other hand, circulating exosomal UCA1 secreted from stressed trophoblasts induced vascular endothelial dysfunction, especially excess ROS production, as measured by exosome extraction and a coculture system. At the molecular level, UCA1 physically bound to ubiquitin-specific peptidase 14 (USP14), which is a deubiquitinating enzyme, and UCA1 functioned as a scaffold to recruit USP14 to profilin 1 (PFN1), an actin-binding protein contributing to endothelial abnormalities and vascular diseases. This ternary complex inhibited the ubiquitination-dependent degradation of PFN1 and prolonged its half-life, further activating the RhoA/Rho-kinase (ROCK) pathway to induce ROS production in endothelial cells. Taken together, these observations suggest a role for the evolution-related UCA1 in the HIF1α-induced adaptive pathogenic mechanism of preeclampsia, promoting the survival of hypoxic trophoblasts and injuring maternal endothelial cells.


Asunto(s)
Preeclampsia , ARN Largo no Codificante , Neoplasias de la Vejiga Urinaria , Enzimas Desubicuitinizantes/metabolismo , Células Endoteliales/metabolismo , Femenino , Humanos , Hipoxia/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Placenta/metabolismo , Preeclampsia/genética , Preeclampsia/metabolismo , Embarazo , Profilinas/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Trofoblastos/metabolismo , Ubiquitina Tiolesterasa , Proteasas Ubiquitina-Específicas/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
13.
Development ; 149(16)2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35950913

RESUMEN

Profilin 4 (Pfn4) is expressed during spermiogenesis and localizes to the acrosome-acroplaxome-manchette complex. Here, we generated PFN4-deficient mice, with sperm displaying severe impairment in manchette formation. Interestingly, HOOK1 staining suggests that the perinuclear ring is established; however, ARL3 staining is disrupted, suggesting that lack of PFN4 does not interfere with the formation of the perinuclear ring and initial localization of HOOK1, but impedes microtubular organization of the manchette. Furthermore, amorphous head shape and flagellar defects were detected, resulting in reduced sperm motility. Disrupted cis- and trans-Golgi networks and aberrant production of proacrosomal vesicles caused impaired acrosome biogenesis. Proteomic analysis showed that the proteins ARF3, SPECC1L and FKBP1, which are involved in Golgi membrane trafficking and PI3K/AKT pathway, are more abundant in Pfn4-/- testes. Levels of PI3K, AKT and mTOR were elevated, whereas AMPK level was reduced, consistent with inhibition of autophagy. This seems to result in blockage of autophagic flux, which could explain the failure in acrosome formation. In vitro fertilization demonstrated that PFN4-deficient sperm is capable of fertilizing zona-free oocytes, suggesting a potential treatment for PFN4-related human infertility.


Asunto(s)
Acrosoma , Profilinas , Espermátides , Espermatogénesis , Acrosoma/metabolismo , Animales , Masculino , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Profilinas/genética , Profilinas/metabolismo , Proteómica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Semen , Motilidad Espermática , Espermátides/metabolismo , Espermatogénesis/genética , Espermatozoides
14.
Environ Toxicol ; 37(10): 2445-2459, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35776891

RESUMEN

Organophosphate flame retardants (OPFRs) have been widely used due to their unique properties. The OPFRs are mainly metabolized in the liver. However, whether the plasma level of OPFRs was involved in the progression of liver cancer remains unclear. Triphenyl phosphate (TPP) is one of the OPFRs that are mostly detected in environment. In this study, we performed CCK8, ATP, and EdU analyses to evaluate the effect of TPP at the concentrations at 0.025-12.8 µM on the proliferation, invasion, and migration of Hep3B, a hepatocellular carcinoma (HCC) cell line. Tumor-bearing mouse model was used for in vivo validation. The results showed that low concentrations of TPP at (0.025-0.1 µM), which are obtained in the plasma of patients with cancers, remarkably promoted cell invasion and migration of Hep3B cells. Animal experiments confirmed that TPP treatment significantly enhanced tumor growth in the xenograft HCC model. To explore the possible molecular mechanisms that might mediate the actions of TPP on Hep3B cells, we profiled gene expression in groups treated with or without TPP at the concentrations of 0.05 and 0.1 µM using transcriptional sequencing. Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment, and Protein-protein interaction (PPI) analyses demonstrated that pathways affected by differentially expressed genes (DEGs) were mainly in nuclear-transcribed mRNA catabolic processes, cytosolic ribosome, and ATPase activity. A 0.05 and 0.1 µM TPP led to up-regulation of a series of genes including EREG, DNPH1, SAMD9, DUSP5, PFN1, CKB, MICAL2, SCUBE3, and CXCL8, but suppressed the expression of MCC. These genes have been shown to be associated with proliferation and movement of cells. Taken together, our findings suggest that low concentration of TPP could fuel the proliferation, invasion, and migration of HCC cells. Thus, TPP is a risk factor in the progression of HCC in human beings.


