Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Ann Vasc Surg ; 105: 227-235, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38609009

RESUMEN

BACKGROUND: Studies have linked matrix metalloproteinases (MMPs) to both thoracic aortic aneurysm and abdominal aortic aneurysm (TAA and AAA). The precise MMPs entailed in this procedure, however, were still unknown. This study used a two-sample Mendelian randomization (MR) analysis to look into the causal relationship between MMPs and the risk of TAA and AAA. METHODS: Eight MMPs, including MMP-1, MMP-2, MMP-3, MMP-8, MMP-9, MMP-10, MMP-12, and MMP-13, were found among people of European ancestry with accessible Genome-Wide Association Studies (GWAS). We employed the findings from Genome-Wide Association Studies (GWAS) for 8 MMPs, and TAA and AAA from the FinnGen consortiums (3,201 cases and 317,899 controls, respectively) were used in a two-sample MR analysis. The primary method of analysis for MR was the inverse variance weighted (IVW) method, along with analyses of heterogeneity and horizontal pleiotropy. 31 single-nucleotide polymorphisms connected to MMP were retrieved. RESULTS: IVW demonstrated a negative causal association between TAA and AAA and serum MMP-12 levels. The incidence of TAA decreased by 1.031% for every 1 ng/mL increase in serum MMP-12 [odds ratio (OR) = 0.897, 95% confidence interval (CI): 0.831-0.968, P = 0.005]. The incidence of AAA fell by 1.653% (OR = 0.835, 95% CI: 0.752-0.926, P = 0.001) for every 1 ng/mL increase in serum MMP-12. There was no horizontal pleiotropy or heterogeneity in the MR data (P > 0.05). CONCLUSIONS: The levels of TAA and AAA and serum MMP-12 are causally related. MMP-12 is a factor that reduces the risk of AAA and TTA. Our study suggested that MMP-12 level is causally associated with a decreased risk of TAA and AAA.


Asunto(s)
Aneurisma de la Aorta Abdominal , Aneurisma de la Aorta Torácica , Metaloproteinasas de la Matriz , Humanos , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/enzimología , Aneurisma de la Aorta Abdominal/diagnóstico por imagen , Aneurisma de la Aorta Abdominal/sangre , Aneurisma de la Aorta Abdominal/epidemiología , Aneurisma de la Aorta Torácica/genética , Aneurisma de la Aorta Torácica/enzimología , Aneurisma de la Aorta Torácica/sangre , Aneurisma de la Aorta Torácica/diagnóstico por imagen , Aneurisma de la Aorta Torácica/epidemiología , Estudios de Casos y Controles , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Incidencia , Metaloproteinasa 12 de la Matriz/genética , Metaloproteinasa 12 de la Matriz/sangre , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/sangre , Análisis de la Aleatorización Mendeliana , Fenotipo , Polimorfismo de Nucleótido Simple , Factores Protectores , Medición de Riesgo , Factores de Riesgo
2.
Bioelectricity ; 6(1): 65-68, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38525485

RESUMEN

Cell Bio conferences-organized jointly by the American Society of Cell Biology (ASCB) and European Molecular Biology Organization (EMBO)-showcase a diverse global community of the brightest researchers in Cell Biology and in emerging interdisciplinary topics, including bioelectricity. In this report, we briefly overview the Cell Bio 2023 subgroup meeting "Bioelectricity in Development, Regeneration, and Cancers." This subgroup meeting featured 12 talks (7 Principal Investigators and 5 junior scientists) exploring the role of bioelectricity in endogenous and diseased states in model systems ranging from cells in culture to single-cell organisms such as yeast all the way to mammalian systems (including tools and technology developed for exploring bioelectricity and electrotaxis in cells and tissues). The subgroup meeting concluded with a discussion on the current challenges and opportunities for the field of bioelectricity.

3.
Dev Dyn ; 253(8): 722-749, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38270285

RESUMEN

BACKGROUND: The two-pore domain potassium (K2P) channels are a major type of potassium channels that maintain the cell membrane potential by conducting passive potassium leak currents independent of voltage change. They play prominent roles in multiple physiological processes, including neuromodulation, perception of pain, breathing and mood control, and response to volatile anesthetics. Mutations in K2P channels have been linked to many human diseases, such as neuronal and cardiovascular disorders and cancers. Significant progress has been made to understand their protein structures, physiological functions, and pharmacological modifiers. However, their expression and function during embryonic development remain largely unknown. RESULTS: We employed the zebrafish model and identified 23 k2p genes using BLAST search and gene cloning. We first analyzed vertebrate K2P channel evolution by phylogenetic and syntenic analyses. Our data revealed that the six subtypes of the K2P genes have already evolved in invertebrates long before the emergence of vertebrates. Moreover, the vertebrate K2P gene number increased, most likely due to two whole-genome duplications. Furthermore, we examined zebrafish k2p gene expression during early embryogenesis by in situ hybridization. Each subgroup's genes showed similar but distinct gene expression domains with some exceptions. Most of them were expressed in neural tissues consistent with their known function of neural excitability regulation. However, a few k2p genes were expressed temporarily in specific tissues or organs, suggesting that these K2P channels may be needed for embryonic development. CONCLUSIONS: Our phylogenetic and developmental analyses of K2P channels shed light on their evolutionary history and potential roles during embryogenesis related to their physiological functions and human channelopathies.


Asunto(s)
Evolución Molecular , Filogenia , Canales de Potasio de Dominio Poro en Tándem , Pez Cebra , Animales , Pez Cebra/genética , Pez Cebra/embriología , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Regulación del Desarrollo de la Expresión Génica , Embrión no Mamífero/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Desarrollo Embrionario/genética
4.
Front Immunol ; 14: 1305937, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38077380

RESUMEN

Introduction: Nonhuman adenoviral (AdV) gene delivery platforms have significant value due to their ability to elude preexisting AdV vector immunity in most individuals. Previously, we have demonstrated that intranasal (IN) immunization of mice with BAd-H5HA, a bovine AdV type 3 (BAdV3) vector expressing H5N1 influenza virus hemagglutinin (HA), resulted in enhanced humoral and cell-mediated immune responses. The BAd-H5HA IN immunization resulted in complete protection following the challenge with an antigenically distinct H5N1 virus compared to the mouse group similarly immunized with HAd-H5HA, a human AdV type 5 (HAdV5) vector expressing HA. Methods: Here, we attempted to determine the activation of innate immune responses in the lungs of mice inoculated intranasally with BAd-H5HA compared to the HAd-H5HA-inoculated group. Results: RNA-Seq analyses of the lung tissues revealed differential expression (DE) of genes involved in innate and adaptive immunity in animals immunized with BAd-H5HA. The top ten enhanced genes were verified by RT-PCR. Consistently, there were transient increases in the levels of cytokines (IL-1α, IL-1ß, IL-5, TNF- α, LIF, IL-17, G-CSF, MIP-1ß, MCP-1, MIP-2, and GM-CSF) and toll-like receptors in the lungs of the group inoculated with BAdV vectors compared to that of the HAdV vector group. Conclusion: These results demonstrate that the BAdV vectors induce enhanced innate and adaptive immunity-related factors compared to HAdV vectors in mice. Thus, the BAdV vector platform could be an excellent gene delivery system for recombinant vaccines and cancer immunotherapy.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Vacunas contra la Influenza , Animales , Bovinos , Ratones , Humanos , Inmunización , Inmunidad Adaptativa , Vacunación , Hemaglutininas
5.
PLoS One ; 18(11): e0294249, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37948436

RESUMEN

The estrogen inhibits colonic smooth muscle contractions, which may lead to constipation. However, the mechanisms of inhibition are poorly understood. Therefore, the present study examined the effect of estrogen on rat colonic smooth muscle contractions and its potential association with the large-conductance Ca2+-activated K+ channels ß1 (BKß1) subunit. Twenty-four female Sprague Dawley rats were randomly assigned to 4 groups. After 2 weeks of intervention, the contraction activity of isolated colonic smooth muscle and the expression of BKß1 in colonic smooth muscle of rats were detected. Additionally, in order to investigate the effects of estrogen on BKß1 expression and calcium mobilization, in vitro experiments were conducted using rat and human colonic smooth muscle cells (SMCs). BKß1 shRNA was used to investigate whether calcium mobilization is affected by BKß1 in colonic SMCs. To explore the relationship between ERß and BKß1, serial deletions, site-directed mutagenesis, a dual-luciferase reporter assay, and chromatin immunoprecipitation assays were employed. In response to E2, colonic smooth muscle strips showed a decrease in tension, while IBTX exposure transiently increased tension. Furthermore, in these muscle tissues, BKß1 and α-SMA were found to be co-expressed. The E2 group showed significantly higher BKß1 expression. In cultured colonic SMCs, the expression of BKß1 was found to increase in the presence of E2 or DPN. E2 treatment reduced Ca2+ concentrations, while BKß1 shRNA treatment increased Ca2+ concentrations relative to the control. ERß-initiated BKß1 expression appears to occur via binding to the BKß1 promoter. These results indicated that E2 may upregulate BKß1 expression via ERß and inhibit colonic smooth muscle contraction through ERß by directly targeting BKß1.


Asunto(s)
Calcio , Receptor beta de Estrógeno , Humanos , Ratas , Femenino , Animales , Calcio/metabolismo , Ratas Sprague-Dawley , Receptor beta de Estrógeno/genética , Estrógenos/farmacología , Contracción Muscular , ARN Interferente Pequeño/farmacología
6.
Cells ; 12(8)2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-37190057

RESUMEN

Developmental patterning is essential for regulating cellular events such as axial patterning, segmentation, tissue formation, and organ size determination during embryogenesis. Understanding the patterning mechanisms remains a central challenge and fundamental interest in developmental biology. Ion-channel-regulated bioelectric signals have emerged as a player of the patterning mechanism, which may interact with morphogens. Evidence from multiple model organisms reveals the roles of bioelectricity in embryonic development, regeneration, and cancers. The Zebrafish model is the second most used vertebrate model, next to the mouse model. The zebrafish model has great potential for elucidating the functions of bioelectricity due to many advantages such as external development, transparent early embryogenesis, and tractable genetics. Here, we review genetic evidence from zebrafish mutants with fin-size and pigment changes related to ion channels and bioelectricity. In addition, we review the cell membrane voltage reporting and chemogenetic tools that have already been used or have great potential to be implemented in zebrafish models. Finally, new perspectives and opportunities for bioelectricity research with zebrafish are discussed.


Asunto(s)
Fenómenos Electrofisiológicos , Pez Cebra , Animales , Ratones , Pez Cebra/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Membrana Celular/metabolismo , Canales Iónicos/genética
7.
Materials (Basel) ; 15(24)2022 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-36556848

RESUMEN

The microstructure and mechanical properties of the welded joints of 6252 armor steel and Q550D high-strength low-alloy (HSLA) steel welded by MIG welding were studied. ER70S-G and ER140S-G were used as fillers to obtain welded joints with good formation and no faults. The joint microstructure (OM) analysis showed that a large Widmanstätten structure was observed at the fusion line on the Q550D side, and the apparent grain sizes changed on the 6252 side. Cylindrical ferrite growth along the bainite grain boundary was observed in the ER70S-G filler weld zone, while the ER140S-G filler weld zone was occupied by lower bainite structures. The XRD phase analysis showed that more Fe-Ni-Cr compounds and less ferrite were formed in the ER140S-G filler weld. The hardness test showed that the hardness of the HAZ on the 6252 side was significantly higher than that of the BM and the WZ, and the welded joint obtained by the ER140S-G filler had a higher hardness. The tensile strength test showed that WZ (>772 MPa) had a higher strength than Q550D BM, and the tensile fracture (SEM) was primarily a ductile fracture. The impact test results showed that the welded joint had better impact resistance at room temperature, but the impact absorption energy of the weld and the heat-affected zone was strongly affected by changes in temperature, and brittle fracture occurred easily at low temperatures.

8.
Dis Markers ; 2022: 7843565, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36061355

RESUMEN

Colorectal cancer (CRC) is one of the most frequent gastrointestinal cancers. MicroRNAs (miRNAs) have been proved to be unusually expressed in CRC progression and thus alter multiple pathological processes in CRC cells. However, the specific roles and mechanisms of miR-22 in CRC have not been clearly reported. MicroRNA-22 (miR-22) and MYC-associated factor X (MAX) expressions were determined by RT-qPCR in CRC tissues and cells. The targeted regulatory effects of miR-22 and MAX were confirmed by luciferase reporter and coimmunoprecipitation assays. Also, gain- and loss-of-function and rescue experiments were used to elucidate the function and mechanism of miR-22 and MAX in CRC cells and the mouse xenograft model. We discovered that miR-22 was hypermethylated and downregulated, while MAX was upregulated in CRC. miR-22 markedly inhibited migration, invasion, glycolysis, and cancer stem cell transcription factors in CRC cells. In addition, it was found that miR-22 can directly target MAX. Additional functional experiments confirmed that MAX overexpression can rescue the effects of miR-22 on the behavior of CRC cells. This study suggested that miR-22, as a cancer suppressor, participates in CRC progression by targeting MAX, which might provide basic information for therapeutic targets for CRC.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Neoplasias Colorrectales , Transición Epitelial-Mesenquimal , MicroARNs , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , MicroARNs/genética
9.
Oncol Rep ; 47(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35029289

RESUMEN

Zinc finger proteins (ZNFs) serve key roles in tumor formation and progression; however, the functions and underlying mechanisms of dysregulated ZNF384 in colorectal cancer (CRC) are yet to be fully elucidated. Therefore, the present study initially aimed to investigate the expression levels of ZNF384 in CRC samples. Moreover, lentiviral ZNF384 overexpression and ZNF384 knockdown models were established in CRC cells. Transwell, wound healing and in vivo tail vein metastasis assays were carried out to evaluate the effects of ZNF384 on CRC cell metastasis. Furthermore, reverse transcription­quantitative PCR, western blotting, serial deletion, site­directed mutagenesis, dual­luciferase reporter and chromatin immunoprecipitation assays were conducted to investigate the potential underlying mechanisms. The results of the present study demonstrated that ZNF384 expression was markedly increased in CRC samples and this was associated with a poor prognosis. Notably, ZNF384 overexpression increased the levels of CRC cell invasion and migration, whereas ZNF384 knockdown inhibited CRC development. Moreover, the results of the present study demonstrated that ZNF384 mediated the expression of MMP2. MMP2 knockdown inhibited ZNF384­mediated CRC cell invasion and migration, whereas MMP2 overexpression ameliorated ZNF384 knockdown­induced inhibition of CRC progression. In addition, the results of the present study demonstrated that hypoxia­inducible factor 1α (HIF­1α) had the ability to bind to the ZNF384 promoter, thereby initiating ZNF384 expression. In human­derived CRC samples, the expression levels of ZNF384 were positively correlated with both MMP2 and HIF­1α expression. Collectively, these findings highlighted that ZNF384 may act as a prognostic marker and regulator of CRC metastasis.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Transactivadores/genética , Dedos de Zinc/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica , Regulación hacia Arriba
10.
Oncol Lett ; 22(6): 812, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34671426

RESUMEN

Colorectal cancer (CRC) is one of the most common types of malignancy worldwide and has a poor prognosis. Non-SMC condensing I complex subunit G (NCAPG) has been reported to be upregulated in numerous types of malignant tumor. However, to the best of our knowledge, its clinicopathological and biological significance in CRC remain to be elucidated. The results of the present study revealed that NCAPG expression levels were upregulated in human CRC tissues and cell lines. The upregulated expression of NCAPG was positively associated with patient clinicopathological characteristics, such as differentiation and tumor size, and independently associated with poor survival. Consistent with the clinical observations, NCAPG was discovered to promote the proliferation and inhibit the apoptosis of CRC cells. Moreover, NCAPG-knockdown inhibited CRC cell proliferation by regulating the PI3K/AKT signaling pathway. Furthermore, NCAPG was identified as a potential target of microRNA (miR)-23b-3p, which was subsequently demonstrated to negatively regulate NCAPG expression. In conclusion, the findings of the current study indicated that the miR-23b-3p/NCAPG/PI3K/AKT signaling axis may play an important role in CRC carcinogenesis, and the status of the molecule may represent a promising prognostic marker for the disease.

11.
Mol Med Rep ; 24(4)2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34368871

RESUMEN

N­methyl D­aspartate receptors (NMDARs) are closely associated with the development, growth and metastasis of cancer. Glutamate receptor, ionotropic, N­methyl D­aspartate­associated protein 1 (GRINA) is a member of the of the NMDAR family, and its aberrant expression is associated with gastric cancer. However, the role of GRINA in colorectal cancer (CRC) is not completely understood. In the present study, expression profiles of GRINA in several CRC databases were obtained and further verified using clinical CRC samples. The effects of GRINA overexpression on CRC progression both in vivo and in vitro were assessed. Briefly, cell proliferation was detected using MTT assay, and cell migration and invasion ability were evaluated by wound healing and Transwell assay. In addition, the molecular mechanism underlying the upregulated expression of GRINA in CRC was investigated. The regulatory association between GRINA and miR­296­3p was detected by luciferase assay, reverse transcription­quantitative PCR and western blotting. The results demonstrated that GRINA expression levels were significantly increased in tumor samples compared with those in healthy samples, and upregulated expression of GRINA was associated with a less favorable prognostic outcome in patients with CRC. GRINA overexpression significantly increased CRC cell proliferation, invasion and migration. Additionally, it was determined that GRINA was post­transcriptionally regulated by microRNA (miR)­296­3p. Together, the results of the present study suggested the potential importance of the miR­296­3p/GRINA axis and highlighted potential novel targets for the management of CRC.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Ácido D-Aspártico/metabolismo , MicroARNs/metabolismo , Receptores de Glutamato/metabolismo , Receptores Ionotrópicos de Glutamato/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Colorrectales/patología , Ácido D-Aspártico/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Persona de Mediana Edad , Pronóstico , Receptores Ionotrópicos de Glutamato/genética , Receptores de N-Metil-D-Aspartato/genética , Neoplasias Gástricas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Genes Chromosomes Cancer ; 60(11): 743-761, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34296799

RESUMEN

Malignant peripheral nerve sheath tumors (MPNSTs) are a type of sarcoma that generally originates from Schwann cells. The prognosis for this type of malignancy is relatively poor due to complicated genetic alterations and the lack of specific targeted therapy. Chromosome fragment 4q22-23 is frequently deleted in MPNSTs and other human tumors, suggesting tumor suppressor genes may reside in this region. Here, we provide evidence that SMARCAD1, a known chromatin remodeler, is a novel tumor suppressor gene located in 4q22-23. We identified two human homologous smarcad1 genes (smarcad1a and smarcad1b) in zebrafish, and both genes share overlapping expression patterns during embryonic development. We demonstrated that two smarcad1a loss-of-function mutants, sa1299 and p403, can accelerate MPNST tumorigenesis in the tp53 mutant background, suggesting smarcad1a is a bona fide tumor suppressor gene for MPNSTs. Moreover, we found that DNA double-strand break (DSB) repair might be compromised in both mutants compared to wildtype zebrafish, as indicated by pH2AX, a DNA DSB marker. In addition, both SMARCAD1 gene knockdown and overexpression in human cells were able to inhibit tumor growth and displayed similar DSB repair responses, suggesting proper SMARCAD1 gene expression level or gene dosage is critical for cell growth. Given that mutations of SMARCAD1 sensitize cells to poly ADP ribose polymerase inhibitors in yeast and the human U2OS osteosarcoma cell line, the identification of SMARCAD1 as a novel tumor suppressor gene might contribute to the development of new cancer therapies for MPNSTs.


Asunto(s)
Carcinogénesis , Neurofibrosarcoma , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Neurofibrosarcoma/genética , Neurofibrosarcoma/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Pez Cebra
13.
FASEB Bioadv ; 3(6): 459-469, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34124601

RESUMEN

Vismodegib, a Smoothened antagonist, is clinically approved for treatment of human basal cell carcinoma (BCC), in the clinical trials of medulloblastoma (MB) and other cancers. However, a significant proportion of these tumors fail to respond to Vismodegib after a period of treatment. Here, we find that AMPK agonists, A769662, and Metformin, can inhibit GLI1 activity and synergize with Vismodegib to suppress MB cell growth in vitro and in vivo. Furthermore, combination of AMPK agonists with Vismodegib is effective in overcoming Vismodegib-resistant MB. This is the first report demonstrating that combining AMPK agonist (Metformin) and SHH pathway inhibitor (Vismodegib) confers synergy for MB treatment and provides an effective chemotherapeutic regimen that can be used to overcome resistance to Vismodegib in SHH-driven cancers.

14.
Dev Dyn ; 250(10): 1477-1493, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33728688

RESUMEN

BACKGROUND: Calcium-activated potassium channels (KCa) are a specific type of potassium channel activated by intracellular calcium concentration changes. This group of potassium channels plays fundamental roles ranging from regulating neuronal excitability to immune cell activation. Many human diseases such as schizophrenia, hypertension, epilepsy, and cancers have been linked to mutations in this group of potassium channels. Although the KCa channels have been extensively studied electrophysiologically and pharmacologically, their spatiotemporal gene expression during embryogenesis remains mostly unknown. RESULTS: Using zebrafish as a model, we identified and renamed 14 KCa genes. We further performed phylogenetic and syntenic analyses on vertebrate KCa genes. Our data revealed that the number of KCa genes in zebrafish was increased, most likely due to teleost-specific whole-genome duplication. Moreover, we examined zebrafish KCa gene expression during early embryogenesis. The duplicated ohnologous genes show distinct and overlapped gene expression. Furthermore, we found that zebrafish KCa genes are expressed in various tissues and organs (somites, fins, olfactory regions, eye, kidney, and so on) and neuronal tissues, suggesting that they may play important roles during zebrafish embryogenesis. CONCLUSIONS: Our phylogenetic and developmental analyses shed light on the potential functions of the KCa genes during embryogenesis related to congenital diseases and human channelopathies.


Asunto(s)
Desarrollo Embrionario/fisiología , Filogenia , Canales de Potasio Calcio-Activados/metabolismo , Pez Cebra/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Canales de Potasio Calcio-Activados/genética , Somitos/metabolismo , Pez Cebra/genética
15.
Hum Cell ; 34(1): 132-151, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32946066

RESUMEN

Colorectal cancer (CRC) is the third leading cause of cancer-related death around the world. In this study, we investigated the roles of LncRNA RP11-462C24.1 in CRC. The expressions of RP11-462C24.1 in CRC tissues and cells were measured. Then, the effects of RP11-462C24.1 on CRC proliferation, cell cycle, apoptosis, and invasion were evaluated both in vivo and in vitro; Last, the underlying mechanisms of concerning the signaling pathway regulated by RP11-462C24.1 was determined. From the results, we found that RP11-462C24.1 was significantly decreased in CRC tumor tissues and the CRC cell lines, which were most significant in SW480 and HT-29 cell lines; moreover, transient overexpression of RP11-462C24.1 suppressed the growth and migration while promoted apoptosis of SW480 and HT-29 cells, while knockdown of RP11-462C24.1 has shown the opposite effects; RP11-462C24.1 may also inhibit the growth of CRC tumors in xenograft mice models; additionally, 70 kD heat shock proteins (HSP70) has been identified as one of the most significantly deferentially expressed genes by RNA-seq, and we further confirmed that RP11-462C24.1 may affect the growth and metathesis of CRC cells via regulating HSP70 and PI3K/AKT signaling pathway. In summary, these results indicated that RP11-462C24 may function as a tumor suppressor in the development of CRC.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica/genética , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Invasividad Neoplásica/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/fisiología , Transducción de Señal/genética , Línea Celular Tumoral , Proliferación Celular , Genes Supresores de Tumor , Humanos , Transducción de Señal/fisiología
16.
Onco Targets Ther ; 13: 11505-11516, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33204105

RESUMEN

PURPOSE: Colorectal cancer (CRC) is one of the most common malignancies in the world. The prognosis of advanced CRC is still poor. The purpose of this study was to identify a gene expression profile associated with CRC that may contribute to the early diagnosis of CRC and improve patient prognosis. PATIENTS AND METHODS: Five pairs of CRC tissues and paracancerous tissues were used to identify causative genes using microarray assays. The prognostic value of Cytochrome C Oxidase Assembly Factor 1 Homolog (COA1) in CRC was assessed in 90 CRC patients. Loss-of-function assays, cell proliferation assays using Celigo and MTT, colony formation assays, a subcutaneous xenograft mouse model, and apoptosis assays were used to define the effects of downregulation of COA1 in CRC cells in vitro and in vivo. The underlying molecular mechanisms of COA1 in CRC were also investigated. RESULTS: The causative gene COA1 was identified through microarray analysis. COA1 expression in CRC was notably associated with pathologic differentiation, tumor size, and tumor depth. COA1 expression may act as an independent prognostic factor for overall survival of CRC. Knockdown of COA1 inhibited the proliferation of CRC cells in vitro and the tumorigenicity of CRC cells in vivo. Decreased COA1 expression induced apoptosis of CRC cells. Based on the microarray assay results comparing HCT116 cells transfected with lentivirus encoding anti-COA1 shRNA or negative control shRNA, ingenuity pathway analysis (IPA) revealed that the PI3K/AKT signaling pathway was significantly enriched. Moreover, CCND1, mTOR, AKT1, and MDM2 were identified as the downstream genes of COA1. CONCLUSION: These findings demonstrate that COA1 promotes CRC cell proliferation and inhibits apoptosis by regulating the PI3K/AKT signaling pathway. Our results implicate COA1 as a potential oncogene involved in tumor growth and progression of CRC.

17.
Comput Math Methods Med ; 2020: 5479279, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32908579

RESUMEN

BACKGROUND: Gastric cancer (GC) is one of the most common malignant tumors in the digestive system with high mortality globally. However, the biomarkers that accurately predict the prognosis are still lacking. Therefore, it is important to screen for novel prognostic markers and therapeutic targets. METHODS: We conducted differential expression analysis and survival analysis to screen out the prognostic genes. A stepwise method was employed to select a subset of genes in the multivariable Cox model. Overrepresentation enrichment analysis (ORA) was used to search for the pathways associated with poor prognosis. RESULTS: In this study, we designed a seven-gene-signature-based Cox model to stratify the GC samples into high-risk and low-risk groups. The survival analysis revealed that the high-risk and low-risk groups exhibited significantly different prognostic outcomes in both the training and validation datasets. Specifically, CGB5, IGFBP1, OLFML2B, RAI14, SERPINE1, IQSEC2, and MPND were selected by the multivariable Cox model. Functionally, PI3K-Akt signaling pathway and platelet-derived growth factor receptor (PDGFR) were found to be hyperactive in the high-risk group. The multivariable Cox regression analysis revealed that the risk stratification based on the seven-gene-signature-based Cox model was independent of other prognostic factors such as TNM stages, age, and gender. CONCLUSION: In conclusion, we aimed at developing a model to predict the prognosis of gastric cancer. The predictive model could not only effectively predict the risk of GC but also be beneficial to the development of therapeutic strategies.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Gástricas/genética , Biología Computacional , Bases de Datos Genéticas , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Masculino , Conceptos Matemáticos , Pronóstico , Modelos de Riesgos Proporcionales , Factores de Riesgo , Neoplasias Gástricas/mortalidad
18.
Biosci Rep ; 40(7)2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32597491

RESUMEN

BACKGROUND: The minichromosome maintenance (MCM) family, a core component of DNA replication, is involved in cell cycle process. Abnormal proliferation has been identified as a crucial process in the evolution of colorectal cancer (CRC). However, the roles of the MCM family in CRC remain largely unknown. METHODS: Here, the expression, prognostic significance and functions of the MCM family in CRC were systematically analyzed through a series of online databases including CCLE, Oncomine, HPA, cBioPortal and cancerSEA. RESULTS: We found all MCM family members were highly expressed in CRC, but only elevation of MCM3 expression was associated with poor prognosis of patients with CRC. Further in vitro and in vivo experiments were performed to examine the role of MCM3 in CRC. Analysis of CCLE database and qRT-PCR assay confirmed that MCM3 was overexpressed in CRC cell lines. Moreover, knockdown of MCM3 significantly suppressed transition of G1 to S phase in CRC cells. Furthermore, down-regulation of MCM3 inhibited CRC cell proliferation, migration, invasion and promoted apoptosis. CONCLUSION: These findings reveal that MCM3 may function as an oncogene and a potential prognosis biomarker. Thus, the association between abnormal expression of MCM3 and the initiation of CRC deserves further exploration.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Componente 3 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Animales , Biomarcadores de Tumor/genética , Carcinogénesis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Colon/patología , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Conjuntos de Datos como Asunto , Progresión de la Enfermedad , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Técnicas de Silenciamiento del Gen , Humanos , Estimación de Kaplan-Meier , Ratones , Componente 3 del Complejo de Mantenimiento de Minicromosoma/genética , Invasividad Neoplásica/genética , Oncogenes , Pronóstico , Supervivencia sin Progresión , Recto/patología , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Genetics ; 215(4): 1067-1084, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32546498

RESUMEN

The roles of bioelectric signaling in developmental patterning remain largely unknown, although recent work has implicated bioelectric signals in cellular processes such as proliferation and migration. Here, we report a mutation in the inwardly rectifying potassium channel (kir) gene, kcnj13/kir7.1, that causes elongation of the fins in the zebrafish insertional mutant Dhi2059. A viral DNA insertion into the noncoding region of kcnj13 results in transient activation and ectopic expression of kcnj13 in the somite and dermomyotome, from which the fin ray progenitors originate. We made an allele-specific loss-of-function kcnj13 mutant by CRISPR (clustered regularly interspaced short palindromic repeats) and showed that it could reverse the long-finned phenotype, but only when located on the same chromosome as the Dhi2059 viral insertion. Also, we showed that ectopic expression of kcnj13 in the dermomyotome of transgenic zebrafish produces phenocopies of the Dhi2059 mutant in a gene dosage-sensitive manner. Finally, to determine whether this developmental function is specific to kcnj13, we ectopically expressed three additional potassium channel genes: kcnj1b, kcnj10a, and kcnk9 We found that all induce the long-finned phenotype, indicating that this function is conserved among potassium channel genes. Taken together, our results suggest that dermomyotome bioelectricity is a new fin-patterning mechanism, and we propose a two-stage bioelectricity model for zebrafish fin patterning. This ion channel-regulated bioelectric developmental patterning mechanism may provide with us new insight into vertebrate morphological evolution and human congenital malformations.


Asunto(s)
Aletas de Animales/fisiología , Animales Modificados Genéticamente/fisiología , Tipificación del Cuerpo , Electricidad , Regulación de la Expresión Génica , Canales de Potasio/metabolismo , Pez Cebra/fisiología , Animales , Fuentes de Energía Bioeléctrica , Células Epiteliales/metabolismo , Músculos/metabolismo , Canales de Potasio/genética , Somitos/metabolismo
20.
Cancers (Basel) ; 12(2)2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32053966

RESUMEN

The mechanisms that drive ductal carcinoma in situ (DCIS) progression to invasive cancer are not clear. Studying DCIS progression in humans is challenging and not ethical, thus necessitating the characterization of an animal model that faithfully resembles human disease. We have characterized a canine model of spontaneous mammary DCIS and invasive cancer that shares histologic, molecular, and diagnostic imaging characteristics with DCIS and invasive cancer in women. The purpose of the study was to identify markers and altered signaling pathways that lead to invasive cancer and shed light on early molecular events in breast cancer progression and development. Transcriptomic studies along the continuum of cancer progression in the mammary gland from healthy, through atypical ductal hyperplasia (ADH), DCIS, and invasive carcinoma were performed using the canine model. Gene expression profiles of preinvasive DCIS lesions closely resemble those of invasive carcinoma. However, certain genes, such as SFRP2, FZD2, STK31, and LALBA, were over-expressed in DCIS compared to invasive cancer. The over-representation of myoepithelial markers, epithelial-mesenchymal transition (EMT), canonical Wnt signaling components, and other pathways induced by Wnt family members distinguishes DCIS from invasive. The information gained may help in stratifying DCIS as well as identify actionable targets for primary and tertiary prevention or targeted therapy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA