Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(12)2022 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-35743036

RESUMEN

Nanos RNA-binding proteins are critical factors of germline development throughout the animal kingdom and their dysfunction causes infertility. During evolution, mammalian Nanos paralogues adopted divergent roles in germ cell biology. However, the molecular basis behind this divergence, such as their target mRNAs, remains poorly understood. Our RNA-sequencing analysis in a human primordial germ cell model-TCam-2 cell line revealed distinct pools of genes involved in the cell cycle process downregulated upon NANOS1 and NANOS3 overexpression. We show that NANOS1 and NANOS3 proteins influence different stages of the cell cycle. Namely, NANOS1 is involved in the G1/S and NANOS3 in the G2/M phase transition. Many of their cell cycle targets are known infertility and cancer-germ cell genes. Moreover, NANOS3 in complex with RNA-binding protein PUM1 causes 3'UTR-mediated repression of FOXM1 mRNA encoding a transcription factor crucial for G2/M phase transition. Interestingly, while NANOS3 and PUM1 act as post-transcriptional repressors of FOXM1, FOXM1 potentially acts as a transcriptional activator of NANOS3, PUM1, and itself. Finally, by utilizing publicly available RNA-sequencing datasets, we show that the balance between FOXM1-NANOS3 and FOXM1-PUM1 expression levels is disrupted in testis cancer, suggesting a potential role in this disease.


Asunto(s)
Células Germinativas , Infertilidad , Animales , Ciclo Celular/genética , División Celular , Proteína Forkhead Box M1/metabolismo , Células Germinativas/metabolismo , Humanos , Infertilidad/metabolismo , Masculino , Mamíferos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo
2.
Biochem Cell Biol ; 99(6): 735-740, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34734756

RESUMEN

Preeclampsia is a severe pregnancy complication characterized by hypertension and may cause maternal morbidity and mortality. A better understanding of the essential genes involved in preeclampsia pathophysiology is urgently needed. This study investigated the function and molecular mechanisms of pumilio RNA binding family member 1 (PUM1) in extravillous trophoblast cells (EVTs). The interaction between protein and mRNA was verified by RNA pull-down assays, RNA immunoprecipitation assays, and luciferase reporter assays. The mRNA and protein levels of the genes involved were determined by RT-qPCR and western blot assays, respectively. Our results demonstrated that PUM1 could bind to the 3'-untranslated region of low-density lipoprotein receptor-related protein 6 (LRP6) mRNA, resulting in reduced expression of LRP6 mRNA and protein. Repression of PUM1 resulted in enhanced colony formation, cell proliferation, migration, and invasion of EVTs. The PUM1-depletion-mediated promotion effects on EVTs could be abrogated by LRP6 knockdown. PUM1 regulates the growth and mobility of EVTs by modulating LRP6 expression. Developing strategies to balance PUM1 and LRP6 levels may be beneficial for the management of preeclampsia patients.


Asunto(s)
Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Trofoblastos , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Embarazo , Proteínas de Unión al ARN , Trofoblastos/metabolismo
3.
Mol Ther ; 28(2): 631-641, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31862314

RESUMEN

Pumilio (PUM) proteins are members of a highly conserved RNA-binding protein family that posttranscriptionally regulate gene expression in many organisms. However, their roles in the placenta are unclear. In the present study, we report the requirement for the PUM homolog 1 (PUM1) gene in preeclampsia (PE). Immunofluorescence and immunohistochemical data showed that PUM1 was highly expressed in human placental villi from women with PE compared to healthy controls (HCs). Further, PUM1 overexpression repressed, and knockdown enhanced, the invasion and proliferation of trophoblasts. Interestingly, PUM1 knockdown promoted trophoblast invasion in a villous explant culture model, while PUM1 overexpression repressed these effects. Furthermore, lncRNA transcriptome sequencing coupled with RNA immunoprecipitation (RIP) revealed that PUM1 inhibits trophoblast invasion in PE by downregulating the expression of lncRNA HOTAIR. Moreover, PUM1 regulates HOTAIR expression via a posttranscriptional mechanism. Using RNA-protein pull-down and mRNA stability assays, we identified PUM1 as a specific binding partner that decreased the half-life of HOTAIR and lowered the steady-state level of HOTAIR expression, suggesting a novel posttranscriptional regulatory mechanism. Collectively, these findings identified a novel RNA regulatory mechanism, revealing a new pathway governing the regulation of PUM1/HOTAIR in trophoblast invasion in the pathogenesis of PE.


Asunto(s)
Regulación de la Expresión Génica , Preeclampsia/genética , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/genética , Trofoblastos/metabolismo , Línea Celular , Movimiento Celular/genética , Proliferación Celular , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Preeclampsia/metabolismo , Embarazo , Estabilidad del ARN
4.
J Cell Mol Med ; 24(7): 4127-4135, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32073729

RESUMEN

Endometrial cancer is one of the most common gynaecological malignancies and the sixth most common cause of cancer-related death among women. Here, we define the role and molecular mechanism of circ_0000043 (hereafter referred to as circ_PUM1) in the development and progression of endometrial carcinoma. QRT-PCR was used to detect the expression of circ_PUM1 in normal endometrial tissue and endometrial carcinoma tissues. Changes in cell function and tumorigenicity in nude mice were examined after circ_PUM1 overexpression or knockdown. Bioinformatic analysis and dual-luciferase reporter assay were used to predict and analyse the miRNAs that circ_PUM1 binds. Gene expression changes were analysed using Western blot. Circ_PUM1 was expressed at significantly higher levels in endometrial cancer tissues than in normal tissues. Up-regulation of circ_PUM1 promoted the proliferation, migration and invasion of endometrial carcinoma cells. Opposite results were observed with circ_PUM1 knockdown, and the tumorigenic ability of endometrial cancer cells after circ_PUM1 knockdown was reduced compared to control cells. Circ_PUM1 is capable of binding to miR-136, and up-regulating its target gene NOTCH3, which can be reversed by overexpression of miR-136. Circ_PUM1 can compete with miR-136, leading to up-regulation of NOTCH3, and thereby promote the development of endometrial cancer.


Asunto(s)
Neoplasias Endometriales/genética , MicroARNs/genética , ARN Circular/genética , Proteínas de Unión al ARN/genética , Animales , Apoptosis/genética , Carcinogénesis/genética , Movimiento Celular , Proliferación Celular/genética , Neoplasias Endometriales/patología , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Xenoinjertos , Humanos , Ratones , Receptor Notch3/genética , Transducción de Señal
5.
Proc Natl Acad Sci U S A ; 114(33): E6902-E6911, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28760986

RESUMEN

PUM1 is an RNA binding protein shown to regulate the stability and function of mRNAs bearing a specific sequence. We report the following: (i) A key function of PUM1 is that of a repressor of key innate immunity genes by repressing the expression of LGP2. Thus, between 12 and 48 hours after transfection of human cells with siPUM1 RNA there was an initial (phase 1) upsurge of transcripts encoding LGP2, CXCL10, IL6, and PKR. This was followed 24 hours later (phase 2) by a significant accumulation of mRNAs encoding RIG-I, SP100, MDA5, IFIT1, PML, STING, and IFNß. The genes that were not activated encoded HDAC4 and NF-κB1. (ii) Simultaneous depletion of PUM1 and LGP2, CXCL10, or IL6 revealed that up-regulation of phase 1 and phase 2 genes was the consequence of up-regulation of LGP2. (iii) IFNß produced 48-72 hours after transfection of siPUM1 was effective in up-regulating LGP2 and phase 2 genes and reducing the replication of HSV-1 in untreated cells. (iv) Because only half of genes up-regulated in phase 1 and 2 encode mRNAs containing PUM1 binding sites, the upsurge in gene expression could not be attributed solely to stabilization of mRNAs in the absence of PUM1. (v) Lastly, depletion of PUM2 does not result in up-regulation of phase 1 or phase 2 genes. The results of the studies presented here indicate that PUM1 is a negative regulator of LGP2, a master regulator of innate immunity genes expressed in a cascade fashion.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad Innata/genética , ARN Helicasas/genética , Proteínas de Unión al ARN/genética , Línea Celular Tumoral , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Humanos , Interferón beta/genética , Interferón beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , ARN Helicasas/metabolismo , Interferencia de ARN , Proteínas de Unión al ARN/metabolismo
6.
Trends Genet ; 32(7): 390-392, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27157388

RESUMEN

Long non-coding RNAs (lncRNAs) are a fascinating, but still largely uncharacterized, class of genes. A recent paper by the Mendell group identifies NORAD, a novel lncRNA that is regulated in response to DNA damage and plays a key role in maintaining genome integrity by modulating the activity the RNA binding proteins PUM2 and PUM1.


Asunto(s)
Daño del ADN/genética , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/genética , Secuencia de Aminoácidos/genética , Animales , Genoma , Humanos , Mamíferos/genética
7.
Neurobiol Learn Mem ; 165: 106902, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30030131

RESUMEN

Animal models have been the mainstay of biological and medical research. Although there are drawbacks to any research tool, we argue that mice have been under-utilized as a tool for predicting human diseases. Here we review four examples from our research group where studying the consequences of altered gene dosage in a mouse led to the discovery of previously unrecognized human syndromes: MECP2 duplication syndrome, SHANK3 duplication syndrome, CIC haploinsufficiency syndrome, and PUM1-related disorders. We also describe the clinical phenotypes of two individuals with CIC haploinsufficiency syndrome who have not been reported previously. To help bring biological insights gained from model systems a step closer to disease gene discovery, we discuss tools and resources that will facilitate this process. Moving back and forth between the lab and the clinic, between studies of mouse models and human patients, will continue to drive disease gene discovery and lead to better understanding of gene functions and disease mechanisms, laying the groundwork for future therapeutic interventions.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades del Sistema Nervioso/genética , Animales , Estudios de Asociación Genética/métodos , Haploinsuficiencia/genética , Humanos , Discapacidad Intelectual Ligada al Cromosoma X/genética , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética
8.
Am J Med Genet A ; 179(6): 1030-1033, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30903679

RESUMEN

PUM1 has been very recently reported as responsible for a new form of developmental disorder named PADDAS syndrome. We describe here an additional patient with early onset developmental delay, epilepsy, microcephaly, and hair dysplasia, with a de novo heterozygous missense variant of PUM1: c.3439C > T, p.(Arg1147Trp). This variant was absent from databases and predicted deleterious by multiple softwares. The same missense variant has been reported by Gennarino et al., in a girl with much more severe epilepsy. Our report is in favor of a variable expressivity of PADDAS syndrome, and broadens the phenotypic spectrum with the description of hair dysplasia.


Asunto(s)
Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mutación Missense , Fenotipo , Proteínas de Unión al ARN/genética , Adolescente , Niño , Preescolar , Epilepsia/diagnóstico , Epilepsia/genética , Femenino , Estudios de Asociación Genética/métodos , Humanos , Hipotricosis/diagnóstico , Hipotricosis/genética , Imagen por Resonancia Magnética , Masculino , Microcefalia/diagnóstico , Microcefalia/genética , Síndrome
9.
Biochem Biophys Res Commun ; 497(1): 313-318, 2018 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-29428722

RESUMEN

BACKGROUND: Abnormal expression of the PUM1 gene (pumilio RNA binding family member 1) is closely related to chromosomal mutations and carcinogenesis. However, there is no report about expression or function of PUM1 in ovarian cancer. The present study explored the role of PUM1 in the development and progression of ovarian cancer. METHODS: Immunohistochemistry was used to detect the expression of PUM1 in normal ovarian tissues and ovarian cancer tissues. The PUM1 gene was silenced using small interfering RNAs in ovarian cancer cell line A2780. MTT, plate colony formation and EdU (5-ethynyl-2'-deoxyuridine) assays were used to detect cell growth, and cell apoptosis was detected by flow cytometry. Wound-healing and Transwell assays were performed to determine cell migration and invasion. Western blotting was used to detect the levels of cancer-related proteins. RESULTS: Immunohistochemistry showed that the level of PUM1 in ovarian cancer tissues was higher than that in normal tissues. The cell proliferation, migration, and invasion ability decreased significantly, while cell apoptosis increased after silencing the PUM1 gene. Moreover, western blotting showed that downregulation of PUM1 decreased the levels of STAT3, BCL2, MMP2, and VEGFA. CONCLUSIONS: Thus, PUM1 promotes the development and progression of ovarian cancer, which may occur via the above-mentioned molecules.


Asunto(s)
Movimiento Celular , Proliferación Celular , Neoplasias Ováricas/patología , Neoplasias Ováricas/fisiopatología , Adulto , Femenino , Humanos , Persona de Mediana Edad , Invasividad Neoplásica , Células Tumorales Cultivadas
10.
J Assist Reprod Genet ; 35(4): 727-731, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29297114

RESUMEN

Accumulating evidence has indicated that the genes involved in meiosis are highly correlated with ovarian function. Pumilio 1 (PUM1) is a RNA-binding protein which is involved in the meiotic process. It has been reported that the Pum1 knockout female mice displayed subfertility due to the decrease in primordial follicle pool. The aim of our study is to investigate whether variants of the PUM1 gene are responsible for primary ovarian insufficiency (POI) in Chinese women. We analyzed coding sequence and untranslated regions of the PUM1 gene in 196 Han Chinese women with non-syndromic POI and 192 controls. Seven novel variants were identified, but one of them was synonymous and six were intronic. Besides, seven known single-nucleotide polymorphisms (SNPs) were found, and there were no significant differences in genotype and allele frequencies of the SNPs between patients and controls. The results suggest that the variants in PUM1 may not contribute to POI in Han Chinese women.


Asunto(s)
Pueblo Asiatico/genética , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple , Insuficiencia Ovárica Primaria/genética , Proteínas de Unión al ARN/genética , Adulto , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Insuficiencia Ovárica Primaria/patología , Pronóstico
11.
Genes (Basel) ; 15(1)2024 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-38275605

RESUMEN

CircRNAs are a class of non-coding RNAs able to regulate gene expression at multiple levels. Their involvement in physiological processes, as well as their altered regulation in different human diseases, both tumoral and non-tumoral, is well documented. However, little is known about their involvement in female reproduction. This study aims to identify circRNAs potentially involved in reproductive women's health. Candidate circRNAs expressed in ovary and sponging miRNAs, already known to be expressed in the ovary, were selected by a computational approach. Using real time PCR, we verified their expression and identified circPUM1 as the most interesting candidate circRNA for further analyses. We assessed the expression of circPUM1 and its linear counterpart in all the follicle compartments and, using a computational and experimental approach, identified circPUM1 direct and indirect targets, miRNAs and mRNAs, respectively, in cumulus cells. We found that both circPUM1 and its mRNA host gene are co-expressed in all the follicle compartments and proposed circPUM1 as a potential regulator of PTEN, finding a strong positive correlation between circPUM1 and PTEN mRNA. These results suggest a possible regulation of PTEN by circPUM1 in cumulus cells and point out the important role of circRNA inside the pathways related to follicle growth and oocyte maturation.


Asunto(s)
MicroARNs , ARN Circular , Femenino , Humanos , Células del Cúmulo/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , ARN Circular/genética , ARN Circular/metabolismo , ARN Mensajero/metabolismo
12.
Cancer Lett ; 581: 216498, 2024 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-38029539

RESUMEN

Abnormal regulation of RNA binding proteins (RBPs) plays an essential role in tumorigenesis and progression, but their functions and mechanisms remain largely elusive. Previously, we reported that Pumilio 1 (PUM1), a RBP, could regulate glycolysis metabolism and promote the progression of gastric cancer (GC). However, the role of PUM1 in tumor immune regulation remains largely elusive. In this study, we report that PUM1 induces immune escape through posttranscriptional regulation of PD-L1 in GC. We used multiplexed immunohistochemistry to analyze the correlation between PUM1 expression and immune microenvironment in GC. The effect of PUM1 deficiency on tumor killing of T cells was examined in vitro and in vivo. The molecular mechanism of PUM1 was evaluated via RNA immunoprecipitation, chromatin immunoprecipitation, Western blot, co-immunoprecipitation, and RNA stability assays. Clinically, elevated PUM1 expression is associated with high-expression of PD-L1, lack of CD8+ T cell infiltration and poor prognosis in GC patients. PUM1 positively regulates PD-L1 expression and PUM1 reduction enhances T cell killing of tumors. Mechanistically, PUM1 directly binds to nucleophosmin/nucleoplasmin 3 (NPM3) mRNA and stabilizes NPM3. NPM3 interacts with NPM1 to promote NPM1 translocation into the nucleus and increase the transcription of PD-L1. PUM1 inhibits the anti-tumor activity of T cells through the PUM1/NPM3/PD-L1 axis. In summary, this study reveals the critical post-transcriptional effect of PUM1 in the modulation of PD-L1-dependent GC immune escape, thus provides a novel indicator and potential therapeutic target for cancer immunotherapy.


Asunto(s)
Neoplasias Gástricas , Humanos , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos , Línea Celular Tumoral , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleoplasminas/metabolismo , Proteínas de Unión al ARN/genética , Neoplasias Gástricas/patología , Microambiente Tumoral
13.
Cytotechnology ; 76(1): 53-68, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38304630

RESUMEN

In recent years, gastric cancer (GC) is still one of the major public health burdens in the world. It is reported that exosome circular RNA (circRNA) is involved in the GC progression. However, the function and potential mechanism of circGMPS in GC remains unclear and needs further exploration. In this study, we isolated and identified exosomes from serum by TEM, NTA analysis and Western blot. RNA expression was evaluated by qRT-PCR. Western blot was employed to examine protein expression. Cell proliferation was measured using CCK-8. Transwell assay was adopted to analyze cell migration and invasion. The relationship between genes was explored through bioinformatics analysis, dual-luciferase reporter gene assay and spearman correlation coefficient. We found that circGMPS was elevated in GC exosomes, tissues and cells. Poor prognosis of GC patients was related to high circGMPS expression. Both exosome co-culture with cells and insertion of circGMPS clearly promoted cell progression. Mechanically, circGMPS sponged miR-144-3p to regulate PUM1. Inhibition of PUM1 or miR-144-3p overexpression inhibited the malignant GC cell progression. Our data confirmed that exosome-derived circGMPS boosted malignant progression by miR-144-3p/PUM1 axis in GC cells, providing strong evidences for circGMPS as a clinical biomarker of GC treatment. Supplementary Information: The online version contains supplementary material available at 10.1007/s10616-023-00597-9.

14.
Genetics ; 227(1)2024 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-38427913

RESUMEN

RNA-binding proteins FBF-1 and FBF-2 (FBFs) are required for germline stem cell maintenance and the sperm/oocyte switch in Caenorhabditis elegans, although the mechanisms controlling FBF protein levels remain unknown. We identified an interaction between both FBFs and CSN-5), a component of the constitutive photomorphogenesis 9 (COP9) signalosome best known for its role in regulating protein degradation. Here, we find that the Mpr1/Pad1 N-terminal metalloprotease domain of CSN-5 interacts with the Pumilio and FBF RNA-binding domain of FBFs and the interaction is conserved for human homologs CSN5 and PUM1. The interaction between FBF-2 and CSN-5 can be detected in vivo by proximity ligation. csn-5 mutation results in the destabilization of FBF proteins, which may explain previously observed decrease in the numbers of germline stem and progenitor cells, and disruption of oogenesis. The loss of csn-5 does not decrease the levels of a related PUF protein PUF-3, and csn-5(lf) phenotype is not enhanced by fbf-1/2 knockdown, suggesting that the effect is specific to FBFs. The effect of csn-5 on oogenesis is largely independent of the COP9 signalosome and is cell autonomous. Surprisingly, the regulation of FBF protein levels involves a combination of COP9-dependent and COP9-independent mechanisms differentially affecting FBF-1 and FBF-2. This work supports a previously unappreciated role for CSN-5 in the stabilization of germline stem cell regulatory proteins FBF-1 and FBF-2.


Asunto(s)
Complejo del Señalosoma COP9 , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Complejo del Señalosoma COP9/metabolismo , Complejo del Señalosoma COP9/genética , Células Germinativas/metabolismo , Oogénesis/genética , Estabilidad Proteica , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Células Madre/metabolismo , Células Madre/citología
15.
Cell Cycle ; 23(6): 682-692, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38794797

RESUMEN

Pumilio RNA-binding family member 1 (PUM1) has been implicated in both the progression of colorectal cancer and the regulation of inflammation. The role of PUM1 in the polarization of tumor-associated macrophages (TAMs) into the M2 phenotype has not yet been reported in hepatocellular carcinoma. Using the PUM1-knockout mice model, flow cytometry, and IHC, we validated the role of PUM1 in hepatocellular carcinoma (HCC) TAMs. One-way analysis of variance (ANOVA) or student's t-tests was used to compare the experimental groups. We found that PUM1 inhibited anti-tumor immunity in HCC through TAM-mediated inhibition of CD8+ T cells. We also showed that PUM1 promotes the transformation of TAMs into pro-tumorigenic M2-like phenotypes by activating cAMP signaling pathway. This study emphasized the potential of PUM1 as a target for immunotherapy in HCC through TAMs. The present study revealed the molecular mechanism underlying the pro-tumor role of PUM1 in HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Macrófagos , Ratones Noqueados , Proteínas de Unión al ARN , Animales , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones , Macrófagos/metabolismo , Macrófagos/inmunología , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/patología , Ratones Endogámicos C57BL , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Humanos , Transducción de Señal , Línea Celular Tumoral
16.
Adv Sci (Weinh) ; 10(27): e2301190, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37469018

RESUMEN

RNA-binding proteins (RBPs) play essential roles in tumorigenesis and progression, but their functions in gastric cancer (GC) remain largely elusive. Here, it is reported that Pumilio 1 (PUM1), an RBP, induces metabolic reprogramming through post-transcriptional regulation of DEP domain-containing mammalian target of rapamycin (mTOR)-interacting protein (DEPTOR) in GC. In clinical samples, elevated expression of PUM1 is associated with recurrence, metastasis, and poor survival. In vitro and in vivo experiments demonstrate that knockdown of PUM1 inhibits the proliferation and metastasis of GC cells. In addition, RNA-sequencing and bioinformatics analyses show that PUM1 is enriched in the glycolysis gene signature. Metabolomics studies confirm that PUM1 deficiency suppresses glycolytic metabolism. Mechanistically, PUM1 binds directly to DEPTOR mRNA pumilio response element to maintain the stability of the transcript and prevent DEPTOR degradation through post-transcriptional pathway. PUM1-mediated DEPTOR upregulation inhibits mTORC1 and alleviates the inhibitory feedback signal transmitted from mTORC1 to PI3K under normal conditions, thus activating the PI3K-Akt signal and glycolysis continuously. Collectively, these results reveal the critical epigenetic role of PUM1 in modulating DEPTOR-dependent GC progression. These conclusions support further clinical investigation of PUM1 inhibitors as a metabolic-targeting treatment strategy for GC.


Asunto(s)
Transducción de Señal , Neoplasias Gástricas , Humanos , Fosfatidilinositol 3-Quinasas , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Gástricas/genética , Serina-Treonina Quinasas TOR/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
17.
Aging (Albany NY) ; 15(12): 5662-5672, 2023 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-37387538

RESUMEN

BACKGROUND: To study the link between macrophage polarization, PUM1/Cripto-1 pathway and ferroptosis in the allogeneic blood transfusion setting. METHODS: This is an exploratory research. The purpose of this study was to investigate the effect of PUM1/Cripto-1 pathway on ferroptosis by regulating macrophage polarization in allogeneic blood transfused mice. Establish in vitro cell models and in vivo rat models. To find out whether PUM1 and Cripto-1 were expressed, RT-qPCR and Western blot analyses were employed. The macrophage polarization markers iNOS, TNF-, IL-1, IL-6, Arg-1, and IL-10 were utilized to identify M1 and M2 macrophages. JC-1 staining was used to detect ATP membrane potential in peripheral blood macrophages. RESULTS: In animal experiments, expression of Cripto-1 was negatively regulated by PUM1 and promoted M1 type polarization of macrophages. Allogeneic blood transfusion assured good state of macrophage mitochondria. Allogeneic blood transfusion inhibited ferroptosis in macrophages by affecting the PUM1/Cripto-1 pathway. In cell experiments, PUM1 regulated Cripto-1 in mouse macrophage RAW264.7. Polarization of RAW264.7 cells was regulated by the PUM1/Cripto-1 pathway. The effect of PUM1/Cripto-1 pathway on macrophage ferroptosis in cell experiments was consistent with that in animal experiments. CONCLUSIONS: In this study, through in vivo cell experiments and in vitro animal experiments, it was successfully proved that PUM1/Cripto-1 pathway affected ferroptosis by regulating macrophage polarization in allogeneic blood transfused mice.


Asunto(s)
Ferroptosis , Trasplante de Células Madre Hematopoyéticas , Ratones , Ratas , Animales , Macrófagos/metabolismo , Células RAW 264.7 , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Transfusión Sanguínea
18.
J Gastrointest Oncol ; 14(1): 233-244, 2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36915463

RESUMEN

Background: Colorectal cancer (CRC) is the third most common cancer and the fourth most common cause of cancer-related death worldwide. Advanced stage CRC, during the recent past, had a dismal prognosis and only a few available treatments. Pumilio homologous protein 1 (PUM1) is reportedly aberrant in human malignancies, including CRC. However, the role of PUM1 in the regulation of tumor-initiating cells (T-ICs) remains unknown. Methods: The levels of messenger RNAs (mRNAs) were determined by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunoblot analyses. Statistical analyses were performed to determine the associations between the levels of PUM1 and tumor features and patient outcomes. Whether PUM1 is a downstream target of miR-218-5p was verified by bioinformatics target gene prediction and qRT-PCR. Results: Herein, it was found that T-ICs, chemoresistance, and recurrent CRC samples all manifest increased PUM1 expression. Functional investigations have shown that PUM1 increased the self-renewal, tumorigenicity, malignant proliferation, and chemoresistance of colorectal cells. PUM1 activates the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) signaling pathway biochemically. Furthermore, it was discovered that miR-218-5p specifically targets T-ICs' PUM1 3'-untranslated region (3'-UTR). More importantly, the PUM1/PI3K/AKT axis regulates CRC cells' responses to treatment with cetuximab, and PUM1 overexpression increased cetuximab resistance. More evidence points to the possibility that low PUM1 may predict cetuximab benefits in CRC patients after analysis of the patient cohort, patient-derived tumor organoids, and patient-derived xenografts (PDXs). Conclusions: Taken together, the result of this work points to the critical function of the miR-218-5p/PUM1/PI3K/AKT regulatory circuit in regulating T-ICs characteristics and thus suggests possible therapeutic targets for CRC.

19.
Cureus ; 14(10): e30577, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36320799

RESUMEN

Epilepsy is a neurological condition brought on by recurrent and spontaneous seizures in patients with hypersynchronous neuronal ensemble activity. These spontaneous seizures appear to be brought on by increased neuronal excitability and synaptic synchronization. The development of neuronal hyperexcitability and acquiring epilepsy is still poorly understood. Cell differentiation and development might be related to the pumilio RNA-binding family member 1 (Pumilio 1 (PUM1)). Complete deficiency of this gene causes misregulation of the proteins involved in the control of neuronal excitability. Furthermore, the voltage-gated sodium channels alpha subunit 2 (SCN2A) triggers action potentials in brain neurons, and a variety of severe hereditary epilepsy syndromes are caused by their mutation. Here, we present a rare case of a seven-year-old female with co-occurrence of two genetic mutations in the pumilio homolog 1 (PUM1) and sodium voltage-gated channel alpha subunit 2 (SCN2A).

20.
Front Pediatr ; 10: 759889, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35386260

RESUMEN

In the recent 3 years, subjects with Pumilio1-associated developmental disability, ataxia, and seizure syndrome have been identified as harboring Pumilio homolog 1 (PUM1) mutations. However, the characteristics of the seizure phenotype remain to be elucidated. We herein described a 3-year-old female proband who was diagnosed with developmental and epileptic encephalopathy presenting with some features suggestive of a Dravet-like syndrome. For genetic analyses, trio-based whole-exome sequencing and array comparative genomic hybridization were performed. Consequently, a de novo heterozygous missense variant was identified in exon 22 of the PUM1 gene: NM_001020658: c.3439C > T (p.Arg1147Trp). Upon thoroughly reviewing the existing literature, nine cases of PUM1 mutation-related epilepsy were identified, and their clinical features were summarized. A relationship between PUM1 mutation and clinical manifestations characteristic of a Dravet-like syndrome was proposed. To our knowledge, this is the first report of a patient with PUM1 mutation presenting with a Dravet-like syndrome.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA