Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Intervalo de año de publicación
1.
FASEB J ; 38(18): e70055, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39305126

RESUMEN

Spinal Muscular Atrophy (SMA) is a neuromuscular disease caused by low levels of the Survival of Motoneuron (SMN) protein. SMN interacts with and regulates the actin-binding protein profilin2a, thereby influencing actin dynamics. Dysfunctional actin dynamics caused by SMN loss disrupts neurite outgrowth, axonal pathfinding, and formation of functional synapses in neurons. Whether the SMN protein directly interacts with and regulates filamentous (F-) and monomeric globular (G-) actin is still elusive. In a quantitative single cell approach, we show that SMN loss leads to dysregulated F-/G-actin fractions. Furthermore, quantitative assessment of cell morphology suggests an F-actin organizational defect. Interestingly, this is mediated by an interaction of SMN with G- and F-actin. In co-immunoprecipitation, in-vitro pulldown and co-localization assays, we elucidated that this interaction is independent of the SMN-profilin2a interaction. Therefore, we suggest two populations being relevant for functional actin dynamics in healthy neurons: SMN-profilin2a-actin and SMN-actin. Additionally, those two populations may influence each other and therefore regulate binding of SMN to actin. In SMA, we showed a dysregulated co-localization pattern of SMN-actin which could only partially rescued by SMN restoration. However, dysregulation of F-/G-actin fractions was reduced by SMN restoration. Taken together, our results suggest a novel molecular function of SMN in binding to actin independent from SMN-profilin2a interaction.


Asunto(s)
Actinas , Atrofia Muscular Espinal , Profilinas , Proteína 1 para la Supervivencia de la Neurona Motora , Actinas/metabolismo , Profilinas/metabolismo , Profilinas/genética , Humanos , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patología , Atrofia Muscular Espinal/genética , Animales , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Ratones , Neuronas Motoras/metabolismo , Unión Proteica
2.
Oncol Lett ; 28(2): 381, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38939626

RESUMEN

Lung metastasis is the second most common type of metastasis in colorectal cancer. Specific treatments for lung metastasis have not been developed since the underlying mechanisms are poorly understood. The present study aimed to elucidate the molecular basis of lung metastasis in colorectal cancer. In a mouse model, cell lines that were highly metastatic to the lungs were established by injecting colorectal cancer cells through the tail vein and removing them from the lungs. Differential gene expression comparing the transfected cells with their parental cells was investigated using DNA microarrays. The results were functionally interpreted using gene enrichment analysis and validated using reverse transcription-quantitative PCR (RT-qPCR). The isoforms of the identified genes were examined by melting curve analysis. The present study established colorectal cancer cell lines that were highly metastatic to the lungs. DNA microarray experiments revealed that genes (N-cadherin, VE-cadherin, Six4, Akt and VCAM1) involved in motility, proliferation and adhesion were upregulated, and genes (tissue inhibitor of metalloproteinase-3 and PAX6) with tumor-suppressive functions were downregulated in metastatic cells. Profilin 2 (PFN2) expression was upregulated in multiple metastatic cell lines using RT-qPCR. Two PFN2 isoforms were overexpressed in metastatic cells. In vitro and in vivo models were established and genes associated with lung metastasis were identified to overcome the heterogeneity of the disease. Overall, aberrant PFN2 expression is unreported in lung metastasis in colorectal cancer. In the present study, two PFN2 isoforms with differential tissue distribution were upregulated in metastatic cells, suggesting that they promote lung metastasis in colorectal cancer.

3.
Cell Mol Biol Lett ; 29(1): 43, 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38539084

RESUMEN

BACKGROUND: Circular RNAs (circRNAs) are single-stranded RNAs with covalently closed structures that have been implicated in cancer progression. However, the regulatory mechanisms remain largely unclear. So, the aim of this study was to reveal the role and regulatory mechanisms of circ-SLC16A1. METHODS: In this study, next-generation sequencing was used to identify abnormally expressed circRNAs between cancerous and para-carcinoma tissues. Fluorescence in situ hybridization and quantitative reverse transcription polymerase chain reaction were performed to assess the expression patterns of circ-solute carrier family 16 member 1 (SLC16A1) in non-small cell lung cancer (NSCLC) cells and tissue specimens. The dual-luciferase reporter assay was utilized to identify downstream targets of circ-SLC16A1. Transwell migration, wound healing, 5-ethynyl-2'-deoxyuridine incorporation, cell counting, and colony formation assays were conducted to assess the proliferation and migration of NSCLC cells. A mouse tumor xenograft model was employed to determine the roles of circ-SLC16A1 in NSCLC progression and metastasis in vivo. RESULTS: The results found that circ-SLC16A1 was upregulated in NSCLC cells and tissues. Downregulation of circ-SLC16A1 inhibited tumor growth by reducing proliferation, lung metastasis, and lymphatic metastasis of NSCLC cells, and arrested the cell cycle in the G1 phase. Also, silencing of circ-SLC16A1 promoted apoptosis of NSCLC cells. The results of bioinformatics analysis and the dual-luciferase reporter assay confirmed that microRNA (miR)-1287-5p and profilin 2 (PFN2) are downstream targets of circ-SLC16A1. PFN2 overexpression or circ-SLC16A1 inhibition restored proliferation and migration of NSCLC cells after silencing of circ-SLC16A1. PFN2 overexpression restored migration and proliferation of NSCLC cells post miR-1287-5p overexpression. CONCLUSIONS: Collectively, these findings show that miR-1287-5p/PFN2 signaling was associated with downregulation of circ-SLC16A1 and reduced invasion and proliferation of NSCLC cells. So, circ-SLC16A1 is identified as a mediator of multiple pro-oncogenic signaling pathways in NSCLC and can be targeted to suppress tumor progression.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , MicroARNs , Animales , Humanos , Ratones , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Modelos Animales de Enfermedad , Hibridación Fluorescente in Situ , Luciferasas , Neoplasias Pulmonares/genética , MicroARNs/genética , Profilinas , ARN Circular/genética
4.
FASEB J ; 37(10): e23170, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37676718

RESUMEN

Small cell lung cancer (SCLC) is one of the most malignant tumors that has an extremely poor prognosis. RNA-binding protein (RBP) and long noncoding RNA (lncRNA) have been shown to be key regulators during tumorigenesis as well as lung tumor progression. However, the role of RBP ELAVL4 and lncRNA LYPLAL1-DT in SCLC remains unclear. In this study, we verified that lncRNA LYPLAL1-DT acts as an SCLC oncogenic lncRNA and was confirmed in vitro and in vivo. Mechanistically, LYPLAL1-DT negatively regulates the expression of miR-204-5p, leading to the upregulation of PFN2, thus, promoting SCLC cell proliferation, migration, and invasion. ELAVL4 has been shown to enhance the stability of LYPLAL1-DT and PFN2 mRNA. Our study reveals a regulatory pathway, where ELAVL4 stabilizes PFN2 and LYPLAL1-DT with the latter further increasing PFN2 expression by blocking the action of miR-204-5p. Upregulated PFN2 ultimately promotes tumorigenesis and invasion in SCLC. These findings provide novel prognostic indicators as well as promising new therapeutic targets for SCLC.


Asunto(s)
Neoplasias Pulmonares , MicroARNs , ARN Largo no Codificante , Carcinoma Pulmonar de Células Pequeñas , Humanos , ARN Largo no Codificante/genética , Profilinas/genética , Carcinoma Pulmonar de Células Pequeñas/genética , Transformación Celular Neoplásica/genética , Carcinogénesis/genética , Neoplasias Pulmonares/genética , MicroARNs/genética , Proteína 4 Similar a ELAV
5.
Cell Tissue Res ; 392(3): 779-791, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36788143

RESUMEN

Previous reports from this laboratory have demonstrated the involvement of histone deacetylase 6 (HDAC6) in sperm motility. As the presence of HDAC6 has also been reported in the earlier stage germ cells, studies were undertaken to explore its role during these stages of spermatogenesis. HDAC6 was overexpressed in GC-1spg cells, which represent the stage between type B spermatogonia and primary spermatocyte, and its effect on germ cell transcriptome was investigated by microarray. Among the many transcripts that were differentially regulated, Profilin 2, reported previously as a neuronal specific isoform, was observed as one of the genes highly upregulated at the transcript level, which was further confirmed by real-time PCR, and the protein confirmed by indirect immunofluorescence (IIF). Profilin 2 colocalized with HDAC6, as seen both in GC-1 cells and sperm. On the sperm, the presence of Profilin 2 was detected throughout the flagella with its colocalization with HDAC6 seen conspicuously in the mid-piece region of the flagella. Co-immunoprecipitation studies confirmed Profilin 2 interaction with HDAC6. Docking studies using Z dock suggested the interaction of 8 residues of HDAC6 with 6 residues of Profilin 2. The novel observation of Profilin 2 in spermatogonial cells, its significant upregulation on HDAC6 overexpression and its interaction with HDAC6 suggests that HDAC6 in collaboration with Profilin 2 may play a role in regulating the movement of germ cells from one stage/compartment to the next.


Asunto(s)
Profilinas , Testículo , Masculino , Ratones , Animales , Testículo/metabolismo , Histona Desacetilasa 6/metabolismo , Profilinas/genética , Profilinas/metabolismo , Regulación hacia Arriba , Motilidad Espermática , Semen/metabolismo
6.
Strahlenther Onkol ; 199(1): 78-89, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36205752

RESUMEN

BACKGROUND: Circular RNA (circRNA) has been proven to play a critical role in breast cancer progression. Therefore, this study was designed to clarify the role and underlying molecular mechanisms of circ-disintegrin and metalloproteinase 9 (circ-ADAM9) in breast cancer. METHODS: A quantitative real-time polymerase chain reaction (RT-qPCR) was conducted to assess the expression levels of circ-ADAM9, microRNA-383-5p (miR-383-5p), and profilin 2 (PFN2). Cellular growth curves of breast cancer cells were determined by colony-forming assay. Cell viability and apoptosis were measured by MTT and flow cytometry, respectively. The protein expression level was analyzed by western blot. Cell migration and invasion were evaluated by wound healing and Transwell assays. A xenograft experiment was established to clarify the functional role of circ-ADAM9 inhibition in vivo. The interactions among circ-ADAM9, miR-383-5p, and PFN2 were analyzed by dual-luciferase reporter, RNA immunoprecipitation (RIP), and RNA pull-down assays. RESULTS: We found that circ-ADAM9 was upregulated in breast cancer tissues and cells compared to controls. Inhibition of circ-ADAM9 expression impaired proliferation, migration, and invasion, but increased radiosensitivity and apoptosis in breast cancer cells; besides, radiotherapy combined with circ-ADAM9 inhibition showed significant inhibitory effects on tumor growth. The functional effects of circ-ADAM9 were related to miR-383-5p, a target of circ-ADAM9. Overexpression of miR-383-5p-mediated malignant behaviors and radiosensitivity of breast cancer cells were dependent on PFN2. CONCLUSION: Circ-ADAM9 was found to participate in breast cancer progression through targeting the miR-383-5p/PFN2 axis.


Asunto(s)
Neoplasias de la Mama , MicroARNs , Humanos , Femenino , Desintegrinas , Neoplasias de la Mama/genética , Neoplasias de la Mama/radioterapia , Mama , Apoptosis/genética , Proliferación Celular/genética , MicroARNs/genética , Línea Celular Tumoral , Proteínas de la Membrana/genética , Proteínas ADAM , Profilinas
7.
Front Cardiovasc Med ; 9: 781753, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35479278

RESUMEN

Cardiovascular diseases (CVD) are the leading cause of death worldwide, wherein myocardial infarction (MI) is the most dangerous one. Promoting angiogenesis is a prospective strategy to alleviate MI. Our previous study indicated that profilin 2 (PFN2) may be a novel target associated with angiogenesis. Further results showed higher levels of serum PFN2 and exosomal PFN2 in patients, mice, and pigs with MI. In this study, we explored whether PFN2 and endothelial cell (EC)-derived exosomal PFN2 could increase angiogenesis and be beneficial for the treatment of MI. Serum PFN2, exosomes, and exosomal PFN2 were elevated in rats with MI. PFN2 and exosomes from PFN2-overexpressing ECs (OE-exo) enhanced EC proliferation, migration, and tube formation ability. OE-exo also significantly increased the vessel number in zebrafish and protected the ECs from inflammatory injury. Moreover, OE-exo-treated mice with MI showed improvement in motor ability, ejection fraction, left ventricular shortening fraction, and left ventricular mass, as well as increased vessel numbers in the MI location, and decreased infarction volume. Mechanistically, PI3K might be the upstream regulator of PFN2, while ERK might be the downstream regulator in the PI3K-PFN2-ERK axis. Taken together, our findings demonstrate that PFN2 and exosomal PFN2 promote EC proliferation, migration, and tube formation through the PI3K-PFN2-ERK axis. Exosomal PFN2 may be a valuable target in the repair of MI injury via angiogenesis.

8.
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
9.
Journal of Chinese Physician ; (12): 1193-1197,1203, 2022.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-956283

RESUMEN

Objective:To explore the effect of miR-3607-3p on the malignant phenotype of cervical cancer cells and related mechanisms.Methods:The OncoLnc bioinformatics website was used to analyze the relationship between the expression of miR-3607-3p and the prognosis of cervical cancer patients. Real time fluorescent quantitative polymerase chain reaction (qRT-PCR) was used to detect the expression of miR-3607-3p in human normal cervical epithelial cells (H8) and cervical cancer cell lines (Hela, HCC94, SiHa, C33A). SiHa cells transfected with negative control nucleotide sequence in vitro were defined as negative control (NC) group, and SiHa cells transfected with miR-3607-3p mimicked sequence was defined as miR-3607-3p group. qRT-PCR was used to detect the expression changes of miR-3607-3p in SiHa cells. Cell counting kit-8 (CCK-8) method and scratch test were used to detect the malignant biological behavior changes of SiHa cells. qRT-PCR and Western blot were used to detect the expression changes of profilin 2 (PFN2) gene, and the dual luciferase reporter gene experiment was used to detect the targeting relationship between miR-3607-3p and PFN2. Results:The survival time of patients with high expression of miR-3607-3p was significantly higher than that of patients with low expression of miR-3607-3p ( P<0.01). Compared with H8 cells, the expression of miR-3607-3p was abnormally low in human cervical cancer cell lines ( P<0.05), and the expression of miR-3607-3p was the lowest in the SiHa cell line ( P<0.01). The expression of miR-3607-3p in the NC group and miR-3607-3p group was (1.04±0.31) and (9.28±1.76), respectively. The expression level of miR-3607-3p in SiHa cells transfected with miR-3607-3p mimic sequence was significantly higher than that in NC group, and the proliferation activity and scratch healing rate were significantly lower than that in NC group (all P<0.01). Dual-luciferase reporter gene assay confirmed that miR-3607-3p directly targeted and binded to PFN2 ( P<0.01). qRT-PCR and Western blot results showed that the expression of PFN2 mRNA and protein in miR-3607-3p group was lower than that in NC group; Western blot results showed the expression of proliferating proteins CDK3 and CDK2 in miR-3607-3p group was lower than that in NC group (all P<0.05), and the expression of epithelial phenotype related proteins Claudin-1 and ZO-1 in miR-3607-3p group was higher than that in NC group (all P<0.05). Conclusions:miR-3607-3p is positively correlated with the survival of cervical cancer patients. Up-regulating the expression of miR-3607-3p can inhibit the proliferation and migration of cervical cancer SiHa cells. The mechanism may be related to the targeted inhibition of PFN2.

10.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-936304

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)
Humanos , Proliferación Celular , Profilinas/metabolismo , Pronóstico , Neoplasias Gástricas/patología , Análisis de Supervivencia
11.
Aging (Albany NY) ; 12(24): 25981-25999, 2020 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-33234737

RESUMEN

Small cell lung cancer (SCLC) is highly aggressive and prone to hypervascular metastases. Recently, we found profilin 2 (PFN2) expression in SCLC but not in normal tissues. Furthermore, PFN2 expression had been shown to promote angiogenesis through exosomes. However, it remains unclear whether PFN2 contributes to the progression and metastasis of SCLC through angiogenesis. We report here that overexpression (OE) of PFN2 increased, whereas its knockdown (KD) decreased the proliferation, migration, and invasion of SCLC cell H446. The exosomes from OE-H446 (SCLC-OE-exo) exhibited similar effects on H446 properties. Culturing of endothelial cells (ECs) in SCLC-OE conditioned medium (CM) or SCLC-OE-exo increased the migration and tube formation ability of ECs, whereas SCLC-KD-CM and SCLC-KD-exo had inhibitory effects. Interestingly, both SCLC- and EC-derived exosomes were internalized in H446 more rapidly than in ECs. More importantly, OE-PFN2 dramatically elevated SCLC growth and vasculature formation as well as lung metastasis in tumor xenograft models. Finally, we found that PFN2 activated Smad2/3 in H446 and pERK in ECs, respectively. Taken together, our study revealed the role of PFN2 in SCLC development and metastasis, as well as tumor angiogenesis through exosomes, providing a new molecular target for SCLC treatment.


Asunto(s)
Exosomas/metabolismo , Neoplasias Pulmonares/genética , Neovascularización Patológica/genética , Profilinas/genética , Carcinoma Pulmonar de Células Pequeñas/genética , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Medios de Cultivo Condicionados , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/patología , Ratones , Ratones SCID , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Carcinoma Pulmonar de Células Pequeñas/irrigación sanguínea , Carcinoma Pulmonar de Células Pequeñas/patología
12.
Int Rev Cell Mol Biol ; 355: 155-204, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32859370

RESUMEN

Actin and microtubules play essential roles in aberrant cell processes that define and converge in cancer including: signaling, morphology, motility, and division. Actin and microtubules do not directly interact, however shared regulators coordinate these polymers. While many of the individual proteins important for regulating and choreographing actin and microtubule behaviors have been identified, the way these molecules collaborate or fail in normal or disease contexts is not fully understood. Decades of research focus on Profilin as a signaling molecule, lipid-binding protein, and canonical regulator of actin assembly. Recent reports demonstrate that Profilin also regulates microtubule dynamics and polymerization. Thus, Profilin can coordinate both actin and microtubule polymer systems. Here we reconsider the biochemical and cellular roles for Profilin with a focus on the essential cytoskeletal-based cell processes that go awry in cancer. We also explore how the use of model organisms has helped to elucidate mechanisms that underlie the regulatory essence of Profilin in vivo and in the context of disease.


Asunto(s)
Citoesqueleto/metabolismo , Neoplasias/metabolismo , Profilinas/metabolismo , Actinas/metabolismo , Animales , Humanos , Microtúbulos/metabolismo
13.
EBioMedicine ; 51: 102583, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31901866

RESUMEN

BACKGROUND: Heterogeneous nuclear ribonucleoprotein (hnRNP) A2/B1 is an important RNA-binding protein that affects the RNA processing, splicing, transport and stability of many genes. hnRNPA2/B1 is expressed during proliferation and metastasis of various cancer types and promotes such processes. However, the precise role and mechanism of hnRNPA2/B1 in breast cancer remain unclear. METHODS: The association of hnRNPA2/B1 with breast cancer metastasis was assessed using tissue chips, mouse models and publicly available data. The role and mechanism of hnRNPA2/B1 in breast cancer metastasis were studied in cell lines and mouse models. FINDINGS: In contrast to other cancer research findings, hnRNPA2/B1 expression was negatively correlated with breast cancer metastasis. hnRNPA2/B1 inhibited MDA-MB-231 triple-negative breast cancer (TNBC) cell metastasis in vitro and in vivo. hnRNPA2/B1 knockout activated ERK-MAPK/Twist and GR-beta/TCF4 pathways but inhibited STAT3 and WNT/TCF4 signalling pathways. Profilin 2 (PFN2) promoted breast cancer cell migration and invasion, whereas hnRNPA2/B1 bound directly to the UAGGG locus in the 3'-untranslated region of PFN2 mRNA and reduced the stability of PFN2 mRNA. INTERPRETATION: Our data supported the role of hnRNPA2/B1 in tumour metastasis risk and survival prediction in patients with breast cancer. The inhibitory role of hnRNPA2/B1 in metastasis was a balance of downstream multiple genes and signalling pathways. PFN2 downregulation by hnRNPA2/B1 might partly explain the inhibitory mechanism of hnRNPA2/B1 in breast cancer metastasis. Therefore, hnRNPA2/B1 might be used as a new prognostic biomarker and valuable molecular target for breast cancer treatments.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Genes Relacionados con las Neoplasias , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Transducción de Señal , Regiones no Traducidas 3'/genética , Actinas/metabolismo , Animales , Neoplasias de la Mama/diagnóstico , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Profilinas/genética , Profilinas/metabolismo , Pronóstico , Estabilidad del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia
14.
Virology ; 533: 108-114, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31150988

RESUMEN

We previously reported that human parainfluenza virus type 2 (hPIV-2) promoted RhoA activation and subsequent filamentous actin (F-actin) formation. Actin-binding proteins, such as profilin and cofilin, are involved in the regulation of F-actin formation by RhoA signaling. In the present study, we identified profilin2 as a key molecule that is involved in hPIV-2-induced F-actin formation. Immunoprecipitation assays demonstrated that hPIV-2 V protein binds to profilin2 but not to profilin1. Mutation of Trp residues within C-terminal region of V protein abolished the binding capacity to profilin2. Depletion of profilin2 resulted in the inhibition of hPIV-2-induced F-actin formation and the suppression of hPIV-2 growth. Overexpression of wild type V but not Trp-mutated V protein reduced the quantity of actin co-immunoprecipitated with profilin2. Taken together, these results suggest that hPIV-2 V protein promotes F-actin formation by affecting actin-profilin2 interaction through its binding to profilin2.


Asunto(s)
Actinas/metabolismo , Virus de la Parainfluenza 2 Humana/metabolismo , Profilinas/metabolismo , Infecciones por Rubulavirus/metabolismo , Infecciones por Rubulavirus/virología , Actinas/genética , Interacciones Huésped-Patógeno , Humanos , Virus de la Parainfluenza 2 Humana/genética , Profilinas/genética , Unión Proteica , Infecciones por Rubulavirus/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
15.
Int J Oncol ; 54(3): 1043-1052, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30628646

RESUMEN

Breast cancer (BC) is a common cancer and leading cause of cancer­associated mortality in women. Abnormal expression of long non­coding RNA FOXD2 adjacent opposite strand RNA 1 (FOXD2­AS1) was associated with the development of a number of tumors. However, whether FOXD2­AS1 is dysregulated in BC and its underlying mechanisms remain unclear. In the present study, it was identified that FOXD2­AS1 expression was upregulated in BC tissue, cell lines and sphere subpopulation. Additionally, the abnormal upregulation of FOXD2­AS1 predicted poor prognosis in patients with BC. Furthermore, downregulation of FOXD2­AS1 decreased cell proliferation, and migratory and invasive abilities in BC cells, and decreased the growth of transplanted tumors in vivo. Downregulation of FOXD2­AS1 decreased the percentage of CD44 antigen+/signal transducer CD24- in breast cancer stem cell (BCSC) cells, and decreased the expression of numerous stem factors, including Nanog, octamer­binding transcription factor 4 (Oct4), and sex determining region Y­box 2 (SOX2), and inhibited the epithelial­mesenchymal transition process. FOXD2­AS1 was identified to be primarily located in the cytoplasm. Using bioinformatics analysis, a reporter gene assay and reverse transcription­polymerase chain reaction assays, it was demonstrated that microRNA (miR)­150­5p was able to bind directly with the 3'­untranslated region of FOXD2­AS1 and PFN2 mRNA. miR­150­5p mimics decreased the cell proliferation, migration and invasion of BC cells. FOXD2­AS1 knockdown significantly inhibited the miR­150­5p inhibitor­induced increase in Nanog, Oct4 and SOX2 expression. The miR­150­5p inhibitor­induced increase in N­cadherin, and decrease in E­cadherin and vimentin was inhibited by FOXD2­AS1 knockdown. Profilin 2 (PFN2) expression was significantly upregulated in BC tissues. Additionally, the abnormal upregulation of PFN2 was associated with poor prognosis in patients with BC. FOXD2­AS1 and PFN2 expression was positively correlated. Collectively, the present results demonstrated the role of the FOXD2­AS1/miR­150­5p/PFN2 axis in the development of BC, and provides novel targets for the treatment of BC, and potential biomarkers for diagnosis and prognosis of BC.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinogénesis/genética , MicroARNs/metabolismo , Profilinas/metabolismo , ARN Largo no Codificante/metabolismo , Regiones no Traducidas 3' , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , Transformación Celular Neoplásica , Citoplasma/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Receptores de Hialuranos/metabolismo , Ratones , MicroARNs/antagonistas & inhibidores , Profilinas/genética , Pronóstico , ARN Largo no Codificante/genética , ARN Mensajero/metabolismo , Células Tumorales Cultivadas
16.
Toxicol Appl Pharmacol ; 355: 68-79, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29802913

RESUMEN

Methamphetamine (MA), a psychostimulant abused worldwide, gives rise to neurotoxicity in the hippocampus, resulting in cognitive impairments and hippocampal volume reduction. The cellular and molecular mechanisms associated with hippocampal impairments due to MA remain unknown. The aim of this study was to investigate the effects of MA on structural alterations and gene expressions in the hippocampus. We analyzed the pattern of volumetric changes in the hippocampus using magnetic resonance imaging (MRI) after acute and chronic administration of MA to cynomolgus macaques. In addition, we performed large-scale transcriptome profiling in the hippocampus using RNA-Seq technology. The hippocampus in response to acute and chronic MA exhibited a significant volumetric atrophy compared with the hippocampus of controls. The genes associated with cytoskeleton organization and phagocytosis were downregulated in the acute MA-treated group compared to the control group. On the other hand, genes associated with synaptic transmission, regulation of neuron differentiation and regulation of neurogenesis were downregulated in the chronic MA-treated group. We confirmed that expression patterns for ADM, BMP4, CHRD, PDYN, UBA1, profilin 2 (PFN2), ENO2 and NSE mRNAs were similar to the results from RNA-Seq based on quantitative RT-PCR. In particular, PFN2 mRNA and protein expression levels, which play important roles in actin cytoskeleton dynamics, were decreased by acute and chronic MA administration. These results not only aid the understanding of cellular and molecular mechanisms regulated by MA in the hippocampus but also suggest basic information aiding biomarker and novel drug development for treating hippocampal impairment caused by MA abuse.


Asunto(s)
Estimulantes del Sistema Nervioso Central/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Metanfetamina/toxicidad , Transcriptoma/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/ultraestructura , Ingestión de Alimentos/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Hipocampo/diagnóstico por imagen , Macaca fascicularis , Imagen por Resonancia Magnética , Neurogénesis/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
17.
J Transl Med ; 14(1): 137, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27188458

RESUMEN

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is one of the most aggressively malignant tumors with dismal prognosis. Profilin 2 (PFN2) is an actin-binding protein that regulates the dynamics of actin polymerization and plays a key role in cell motility. Recently, PFN2 have emerged as significant regulators of cancer processes. However, the clinical significance and biological function of PFN2 in ESCC remain unclear. METHODS: PFN2 protein expression was validated by immunohistochemistry (IHC) on tissue microarray from Chinese Han and Kazakh populations with ESCC. The associations among PFN2 expression, clinicopathological features, and prognosis of ESCC were analyzed. The effects on cell proliferation, invasion and migration were examined using MTT and Transwell assays. Markers of epithelial-mesenchymal transition (EMT) were detected by Western blot analysis. RESULTS: Compared with normal esophageal epithelium (NEE), PFN2 protein expression was markedly increased in low-grade intraepithelial neoplasia (LGIN), high-grade intraepithelial neoplasia (HGIN), and ESCC, increased gradually from LGIN to ESCC, and finally reached high grade in HGIN in the Han population. Similarly, PFN2 protein was more overexpressed in ESCC than in NEE in the Kazakh population. The results of Western blot analysis also showed that PFN2 expression was significantly higher in the ESCC tissue than in a matched adjacent non-cancerous tissue. PFN2 expression was positively correlated with invasion depth and lymph node metastasis. High PFN2 expression was significantly correlated with short overall survival (OS) (P = 0.023). Cox regression analysis revealed that PFN2 expression was an independent prognostic factor for poor OS in ESCC. Downregulation of PFN2 inhibited, rather than proliferated, cell invasion and migration, as well as induced an EMT phenotype, including increased expression of epithelial marker E-cadherin, decreased mesenchymal marker Vimentin, Snail, Slug and ZEB1, and morphological changes in ESCC cells in vitro. CONCLUSIONS: Our findings demonstrate that PFN2 has a novel role in promoting ESCC progression and metastasis and portending a poor prognosis, indicating that PFN2 could act as an early biomarker of high-risk population. Targeting PFN2 may offer a promising therapeutic strategy for ESCC treatment.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Terapia Molecular Dirigida , Profilinas/metabolismo , Adulto , Anciano , Pueblo Asiatico , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patología , Línea Celular Tumoral , Movimiento Celular , Forma de la Célula , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Epitelio/metabolismo , Epitelio/patología , Carcinoma de Células Escamosas de Esófago , Etnicidad , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Metástasis Linfática , Persona de Mediana Edad , Invasividad Neoplásica , Fenotipo , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Pronóstico , Modelos de Riesgos Proporcionales , ARN Interferente Pequeño/metabolismo , Curva ROC , Transfección , Regulación hacia Arriba
18.
Biosci Biotechnol Biochem ; 79(9): 1438-46, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25964982

RESUMEN

We investigated the role of profilin 2 in the stemness, migration, and invasion of HT29 cancer stem cells (CSCs). Increased and decreased levels of profilin 2 significantly enhanced and suppressed the self-renewal, migration, and invasion ability of HT29 CSCs, respectively. Moreover, profilin 2 directly regulated the expression of stemness markers (CD133, SOX2, and ß-catenin) and epithelial mesenchymal transition (EMT) markers (E-cadherin and snail). CD133 and ß-catenin were up-regulated by overexpression of profilin 2 and down-regulated by depletion of profilin 2. SOX2 was decreased by profilin 2 depletion. E-cadherin was not influenced by profilin 2- overexpression but increased by profilin 2- knockdown. The expression of snail was suppressed by profilin 2- knockdown. We speculated that stemness and the EMT are closely linked through profilin 2-related pathways. Therefore, this study indicates that profilin 2 affects the metastatic potential and stemness of colorectal CSCs by regulating EMT- and stemness-related proteins.


Asunto(s)
Neoplasias Colorrectales/genética , Proteínas de Neoplasias/biosíntesis , Células Madre Neoplásicas , Profilinas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HT29 , Humanos , Invasividad Neoplásica/genética , Profilinas/biosíntesis , Vía de Señalización Wnt/genética , beta Catenina/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA