Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 972
Filtrar
1.
Dis Model Mech ; 17(3)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38501211

RESUMEN

Mitogen-activated protein 3 kinase 1 (MAP3K1) has a plethora of cell type-specific functions not yet fully understood. Herein, we describe a role for MAP3K1 in female reproductive tract (FRT) development. MAP3K1 kinase domain-deficient female mice exhibited an imperforate vagina, labor failure and infertility. These defects corresponded with shunted Müllerian ducts (MDs), the embryonic precursors of FRT, that manifested as a contorted caudal vagina and abrogated vaginal-urogenital sinus fusion in neonates. The MAP3K1 kinase domain is required for optimal activation of the Jun-N-terminal kinase (JNK) and cell polarity in the MD epithelium, and for upregulation of WNT signaling in the mesenchyme surrounding the caudal MD. The MAP3K1-deficient epithelial cells and MD epithelium had reduced expression of WNT7B ligands. Correspondingly, conditioned media derived from MAP3K1-competent, but not -deficient, epithelial cells activated a TCF/Lef-luciferase reporter in fibroblasts. These observations indicate that MAP3K1 regulates MD caudal elongation and FRT development, in part through the induction of paracrine factors in the epithelium that trans-activate WNT signaling in the mesenchyme.


Asunto(s)
Células Epiteliales , Quinasa 1 de Quinasa de Quinasa MAP , Vagina , Animales , Femenino , Ratones , Células Epiteliales/metabolismo , Epitelio/metabolismo , Vagina/metabolismo , Vía de Señalización Wnt , Quinasa 1 de Quinasa de Quinasa MAP/genética , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo
2.
J Immunol Res ; 2023: 6696967, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37928434

RESUMEN

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by the production of autoantibodies and tissue inflammation. Mesenchymal stem cells (MSCs) have emerged as a promising candidate therapy for SLE owing to the immunomodulatory and regenerative properties. Circulating miRNAs are small, single-stranded noncoding RNAs in a variety of body fluids that regulate numerous immunologic and inflammatory pathways. Recent studies have revealed many differentially expressed circulating miRNAs in autoimmune diseases including SLE. However, the role of circulating miRNAs in SLE has not been extensively studied. Here, we performed small RNA sequencing analysis to compare the circulating miRNA profiles of SLE patients before and after MSC transplantation (MSCT), and identified a significant decrease of circulating miR-320b level during MSCT. Importantly, we found that the expression of circulating miR-320b and its target gene MAP3K1 was closely associated with SLE disease activity. The in vitro experiments showed that decreased MAP3K1 level in SLE peripheral blood mononuclear cells (PBMCs) was involved in CD4+ T-cell proliferation. In MRL/lpr mice, miR-320b overexpression aggravated symptoms of SLE, while miR-320b inhibition could promote disease remission. Besides, MSCs regulate miR-320b/MAP3K1 expression both in vitro and in vivo. Our results suggested that circulating miR-320b and MAP3K1 may be involved in CD4+ T-cell proliferation in SLE. This trial is registered with NCT01741857.


Asunto(s)
Lupus Eritematoso Sistémico , Quinasa 1 de Quinasa de Quinasa MAP , MicroARNs , Animales , Humanos , Ratones , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Leucocitos Mononucleares/metabolismo , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/terapia , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Ratones Endogámicos MRL lpr , MicroARNs/genética , MicroARNs/metabolismo
3.
Kaohsiung J Med Sci ; 39(9): 896-903, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37314251

RESUMEN

The incidence of early-onset colorectal cancer (CRC), which affects people under 50, is increasing for unknown reasons. Additionally, no underlying genetic cause is found in 20%-30% of patients suspected of having familial CRC syndrome. Whole exome sequencing (WES) has generated evidence for new genes associated with CRC susceptibility, but many patients remain undiagnosed. This study applied WES in five early-onset CRC patients from three unrelated families to identify novel genetic variants that could be linked to rapid disease development. Furthermore, the candidate variants were validated using Sanger sequencing. Two heterozygote variations, c.1077-2A>G and c.199G>A, were found in the MSH2 and the MLH1 genes, respectively. Sanger sequencing analysis confirmed that these (likely) pathogenic mutations segregated in all the affected families' members. In addition, we identified a rare heterozygote variant (c.175C>T) with suspected pathogenic potential in the MAP3K1 gene; formally the variant is of uncertain significance (VUS). Our findings support the hypothesis that CRC onset may be oligogenic and molecularly heterogeneous. Larger and more robust studies are needed to understand the genetic basis of early-onset CRC development, combined with novel functional analyses and omics approaches.


Asunto(s)
Neoplasias Colorrectales , Quinasa 1 de Quinasa de Quinasa MAP , Humanos , Proteína 2 Homóloga a MutS/genética , Secuenciación del Exoma , Mutación/genética , Neoplasias Colorrectales/genética , Predisposición Genética a la Enfermedad , Homólogo 1 de la Proteína MutL/genética , Quinasa 1 de Quinasa de Quinasa MAP/genética
4.
Cell Oncol (Dordr) ; 46(5): 1213-1234, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37166744

RESUMEN

PURPOSE: In this study, we assessed whether the overexpression of MAP3K1 promotes the proliferation, migration, and invasion of breast cancer cells, which affect the prognosis of hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative early stage breast cancer. METHODS: Two HR-positive, HER2-negative breast cancer cell lines (MCF7 and T-47D) overexpressing MAP3K1 were transfected with two MAP3K1 short hairpin RNA plasmids (shMAP3K1 [#3] and shMAP3K1 [#5]). The proliferation, migration, and invasion of these cells were then examined. We assessed whether shMAP3K1 affects the cell cycle, levels of downstream signaling molecules (ERK, JNK, p38 MAPK, and NF-κB), and sensitivity to chemotherapeutic and hormonal agents. To assess the anti-tumor effect of MAP3K1 knockdown in the breast cancer orthotopic model, MCF7 and T-47D cells treated with or without shMAP3K1 (#3) and shMAP3K1 (#5) were inoculated into the mammary fat pads of mice. In total, 182 patients with HR-positive, HER2-negative T1 and T2 breast cancer and 0-3 nodal metastases were included. Additionally, 73 patients with T1 and T2 breast cancer and negative nodes who received adjuvant endocrine therapy alone were selected as an independent validation cohort. RESULTS: In both cell lines, shMAP3K1 (#3) and shMAP3K1 (#5) significantly reduced cell growth, migration, and invasion by downregulating MMP-9 and by blocking the G2/M phase of the cell cycle and its regulatory molecule cyclin B1. Moreover, both shMAP3K1 (#3) and shMAP3K1 (#5) downregulated ERK-, JNK-, p38 MAPK-, and NF-κB-dependent gene transcription and enhanced the sensitivity of both cell lines to doxorubicin, docetaxel, and tamoxifen. We observed that both shMAP3K1 (#3) and shMAP3K1 (#5) inhibited tumor growth compared with that in the scrambled group of MCF7 and T-47D cell orthotopic tumors. Patients with MAP3K1 overexpression exhibited significantly poorer 10-year disease-free survival (DFS) (70.4% vs. 88.6%, p = 0.003) and overall survival (OS) (81.9% vs. 96.3%, p = 0.001) than those without MAP3K1 overexpression. Furthermore, phospho-ERK (p < 0.001) and phospho-JNK (p < 0.001) expressions were significantly associated with MAP3K1 expression, and both phospho-ERK and phospho-JNK expressions were significantly correlated with poor 10-year DFS and OS. These biological findings, including a significant association between DFS and OS, and the expressions of MAP3K1, phospho-ERK, and phospho-JNK were further validated in an independent cohort. Multivariate analysis identified MAP3K1 expression as an independent poor prognostic factor for DFS and OS. CONCLUSION: Our results indicate that the overexpression of MAP3K1 plays a major role in the poor prognosis of HR-positive, HER2-negative early stage breast cancer.


Asunto(s)
Neoplasias de la Mama , Quinasa 1 de Quinasa de Quinasa MAP , Humanos , Animales , Ratones , Femenino , Neoplasias de la Mama/patología , FN-kappa B , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Tamoxifeno , Supervivencia sin Enfermedad , Proteínas Quinasas p38 Activadas por Mitógenos , Quinasa 1 de Quinasa de Quinasa MAP/genética
5.
Anim Biotechnol ; 34(3): 686-697, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37014133

RESUMEN

MAP3K1 is a significant member of the MAPK family, and its expressed MEKK1 protein has a wide range of biological activities and is an essential node in the MAPK signaling pathway. A significant number of studies have revealed that MAP3K1 plays a complicated function in the control of cell proliferation, apoptosis, invasion and movement, participates in the regulation of the immune system, and plays an important role in wound healing, tumorigenesis and other processes. In this study, we looked at the involvement of MAP3K1 in the control of hair follicle stem cells (HFSCs). Overexpression of MAP3K1 significantly promoted the proliferation of HFSCs by inhibiting apoptosis and promoting the transition from S phase to G2 phase. The transcriptome identified 189 (MAP3K1_OE) and 414 (MAP3K1_sh) differential genes. The two pathways with the most significant enrichment of differentially expressed genes were the IL-17 signaling pathway and TNF signaling pathway, and the significantly enriched terms in the GO enrichment analysis involved regulation of response of external stimulus, inflammatory and cytokine. Indicate that MAP3K1 can function as a promoting factor in HFSCs through the induction of cell cycle transition from S phase to G2 phase can inhibition apoptosis by mediating crosstalk among several pathways and cytokines.HIGHLIGHTSAbnormal MAP3K1 expression in hair follicle stem cells (HFSCs) can impair HFSC proliferation and apoptosis.MAP3K1 controls hair follicle stem cell proliferation via modulating cell apoptosis and the ratio of cells in S phase/G2 phase.The differential genes shared by MAP3K1_sh and MAP3K1_OE are enriched in GO terms such as inflammation, adipocyte differentiation, acute inflammation, and so on.


Asunto(s)
Folículo Piloso , Quinasa 1 de Quinasa de Quinasa MAP , Animales , Folículo Piloso/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Células Madre/metabolismo , Perfilación de la Expresión Génica , Citocinas/genética , Citocinas/metabolismo , Inflamación/metabolismo
6.
Genes (Basel) ; 14(3)2023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36980976

RESUMEN

Polymorphisms rs2472493 near ABCA1, rs7636836 in FNDC3B, and rs61275591 near the ANKRD55-MAP3K1 genes were previously reported to exhibit genome-wide significance in primary open-angle glaucoma (POAG). Since these polymorphisms have not been investigated in the Arab population of Saudi Arabia, we examined their association with POAG in a Saudi cohort. Genotyping was performed in 152 POAG cases and 246 controls using Taqman real-time assays and their associations with POAG and clinical markers, such as intraocular pressure, cup/disc ratio, and the number of antiglaucoma medications, were tested by statistical methods. There was no association observed between POAG and the minor allele frequencies of rs2472493[G], rs7636836[T], or rs61275591[A]. None of the genetic models such as co-dominant, dominant, recessive, over-dominant, and log-additive demonstrated any genotype link. The Rs2472493 genotype showed a modest association (p = 0.044) with the number of antiglaucoma medications in the POAG group, but no significant genotype effect on post hoc analysis. In addition, a G-T allelic haplotype of rs2472493 (ABCA1) and rs7636836 (FNDC3B) did show an over two-fold increased risk of POAG (odds ratio = 2.18), albeit non-significantly (p = 0.092). Similarly, no other allelic haplotype of the three variants showed any significant association with POAG. Our study did not replicate the genetic association of rs2472493 (ABCA1), rs763683 (FNDC3B), and rs61275591 (ANKRD55-MAP3K1) in POAG and related clinical phenotypes, suggesting that these polymorphisms are not associated with POAG in a Saudi cohort of Arab ethnicity. However, large population-based multicenter studies are needed to validate these results.


Asunto(s)
Glaucoma de Ángulo Abierto , Quinasa 1 de Quinasa de Quinasa MAP , Humanos , Glaucoma de Ángulo Abierto/genética , Glaucoma de Ángulo Abierto/epidemiología , Agentes Antiglaucoma , Arabia Saudita/epidemiología , Estudios de Casos y Controles , Polimorfismo Genético , Quinasa 1 de Quinasa de Quinasa MAP/genética , Fibronectinas/genética , Transportador 1 de Casete de Unión a ATP/genética , Proteínas Portadoras/genética
7.
Cancer Rep (Hoboken) ; 6(1): e1773, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36560873

RESUMEN

BACKGROUND: Accumulating studies have evaluated the association between MAP3K1 polymorphisms and cancer prognosis. However, the results of these studies are conflicting. Given the potential impact of MAP3K1 rs889312 SNP on the prognosis of various cancers, this meta-analysis was performed to obtain solid and credible evidence. METHODS AND MATERIALS: This study was performed according to the PRISMA 2020 statement. A comprehensive article search was conducted to find and select articles from multiple databases, including PubMed, Google Scholar, Web of Science, EMBASE and the Cochrane Library, published up to 15th September 2022. The data analysis was performed with Review Manager v5.2. Pooled HR with its 95% CI and p-value was calculated where HR >1 suggests worse/poor survival and HR <1 suggests better survival of cancer patients. RESULTS: A total of five articles comprising 24 439 patients were included for both qualitative and quantitative data synthesis. It was found that only the dominant genetic model (AC + CC vs. AA) showed a statistically significant poor overall survival for MAP3K1 rs889312 polymorphism (HR = 1.25, 95% CI = 1.06-1.47, p = .01). In addition, publication bias analysis by the Egger's test and the Begg-Mazumdar test reported no significant bias in the analysis of overall survival (p > .05). CONCLUSIONS: The present study concludes that MAP3K1 gene rs889312 polymorphism plays a prognostic role in the survival of cancer patients. However, future research is recommended that will analyze more MAP3K SNPs along with rs889312, which may reveal more credible outcomes in terms of cancer prognosis.


Asunto(s)
Quinasa 1 de Quinasa de Quinasa MAP , Neoplasias , Humanos , Neoplasias/diagnóstico , Neoplasias/genética , Polimorfismo de Nucleótido Simple , Pronóstico
8.
BMC Gastroenterol ; 22(1): 513, 2022 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-36510163

RESUMEN

BACKGROUND/OBJECTIVES: The hormone-dependent effect of MAP3K1 gene polymorphisms may explain sex-specific differences in gastric cancer (GC) risk. Phytoestrogens have been shown to interact with this genetic factor. Here, we investigated the association between MAP3K1 gene polymorphisms and GC risk by sex and whether these associations differ depending on soy products intake. METHODS: Participants aged 20-79 years were recruited from two hospitals between December 2002 and September 2006. In all, 440 cases and 485 controls were recruited, among, 246 pairs of cases and controls, matched by sex, age (± 5 years), study admission period (± 1 years), and hospital, were included for the analysis. RESULTS: In dominant model, men with the A allele of rs252902 showed significantly increased GC risk (odd ratio; OR=2.19, 95% confidence interval; CI=1.31-3.64) compared to GG homozygotes. When stratified by intake of soy products, men with the A allele of rs252902 and low intake of soy products showed significantly higher GC risk (OR=3.29, 95% CI=1.55-6.78) than that in GG homozygotes. CONCLUSIONS: Men with the risk allele of MAP3K1 had a significantly increased GC risk compared to GG homozygotes; this trend was more pronounced in those with low intake of soy products.


Asunto(s)
Quinasa 1 de Quinasa de Quinasa MAP , Neoplasias Gástricas , Masculino , Femenino , Humanos , Neoplasias Gástricas/genética , Estudios de Casos y Controles , Polimorfismo de Nucleótido Simple , Alelos , Oportunidad Relativa , Factores de Riesgo , Predisposición Genética a la Enfermedad , Quinasa 1 de Quinasa de Quinasa MAP/genética
9.
Mol Med Rep ; 26(5)2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36102299

RESUMEN

The 46, XY disorder of sex development (DSD) is the main cause of birth defects; however, as it is a group of highly heterogeneous diseases, >50% of cases are not accurately diagnosed. Identification of more cases will improve understanding of the relationship between genotype and phenotype for DSD. The present study conducted a systematic analysis of the clinical characteristics of a proband with 46, XY DSD, applied genetic analysis by whole­exome sequencing to this pedigree and performed bioinformatics analysis of the identified variant. The proband presented with a short penis, lack of testicles and partial growth hormone (GH) deficiency at 1 year old. Histopathological examination revealed there were oviduct, epididymis and fibrous vascular tissue on both sides of the abdomen. The last follow­up at 5 years of age revealed that the patient exhibited restricted growth, a 1.5­cm penis and lack of testicles. Notably, a novel pathogenic mitogen­activated protein kinase kinase kinase 1 (MAP3K1) variant (c.3020A>G) was identified in the proband, resulting in a change in the 1,007th amino acid (glutamine) of the encoded protein. This variant caused the uncharged neutral glutamine to be replaced by a positively charged basic arginine. p.Gln1007 in MAP3K1 was confirmed to be conserved across various species. Pathogenicity analysis using bioinformatics tools suggested that this MAP3K1 variant may cause functional defects. In conclusion, the present study identified a novel MAP3K1 variant that was the cause of 46, XY DSD and partial GH deficiency. The present findings extend the mutation spectrum of MAP3K1 and provide novel characteristics of 46, XY DSD.


Asunto(s)
Trastorno del Desarrollo Sexual 46,XY , Quinasa 1 de Quinasa de Quinasa MAP , Trastorno del Desarrollo Sexual 46,XY/diagnóstico , Trastorno del Desarrollo Sexual 46,XY/genética , Glutamina/genética , Hormona del Crecimiento/genética , Humanos , Quinasa 1 de Quinasa de Quinasa MAP/genética , Masculino , Fenotipo
10.
Taiwan J Obstet Gynecol ; 61(5): 903-905, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36088066

RESUMEN

OBJECTIVE: Swyer syndrome, or 46, XY complete gonadal dysgenesis, is a disorder of human sexual development which present with female external genitalia, lack of female reproductive organs, and a 46, XY karyotype. Many genes that participate in human sexual development have been implicated in the pathogenesis of 46, XY gonadal dysgenesis. CASE REPORT: A 18-year-old phenotypically female was presented with primary amenorrhea. Surveillance revealed hypergonadotropic hypogonadism, a normal male 46, XY karyotype and absent of functional gonad, which was confirmed by pathological examination of the streak gonad. Whole exome sequencing showed germline mutations of a novel missense variant, c.570G > C, p.Lys190Asn, in exon 2 of MAP3K1 gene. CONCLUSION: Given evolutionary conservation of lysine residue at position 190, the amino acid substitution may interfere with interaction between MAP3K1 and RHOA, and contributes to complete gonadal dysgenesis in the context of 46,XY.


Asunto(s)
Disgenesia Gonadal 46 XY , Disgenesia Gonadal , Quinasa 1 de Quinasa de Quinasa MAP , Síndrome de Turner , Adolescente , Femenino , Disgenesia Gonadal 46 XY/genética , Humanos , Cariotipificación , Quinasa 1 de Quinasa de Quinasa MAP/genética , Masculino , Mutación Missense
11.
Sci Rep ; 12(1): 11482, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35798792

RESUMEN

Epithelial development starts with stem cell commitment to ectoderm followed by differentiation to the basal keratinocytes. The basal keratinocytes, first committed in embryogenesis, constitute the basal layer of the epidermis. They have robust proliferation and differentiation potential and are responsible for epidermal expansion, maintenance and regeneration. We generated basal epithelial cells in vitro through differentiation of mouse embryonic stem cells (mESCs). Early on in differentiation, the expression of stem cell markers, Oct4 and Nanog, decreased sharply along with increased ectoderm marker keratin (Krt) 18. Later on, Krt 18 expression was subdued when cells displayed basal keratinocyte characteristics, including regular polygonal shape, adherent and tight junctions and Krt 14 expression. These cells additionally expressed abundant Sca-1, Krt15 and p63, suggesting epidermal progenitor characteristics. Using Map3k1 mutant mESCs and environmental dioxin, we examined the gene and environment effects on differentiation. Neither Map3k1 mutation nor dioxin altered mESC differentiation to ectoderm and basal keratinocytes, but they, individually and in combination, potentiated Krt 1 expression and basal to spinous differentiation. Similar gene-environment effects were observed in vivo where dioxin exposure increased Krt 1 more substantially in the epithelium of Map3k1+/- than wild type embryos. Thus, the in vitro model of epithelial differentiation can be used to investigate the effects of genetic and environmental factors on epidermal development.


Asunto(s)
Dioxinas , Queratinocitos , Quinasa 1 de Quinasa de Quinasa MAP , Células Madre Embrionarias de Ratones , Animales , Diferenciación Celular , Dioxinas/farmacología , Células Epidérmicas , Epidermis/metabolismo , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Quinasa 1 de Quinasa de Quinasa MAP/genética , Ratones , Células Madre Embrionarias de Ratones/citología , Mutación
12.
DNA Cell Biol ; 41(6): 590-599, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35533015

RESUMEN

The liver of poultry is the primary site of lipid synthesis. The excessive production of lipids accumulates in liver tissues causing lipid metabolism disorders, which result in fatty liver disease and have a transgenerational effect of acquired phenotypes. However, its specific mechanisms have not yet been fully understood. In this study, the differentially expressed miR-375 as well as its target gene MAP3K1 (mitogen-activated protein kinase kinase kinase 1) were screened out by interaction network analysis of microRNA sequencing results and transcriptome profiling in the fatty liver group of the F0-F3 generation (p < 0.05 or p < 0.01). Furthermore, the results showed that the number of lipid droplets and triglyceride content were significantly decreased after upregulation of miR-375 in primary hepatocyte culture in vitro (p < 0.05 or p < 0.01). The MAP3K1 knockdown group exhibited the opposite trends (p < 0.05 or p < 0.01). P53, Bcl-x, PMP22, and CDKN2C related to cell proliferation were significantly upregulated or downregulated after knocking down MAP3K1 (p < 0.05). This research uniquely revealed that silencing miR-375 inhibits lipid biosynthesis and promotes cell proliferation, which may be due to the partial regulation of the expression level of MAP3K1, thereby further participating in the transgenerational inheritance process of regulating liver lipid metabolism. These results reveal the pathogenesis of fatty liver in noncoding RNA and provide good candidate genes for breeding progress of disease resistance in chickens.


Asunto(s)
Hígado Graso , Quinasa 1 de Quinasa de Quinasa MAP , MicroARNs , Animales , Pollos/genética , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/veterinaria , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Aves de Corral , Triglicéridos/metabolismo
13.
Biochem Biophys Res Commun ; 612: 188-195, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35567901

RESUMEN

Glioblastoma (GBM) stem cells (GSCs) are responsible for GBM initiation, progression, infiltration, standard therapy resistance, and recurrence. However, the mechanisms underlying GSC invasion remain incompletely understood. Using public single-cell RNA-Seq data, we identified MAP3K1 as a master regulator of infiltrative GSCs through c-JUN signaling regulation. MAP3K1 knockdown significantly decreased GSC invasion capacity, proliferation, and stemness in vitro. Moreover, in an orthotopic xenograft model, knockdown of MAP3K1 prominently suppressed GSC infiltration along the corpus callosum and tumor progression and prolonged mouse survival. Mechanistically, MAP3K1 regulates GSC invasion through phosphorylation of downstream c-JUN at serine 63 and 73, as confirmed using the CPTAC phosphoproteome dataset. Furthermore, the c-JUN inhibitor JNK-IN-8 significantly decreased GSC invasion, proliferation, and stemness. Taken together, our study demonstrates that MAP3K1 regulates GSC invasion and tumor progression via activation of c-JUN signaling and indicates that the MAP3K1/c-JUN signaling axis is a therapeutic target for infiltrative GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Quinasa 1 de Quinasa de Quinasa MAP , Animales , Benzamidas , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular , Glioblastoma/patología , Humanos , Ratones , Células Madre Neoplásicas/patología , Piridinas , Pirimidinas
14.
J Clin Lab Anal ; 36(6): e24470, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35524422

RESUMEN

BACKGROUND: Papillary thyroid carcinoma (PTC) grows slowly but has a great risk of metastasis. MicroRNAs are well known as vital tumor-related gene regulators. In PTC, the role of miR-203a-3p and the underlying mechanisms remain not completely understood. METHODS: We conducted CCK8 assay, wound healing assay, transwell experiment and flow cytometry analyses to investigate the function of miRNA-203a-3p. The interaction of miRNA-203a-3p with its gene MAP3K1 was characterized by quantitative real-time polymerase chain reaction, western blotting and luciferase assay. RESULTS: We found that the levels of miRNA-203a-3p were statistically decreased in PTC tissues. When mimics were delivered to TPC-1 and KTC-1 cells to upregulate miR-203a-3p, it was observed that cell proliferation, metastatic abilities and cell cycle process were prevented but cell apoptosis was enhanced. Furthermore, we proved the interaction between MAP3K1 and miR-203a-3p. Intriguingly, similar to miR-203a-3p mimics, siMAP3K1 showed a tumor-suppressive effect, and this effect could be reversed when miR-203a-3p was simultaneously inhibited. Finally, selected autophagy-linked proteins such as LC3 Beclin-1 were detected and found to be increased when miR-203a-3p was upregulated or MAP3K1 was inhibited. CONCLUSION: Overall, miR-203a-3p inhibits the oncogenic characteristics of TPC-1 and KTC-1 cells via suppressing MAP3K1 and activating autophagy. Our findings might enrich the understanding and the therapeutic strategies of PTC.


Asunto(s)
Carcinoma Papilar , Quinasa 1 de Quinasa de Quinasa MAP , MicroARNs , Neoplasias de la Tiroides , Autofagia/genética , Carcinoma Papilar/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Quinasa 1 de Quinasa de Quinasa MAP/genética , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología
15.
Proc Natl Acad Sci U S A ; 119(18): e2115071119, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35476515

RESUMEN

Activation of inhibitor of nuclear factor NF-κB kinase subunit-ß (IKKß), characterized by phosphorylation of activation loop serine residues 177 and 181, has been implicated in the early onset of cancer. On the other hand, tissue-specific IKKß knockout in Kras mutation-driven mouse models stalled the disease in the precancerous stage. In this study, we used cell line models, tumor growth studies, and patient samples to assess the role of IKKß and its activation in cancer. We also conducted a hit-to-lead optimization study that led to the identification of 39-100 as a selective mitogen-activated protein kinase kinase kinase (MAP3K) 1 inhibitor. We show that IKKß is not required for growth of Kras mutant pancreatic cancer (PC) cells but is critical for PC tumor growth in mice. We also observed elevated basal levels of activated IKKß in PC cell lines, PC patient-derived tumors, and liver metastases, implicating it in disease onset and progression. Optimization of an ATP noncompetitive IKKß inhibitor resulted in the identification of 39-100, an orally bioavailable inhibitor with improved potency and pharmacokinetic properties. The compound 39-100 did not inhibit IKKß but inhibited the IKKß kinase MAP3K1 with low-micromolar potency. MAP3K1-mediated IKKß phosphorylation was inhibited by 39-100, thus we termed it IKKß activation modulator (IKAM) 1. In PC models, IKAM-1 reduced activated IKKß levels, inhibited tumor growth, and reduced metastasis. Our findings suggests that MAP3K1-mediated IKKß activation contributes to KRAS mutation-associated PC growth and IKAM-1 is a viable pretherapeutic lead that targets this pathway.


Asunto(s)
Quinasa 1 de Quinasa de Quinasa MAP , Neoplasias Pancreáticas , Humanos , Quinasa I-kappa B/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas , Neoplasias Pancreáticas
16.
Sex Dev ; 16(2-3): 92-97, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35290982

RESUMEN

Pathogenic variants in the MAP3K1 gene are an important cause of 46,XY non-syndromic partial and complete gonadal dysgenesis, accounting for at least 4% of cases. Inheritance occurs in a sex-limited, autosomal dominant fashion with virtually complete penetrance in 46,XY individuals. 46,XX carriers appear to have normal fertility and no developmental abnormalities. Pathogenic variants occur almost exclusively within known domains of the MAP3K1 protein, facilitating annotation when identified. Where studied, these variants have been modeled to alter the local MAP3K1 folding and surface domains and have been shown to alter interactions with known binding partners. The net effect of these variants is to increase phosphorylation of downstream targets ERK1, ERK2, and p38, resulting in multiple gain-of-function effects interfering with testis determination and enabling ovarian determination.


Asunto(s)
Disgenesia Gonadal 46 XY , Disgenesia Gonadal , Quinasa 1 de Quinasa de Quinasa MAP , Masculino , Humanos , Quinasa 1 de Quinasa de Quinasa MAP/genética , Disgenesia Gonadal 46 XY/genética , Disgenesia Gonadal 46 XY/patología , Disgenesia Gonadal/genética , Heterocigoto , Testículo/patología
17.
Cell Cycle ; 21(11): 1194-1211, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35230926

RESUMEN

Colon cancer (CC) is a common malignant tumor of the digestive tract. Circular RNAs (circRNAs) play important roles in the progression of CC. This study aimed to explore the role and mechanism of circRNA_0085315 in CC. In this study, we used qRT-PCR and Western blot assays to analyze the expressions of circRNA, miRNA, and mRNA as well as the expression of the related proteins. Luciferase reporter, RNA pull-down, and qRT-PCR assays were used to prove the relationship among circRNA, miRNA, and mRNA. CCK-8, colony formation, and transwell assays were used to perform the analysis of cell proliferation, migration, and invasion. Our results showed that the higher circRNA_0085315 expression led to the poorer prognosis of CC patients. The function of circRNA_0085315 as a ceRNA in competing with MAP3K1 mRNA to sponge miR-1200. CircRNA_0085315 sponged miR-1200 to promote cell proliferation, migration, and invasion and affected the expression of Ki67, MMP2, E-cadherin, and N-cadherin, but not circRNA_0085315-mut without the binding site of miR-1200. MAP3K1-overexpression or miR-1200 mimics prevented the suppression on the enhanced cell proliferation, migration, and invasion caused by circRNA_0085315-overexpression. circRNA_0085315 increased the phosphorylation levels of JNK, p38, and ERK1/2 by stimulating MAP3K1 up-regulation caused by miR-1200 inhibition. In conclusion, circRNA_0085315 serves as a ceRNA and promotes CC progression through the activation of the MAPK signaling pathway mediated via the miR-1200/MAP3K1 axis, suggesting that circRNA_0085315 may be a promising diagnostic and therapeutic target for CC.


Asunto(s)
Neoplasias del Colon , Quinasa 1 de Quinasa de Quinasa MAP , MicroARNs , ARN Circular , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Humanos , Quinasa 1 de Quinasa de Quinasa MAP/genética , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Sistema de Señalización de MAP Quinasas , MicroARNs/genética , MicroARNs/metabolismo , ARN Circular/genética , ARN Circular/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
18.
Hepatol Commun ; 6(1): 133-160, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34558855

RESUMEN

Alcohol-associated liver disease (ALD) is a significant clinical problem for which the most effective therapy is alcohol abstinence. The two aims of this study were, first, to identify the liver transcriptome, fecal microbiome, and portal serum metabolome at peak injury and during early and late resolution from ALD; and second, to integrate their interactions and understand better the pathogenesis of ALD. To provoke alcohol-induced liver injury, female and male wild-type mice were fed the control or ethanol Lieber-DeCarli diets for 6 weeks. To study early and late resolution, alcohol was withdrawn from the diet and mice were sacrificed after 3 and 14 days, respectively. At peak injury, there was increased signal transducer and activator of transcription (Stat3), Rho-GTPases, Tec kinase and glycoprotein VI (Gp6), and decreased peroxisome proliferator-activated receptor signaling. During resolution from ALD, there was up-regulation of vitamin D receptor/retinoid X receptor, toll-like receptor, p38 and Stat3, and down-regulation of liver X receptor signaling. Females showed significant changes in catabolic pathways, whereas males increased cellular stress, injury, and immune-response pathways that decreased during resolution. The bacterial genus Alistipes and the metabolite dipeptide glycyl-L-leucine increased at peak but decreased during resolution from ALD in both genders. Hepatic induction of mitogen-activated protein kinase (Map3k1) correlated with changes in the microbiome and metabolome at peak but was restored during ALD resolution. Inhibition of MAP3K1 protected from ALD in mice. Conclusion: Alcohol abstinence restores the liver transcriptome, fecal microbiome, and portal serum metabolome in a gender-specific manner. Integration of multiomics data identified Map3k1 as a key gene driving pathogenesis and resolution from ALD.


Asunto(s)
Hepatopatías Alcohólicas/genética , Hepatopatías Alcohólicas/metabolismo , Metaboloma , Microbiota , Transcriptoma , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Heces/microbiología , Femenino , Hepatocitos/metabolismo , Hepatopatías Alcohólicas/microbiología , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Masculino , Ratones Endogámicos C57BL , Regulación hacia Arriba
19.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34911761

RESUMEN

Arterial remodeling is an important adaptive mechanism that maintains normal fluid shear stress in a variety of physiologic and pathologic conditions. Inward remodeling, a process that leads to reduction in arterial diameter, plays a critical role in progression of such common diseases as hypertension and atherosclerosis. Yet, despite its pathogenic importance, molecular mechanisms controlling inward remodeling remain undefined. Mitogen-activated protein kinases (MAPKs) perform a number of functions ranging from control of proliferation to migration and cell-fate transitions. While the MAPK ERK1/2 signaling pathway has been extensively examined in the endothelium, less is known about the role of the MEKK3/ERK5 pathway in vascular remodeling. To better define the role played by this signaling cascade, we studied the effect of endothelial-specific deletion of its key upstream MAP3K, MEKK3, in adult mice. The gene's deletion resulted in a gradual inward remodeling of both pulmonary and systematic arteries, leading to spontaneous hypertension in both vascular circuits and accelerated progression of atherosclerosis in hyperlipidemic mice. Molecular analysis revealed activation of TGFß-signaling both in vitro and in vivo. Endothelial-specific TGFßR1 knockout prevented inward arterial remodeling in MEKK3 endothelial knockout mice. These data point to the unexpected participation of endothelial MEKK3 in regulation of TGFßR1-Smad2/3 signaling and inward arterial remodeling in artery diseases.


Asunto(s)
Hipertensión Pulmonar/patología , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , MAP Quinasa Quinasa Quinasa 3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Remodelación Vascular/fisiología , Animales , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Genotipo , Miembro Posterior/irrigación sanguínea , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipertensión Pulmonar/metabolismo , Isquemia , Quinasa 1 de Quinasa de Quinasa MAP/genética , MAP Quinasa Quinasa Quinasa 3/genética , Ratones , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/toxicidad , Transducción de Señal , Tamoxifeno/toxicidad , Factor de Crecimiento Transformador beta/genética
20.
PLoS Negl Trop Dis ; 15(12): e0010027, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34879059

RESUMEN

BACKGROUND: The metacestode larval stage of the fox-tapeworm Echinococcus multilocularis causes alveolar echinococcosis by tumour-like growth within the liver of the intermediate host. Metacestode growth and development is stimulated by host-derived cytokines such as insulin, fibroblast growth factor, and epidermal growth factor via activation of cognate receptor tyrosine kinases expressed by the parasite. Little is known, however, concerning signal transmission to the parasite nucleus and cross-reaction with other parasite signalling systems. METHODOLOGY/PRINCIPAL FINDINGS: Using bioinformatic approaches, cloning, and yeast two-hybrid analyses we identified a novel mitogen-activated kinase (MAPK) cascade module that consists of E. multilocularis orthologs of the tyrosine kinase receptor interactor Growth factor receptor-bound 2, EmGrb2, the MAPK kinase kinase EmMEKK1, a novel MAPK kinase, EmMKK3, and a close homolog to c-Jun N-terminal kinase (JNK), EmMPK3. Whole mount in situ hybridization analyses indicated that EmMEKK1 and EmMPK3 are both expressed in E. multilocularis germinative (stem) cells but also in differentiated or differentiating cells. Treatment with the known JNK inhibitor SP600125 led to a significantly reduced formation of metacestode vesicles from stem cells and to a specific reduction of proliferating stem cells in mature metacestode vesicles. CONCLUSIONS/SIGNIFICANCE: We provide evidence for the expression of a MEKK1-JNK MAPK cascade module which, in mammals, is crucially involved in stress responses, cytoskeletal rearrangements, and apoptosis, in E. multilocularis stem cells. Inhibitor studies indicate an important role of JNK signalling in E. multilocularis stem cell survival and/or maintenance. Our data are relevant for molecular and cellular studies into crosstalk signalling mechanisms that govern Echinococcus stem cell function and introduce the JNK signalling cascade as a possible target of chemotherapeutics against echinococcosis.


Asunto(s)
Echinococcus multilocularis/enzimología , Proteínas del Helminto/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Células Madre/enzimología , Animales , Proliferación Celular , Echinococcus multilocularis/genética , Echinococcus multilocularis/crecimiento & desarrollo , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Proteínas del Helminto/genética , MAP Quinasa Quinasa 4/genética , Quinasa 1 de Quinasa de Quinasa MAP/genética , MAP Quinasa Quinasa Quinasa 3/genética , MAP Quinasa Quinasa Quinasa 3/metabolismo , Sistema de Señalización de MAP Quinasas , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...