Asunto(s)
Carcinoma Hepatocelular , Retardadores de Llama , Neoplasias Hepáticas , Animales , Proteínas de Unión al Calcio/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular , Proliferación Celular/genética , Retardadores de Llama/toxicidad , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones , Organofosfatos/farmacología , Profilinas/genética , Profilinas/metabolismo
15.
F S Sci ; 3(1): 35-48, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35559994

RESUMEN

OBJECTIVE: To investigate the regulatory mechanisms and signaling molecules underlying hatching in mouse embryos. DESIGN: Experimental laboratory study using a mouse embryo model. SETTING: University-based basic scientific research laboratory. ANIMALS: A total of 40 B6C3F1 × B6D2F1 mouse embryos were used in this study. INTERVENTION(S): Frozen/thawed mouse embryos, at the 8-cell stage, were cultured in vitro for 2 days. The resulting hatching and prehatching blastocysts were then used for complementary deoxyribonucleic acid (cDNA) library preparation and ribonucleic acid (RNA) sequencing analysis (n = 8 for each group). Differentially expressed genes were then used for downstream functional analysis. In addition, a list of genes related to developmental progression in humans was used to identify genes that were potentially related to the hatching of human embryos. MAIN OUTCOME MEASURE(S): Differentially expressed genes, enriched Gene Ontology terms and canonical pathways, clustered gene networks, activated upstream regulators, and common genes between a gene list of hatching-related genes in mice and a gene list associated with developmental progression in humans. RESULT(S): A total 275 differentially expressed genes were identified between hatching and prehatching blastocysts: 230 up-regulated and 45 down-regulated genes. Functional enrichment analysis suggested that blastocyst hatching in vitro is an adenosine triphosphate (ATP)-dependent process that involves protein biosynthesis and organization of the cytoskeleton. Furthermore, by regulating cell motility, the RhoA signaling pathway (including Arpc2, Cfl1, Gsn, Pfn1, Tpi1, Grb2, Tmsb10, Enah, and Rnd3 genes) may be a crucial signaling pathway during hatching. We also identified a cluster of genes (Krt8, Krt7, Cldn4, and Aqp3) that exerted functional roles in cell-cell junctions and water homeostasis during hatching. Moreover, some growth factors (angiotensinogen and fibroblast growth factor 2) and endocrine factors (estrogen receptor and prolactin) were predicted to be involved in the regulation of embryo hatching. In addition, we identified 81 potential genes that are potentially involved in the hatching process in human embryos. CONCLUSION(S): Our analysis identified potential genes and molecular regulatory pathways involved in the blastocyst hatching process in mice; we also identified genes that may potentially regulate hatching in human embryos. Our findings enhance our knowledge of embryo development and provide useful information for further exploring the mechanisms underlying embryo hatching.


Asunto(s)
Blastocisto , ARN , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Blastocisto/metabolismo , Embrión de Mamíferos , Desarrollo Embrionario/genética , Femenino , Humanos , Parto , Embarazo , Profilinas/metabolismo , ARN/metabolismo
16.
Thorac Cancer ; 13(11): 1597-1610, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35445786

RESUMEN

BACKGROUND: Circular RNAs (circRNAs) participate in the occurrence and progression of many cancers. CircRNA ataxin 7 (circATXN7) (circBase ID: hsa_circ_0066436) plays a promoting influence on gastric cancer progression. However, the biological role of circATXN7 in non-small cell lung cancer (NSCLC) is indistinct. METHODS: Levels of circATXN7, microRNA (miR)-7-5p, and profilin 2 (PFN2) mRNA were detected using quantitative real-time polymerase chain reaction (RT-qPCR). Proliferation, apoptosis, metastasis, and invasion were analyzed using cell counting kit-8 (CCK-8), colony formation, 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, and transwell assays. Protein levels were analyzed using western blotting (WB) and immunohistochemistry (IHC). The relationship between circATXN7 or PFN2 and miR-7-5p was analyzed by dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. The biological function of circATXN7 was verified by xenograft assay. RESULTS: CircATXN7 and PFN2 were highly expressed in NSCLC, whereas miR-7-5p expression had the opposite trend. CircATXN7 overexpression constrained apoptosis and promoted proliferation, metastasis, invasion, and epithelial-mesenchymal transition of NSCLC cells, but circATXN7 silencing played the opposing influence and repressed xenograft tumor growth in vivo. CircATXN7 served as a miR-7-5p sponge, and circATXN7 regulated malignant behaviors of NSCLC cells through sponging miR-7-5p. PFN2 acted as a miR-7-5p target. PFN2 silencing overturned the promoting effect of miR-7-5p inhibitor on NSCLC cell malignancy, while PFN2 overexpression reversed the inhibitory impact of miR-7-5p mimic on NSCLC cell malignancy. CONCLUSION: CircATXN7 accelerated the malignancy of NSCLC cells through adsorbing miR-7-5p and upregulating PFN2, offering evidence to support circATXN7 as a target for NSCLC treatment.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , MicroARNs , Ataxina-7/genética , Ataxina-7/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Proliferación Celular/genética , Regulación hacia Abajo , Humanos , Neoplasias Pulmonares/patología , MicroARNs/genética , MicroARNs/metabolismo , Profilinas/genética , Profilinas/metabolismo , ARN Circular/genética
17.
Sci Rep ; 12(1): 5432, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35361841

RESUMEN

Vegetables represent a major source of Ni exposure. Environmental contamination and cultural practices can increase Ni amount in tomato posing significant risk for human health. This work assesses the tomato (Solanum lycopersicum L.) response to Ni on the agronomic yield of fruits and the related production of allergens. Two cultivars were grown in pots amended with Ni 0, 30, 60, 120, and 300 mg kg-1, respectively. XRF and ICP-MS analyses highlighted the direct increase of fruit Ni content compared to soil Ni, maintaining a stable biomass. Leaf water content increased at Ni 300 mg kg-1. Total protein content and individual allergenic components were investigated using biochemical (RP-HPLC and N-terminal amino acid sequencing) and immunological (inhibition tests of IgE binding by SPHIAa assay on the FABER testing system) methodologies. Ni affected the fruit tissue concentration of pathogenesis-related proteins and relevant allergens (LTP, profilin, Bet v 1-like protein and TLP). This study elucidates for the first time that tomato reacts to exogenous Ni, uptaking the metal while changing its allergenic profiles, with potential double increasing of exposure risks for consumers. This evidence highlighted the importance of adequate choice of low-Ni tomato cultivars and practices to reduce Ni uptake by potentially contaminated matrices.


Asunto(s)
Alérgenos , Níquel , Solanum lycopersicum , Frutas , Solanum lycopersicum/efectos de los fármacos , Solanum lycopersicum/metabolismo , Níquel/toxicidad , Profilinas/metabolismo
18.
Nan Fang Yi Ke Da Xue Xue Bao ; 42(2): 215-222, 2022 Feb 20.
Artículo en Chino | MEDLINE | ID: mdl-35365445

RESUMEN

OBJECTIVE: To investigate the expression of profilin 2 (PFN2) in gastric cancer and assess its potential value as a novel prognostic indicator and a therapeutic target. METHODS: We collected gastric cancer and paired adjacent tissues from 100 patients for immunohistochemical detection of PFN2 expression. According to the expression level of PFN2, the patients were divided into two groups with high (46 cases) and low (48 cases) PNF2 expression in cancer tissues, and also into two groups with high (26 cases) and low (49 cases) PNF2 expression in adjacent tissues. Chi-square test, Spearman correlation and KaplanMeier survival analysis were used to analyze the relationship between PFN2 protein expression level and the patients' clinical parameters. We also tested the effects of PFN2 knockdown and overexpression on the proliferation and migration of MKN-45 cells using Transwell assay and CCK-8 assay. RESULTS: The expression of PFN2 protein was significantly higher in gastric cancer tissues than in adjacent tissues (P < 0.01). PFN2 expression was positively correlated with M-stage of gastric cancer and VEGFR expression in the tumor tissues (P < 0.01). A high expression of PFN2 protein was significantly correlated with a poor prognosis of gastric cancer patients (P < 0.01), and was an independent predictor of the prognosis of gastric cancer. In MKN-45 cells, the cells overexpressing PFN2 showed significantly stronger proliferation and migration abilities than those with PFN2 knockdown (P < 0.001). CONCLUSION: PFN2 protein is highly expressed in gastric cancer tissues to promote the proliferation and migration of the tumor cells. PFN2 may serve as a potential diagnostic marker, a prognostic indicator and a therapeutic target for gastric cancer.


Asunto(s)
Profilinas , Neoplasias Gástricas , Proliferación Celular , Humanos , Profilinas/metabolismo , Pronóstico , Neoplasias Gástricas/patología , Análisis de Supervivencia
19.
Acta Biochim Pol ; 69(1): 147-153, 2022 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-35232010

RESUMEN

The microRNA-151 (miR-151) has been reported to be involved in the growth, development, and tumorigenesis of different types of human cancers. This study was designed to unravel the role and therapeutic potential of miR-151 in glioma. The results showed glioma was found to be associated with significant (P<0.05) downregulation of miR-151. Low expression of miR-151 was also associated with poor survival of the glioma patients. Overexpression of miR-151 resulted in a significant (P<0.05) decline of glioma cell proliferation and colony formation. The sensitivity of the glioma cells to adriamycin also increased significantly (P<0.05) upon miR-151 overexpression. Additionally, overexpression of miR-151 also suppressed the migration and invasion of the human glioma cells. This was also associated with alteration in the expression of epithelial mesenchymal transition proteins. The expression of E-cadherin was increased while as that of N-cadherin, vimentin, and Snail was considerably decreased upon miR-151 overexpression. Bioinformatic analysis and ducal luciferase assay showed miR-151 targets profilin 2 (PFN2) in human glioma cells. The expression of PFN2 was found to be significantly (P<0.05) upregulated in human glioma tissues cells and cell lines. Nonetheless, the PFN2 expression was considerably suppressed upon miR-151 overexpression. Knockdown of PFN2 resulted in decrease of glioma cells proliferation. In contrary, overexpression of PFN2 could avoid the tumor-suppressive effects of miR-151. Taken together, present study points towards the tumor-suppressive effects of miR-151 and prospective therapeutic implications in human glioma.


Asunto(s)
Transición Epitelial-Mesenquimal , Glioma/metabolismo , MicroARNs/metabolismo , Profilinas/metabolismo , Adulto , Cadherinas/metabolismo , Carcinogénesis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Doxorrubicina/farmacología , Femenino , Humanos , Masculino , Regulación hacia Arriba
20.
J Invest Dermatol ; 142(9): 2455-2463.e9, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35148999

RESUMEN

PFN1 is an actin-binding protein that regulates actin polymerization, cell proliferation, apoptosis, angiogenesis, and carcinogenesis. Its dysregulation has been reported in diverse pathologic diseases; however, the role of PFN1 in psoriasis has not yet been elucidated. In this study, we show that PFN1 expression is increased in both skin and serum of patients with psoriasis. PFN1 was markedly expressed in the epidermis of psoriatic lesions, and its expression positively correlated with psoriasis severity. IL-17A treatment of keratinocytes increased PFN1 expression, whereas TNF-α induced PFN1 expression and secretion. In addition, knockdown of PFN1 with short hairpin RNA resulted in an altered expression of psoriasis-associated inflammatory markers, HBD2, S100A7, S100A9, and Ki-67, and recombinant PFN1 suppressed the IL-17A‒induced inflammatory response in keratinocytes. Interestingly, recombinant PFN1 also suppressed IL-17A‒induced IκBζ, an important player in immune response in psoriasis. Collectively, our results show that PFN1 acts as a negative regulator of psoriatic inflammation through the suppression of IκBζ and that the balanced level of PFN1 is important for IκBζ regulation. Thus, the expression of PFN1 can be used as a biomarker for psoriasis severity, and it can be considered as a possible target for the treatment of psoriasis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Profilinas , Psoriasis , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Humanos , Interleucina-17/farmacología , Queratinocitos/metabolismo , Profilinas/genética , Profilinas/metabolismo , Psoriasis/patología , Piel/patología , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA