Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
BMC Cancer ; 21(1): 803, 2021 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-34253170

RESUMEN

BACKGROUND: Although the major anticancer effect of metformin involves AMPK-dependent or AMPK-independent mTORC1 inhibition, the mechanisms of action are still not fully understood. METHODS: To investigate the molecular mechanisms underlying the effect of metformin on the mTORC1 inhibition, MTT assay, RT-PCR, and western blot analysis were performed. RESULTS: Metformin induced the expression of ATF4, REDD1, and Sestrin2 concomitant with its inhibition of mTORC1 activity. Treatment with REDD1 or Sestrin2 siRNA reversed the mTORC1 inhibition induced by metformin, indicating that REDD1 and Sestrin2 are important for the inhibition of mTORC1 triggered by metformin treatment. Moreover, REDD1- and Sestrin2-mediated mTORC1 inhibition in response to metformin was independent of AMPK activation. Additionally, lapatinib enhances cell sensitivity to metformin, and knockdown of REDD1 and Sestrin2 decreased cell sensitivity to metformin and lapatinib. CONCLUSIONS: ATF4-induced REDD1 and Sestrin2 expression in response to metformin plays an important role in mTORC1 inhibition independent of AMPK activation, and this signalling pathway could have therapeutic value.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Metformina/farmacología , Metformina/uso terapéutico , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Humanos , Transfección
3.
Brain Sci ; 13(11)2023 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-38002533

RESUMEN

To date, budget management in virtual shopping training has not been given much importance. The main objective of this study was to investigate the effects of virtual shopping budget-management training on executive functions and brain activation. Sixteen participants were randomly assigned to the experimental group that received virtual shopping budget-management training or the waitlist control group for a total of 16 sessions. To examine the effects of virtual shopping budget-management training on brain activation, HbO2 was measured in the prefrontal cortex via functional near-infrared spectroscopy (fNIRS) during the Trail Making Test Part B (TMT-B) and Stroop test. Mann-Whitney and Wilcoxon signed-rank tests were used to compare outcomes between and within the two groups. The virtual shopping budget-management training showed no significant difference in all outcomes between both groups (p > 0.05). No significant differences were observed in HbO2 levels during both TMT-B (p > 0.05) and the Stroop test (p > 0.05). However, in the pre-post comparisons, there was a significant difference in the TMT-B (p < 0.05) and Stroop test (p < 0.05) in the experimental group. In this study, although we did not find a distinct advantage in training, it confirmed its potential for clinical benefits in healthy young adults through training.

4.
PeerJ ; 10: e13461, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35607451

RESUMEN

Background: Melatonin, a neurohormone, maybe involved in physiological processes, such as antioxidation, anti-inflammation, and hair growth. In the present study, we investigated the effects of melatonin on proliferation and intracellular signaling in DP cells using a three-dimensional (3D) spheroid culture system that mimics the in vivo hair follicle system. Methods: DP cells were incubated in monolayer (2D) and 3D spheroid culture systems. The expression levels of melatonin receptors in DP cells were analyzed using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blotting. The effect of melatonin on the hair-inductive property of DP cells was analyzed using a WST-1-based proliferation assay, determination of DP spheroid size, expression analysis of DP signature genes, and determination of ß-catenin stabilization in DP cells. The AKT/GSK3ß/ß-catenin signaling pathway associated with melatonin-induced ß-catenin stabilization in DP cells was investigated by analyzing changes in upstream regulator proteins, including AKT, GSK3ß, and their phosphorylated forms. Results: The expression levels of the melatonin receptors were higher in human DP cells than in human epidermal keratinocytes and human dermal fibroblast cells. Comparing the expression level according to the human DP cell culture condition, melatonin receptor expression was upregulated in the 3D culture system compared to the traditional two-dimensional monolayer culture system. Cell viability analysis showed that melatonin concentrations up to 1 mM did not affect cell viability. Moreover, melatonin increased the diameter of DP cell 3D spheroids in a dose-dependent manner. Immunoblotting and qRT-PCR analysis revealed that melatonin upregulated the expression of hair growth-related genes, including alkaline phosphatase, bone morphogenetic protein 2, versican, and wingless-int 5A, in a melatonin receptor-dependent manner. Cell fractionation analysis showed that melatonin increased the nuclear localization of ß-catenin. This result correlated with the increased transcriptional activation of T-cell factor/lymphoid enhancer factor-responsive luciferase induced by melatonin treatment. Interestingly, melatonin induced the phosphorylation of protein kinase B/AKT at serine 473 residue and GSK-3ß at serine 9 residue. To determine whether AKT phosphorylation at serine 473 induced ß-catenin nuclear translocation through GSK3ß phosphorylation at serine 9, the PI3K/AKT inhibitor LY294002 was cotreated with melatonin. Immunoblotting showed that LY294002 inhibited melatonin-induced phosphorylation of GSK3ß at serine 9 residue and ß-catenin activation. Conclusion: Collectively, this report suggests that melatonin promotes growth properties by activating the AKT/GSK3ß/ß-catenin signaling pathway through melatonin receptors.


Asunto(s)
Melatonina , Proteínas Proto-Oncogénicas c-akt , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Melatonina/farmacología , Receptores de Melatonina , Células Cultivadas , beta Catenina/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal
5.
J Breast Cancer ; 25(5): 387-403, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36314765

RESUMEN

PURPOSE: Phosphorylated AKT1 (p-AKT1) at Ser473 is a functional isoform of AKT and a key component of the PI3K/mTOR/AKT pathway. This study aimed to evaluate the prognostic significance of p-AKT1 (Ser473) based on the molecular subtypes of breast cancer. METHODS: To investigate the prognostic value of p-AKT1 (Ser473), we performed a retrospective chart review of patients with breast cancer. Data on p-AKT1 (Ser473) positivity, hormone receptor (HR) status, human epidermal growth factor receptor 2 (HER2) expression status, and other clinicopathological factors were obtained. Furthermore, the therapeutic effect of blocking p-AKT1 (Ser473) in breast cancer cells was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, cell apoptosis assay, apoptosis protein array, and western blot analysis. RESULTS: A total of 3,044 patients were evaluated, and the median follow-up time was 43 (range: 0-125) months. In patients with HR-positive and HER2-positive disease, the p-AKT1 (Ser473)-positive group had worse disease-free survival (DFS) than the p-AKT1 (Ser473)-negative group (hazard ratio, 1.9; 95% confidence interval, 1.1-3.5; p = 0.024). In the multivariate analysis, p-AKT1 (Ser473) remained a significantly worse prognostic factor in patients with HR-positive/HER2-positive breast cancer (p = 0.03). There was no difference in DFS according to p-AKT1 (Ser473) status among patients with other breast cancer subgroups. In vitro analysis showed that blocking p-AKT1 (Ser473) levels enhanced trastuzumab-induced cell death in HR-positive/HER2-positive and p-AKT1 (Ser473)-positive breast cancer cells. CONCLUSION: p-AKT1 (Ser473) is a prognostic marker for poor outcomes in patients with HR-positive/HER2-positive breast cancer and may have a potential value as a therapeutic target.

6.
Sci Rep ; 11(1): 10037, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976340

RESUMEN

Although a considerable volume of data supporting induction or aggravation of psoriasis because of angiotensin-converting enzyme (ACE) inhibitor use exists, it remains insufficient for definitive conclusions. Therefore, we aimed to evaluate the association between ACE inhibitor use and psoriasis incidence through a systematic literature review and meta-analysis. We searched for qualifying studies across PubMed, Web of Science, and Embase. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to evaluate the strength of the association between ACE inhibitor use and psoriasis incidence. Eight studies with a total of 54,509 patients with a psoriasis diagnosis were included in this meta-analysis. The pooled OR for psoriasis incidence among ACE inhibitor users was 1.52 (95% CI, 1.16-2.00) compared to that among non-users. From subgroup analysis by continent, the OR for ACE inhibitor users versus non-users was 2.37 (95% CI 1.28-4.37) in Asia. Per the subgroup analysis by climate, the OR for ACE inhibitor users vs non-users in dry climate was 3.45 (95% CI: 2.05-5.79) vs 1.32 (95% CI 1.01-1.73) in temperate climate. Our results reveal a significant association between ACE inhibitor use and psoriasis incidence.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Psoriasis/epidemiología , Humanos , Incidencia , Psoriasis/inducido químicamente
7.
J Microbiol Biotechnol ; 31(7): 933-941, 2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34099599

RESUMEN

Ginsenoside Rg4 is a rare ginsenoside that is naturally found in ginseng, and exhibits a wide range of biological activities including antioxidant and anti-inflammatory properties in several cell types. The purpose of this study was to use an in vivo model of hair follicle (HF)-mimic based on a human dermal papilla (DP) spheroid system prepared by three-dimensional (3D) culture and to investigate the effect of Rg4 on the hair-inductive properties of DP cells. Treatment of the DP spheroids with Rg4 (20 to 50 µg/ml) significantly increased the viability and size of the DP spheres in a dose-dependent manner. Rg4 also increased the mRNA and protein expression of DP signature genes that are related to hair growth including ALP, BMP2, and VCAN in the DP spheres. Analysis of the signaling molecules and luciferase reporter assays further revealed that Rg4 induces the activation of phosphoinositide 3-kinase (PI3K)/AKT and the inhibitory phosphorylation of GSK3ß, which activates the WNT/ß-catenin signaling pathway. These results correlated with not only the increased nuclear translocation of ß-catenin following the treatment of the DP spheres with Rg4 but also the significant elevation of mRNA expression of the downstream target genes of the WNT/ß-catenin pathway including WNT5A, ß-catenin, and LEF1. In conclusion, these results demonstrated that ginsenoside Rg4 promotes the hair-inductive properties of DP cells by activating the AKT/GSK3ß/ß-catenin signaling pathway in DP spheres, suggesting that Rg4 could be a potential natural therapy for hair growth.


Asunto(s)
Dermis/efectos de los fármacos , Ginsenósidos/farmacología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Cabello/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , beta Catenina/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Dermis/citología , Dermis/metabolismo , Cabello/crecimiento & desarrollo , Folículo Piloso/citología , Folículo Piloso/efectos de los fármacos , Folículo Piloso/crecimiento & desarrollo , Folículo Piloso/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Esferoides Celulares , Proteínas Wnt/metabolismo
8.
Anticancer Res ; 41(7): 3481-3487, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34230143

RESUMEN

BACKGROUND/AIM: Metformin is an antidiabetic drug that has been reported to have antitumor activity in many cancer types. This study investigated the molecular mechanisms underlying the antitumor effect of metformin. MATERIALS AND METHODS: We investigated the molecular mechanism of the antitumor effect of metformin alone and in combination with AKT serine/threonine kinase (AKT) inhibition via cell viability and western blot analyses. RESULTS: Notably, metformin increased the phosphorylation of AKT at serine 473 using protein array screening. Metformin-induced AKT activation was markedly suppressed by siRNA targeting activating transcription factor 4 (ATF4) but not AMP-activated protein kinase α. These results indicate that AKT activation by metformin was induced in an ATF4-dependent and AMPKα-independent manner. Treatment using metformin combined with MK-2206, an AKT inhibitor, or a siRNA for AKT markedly reduced the viability of cells compared with those cells treated with these agents alone. In addition, MK-2206 increased cell sensitivity to the combination of metformin with ionizing radiation or cisplatin. CONCLUSION: Inhibition of AKT can enhance the antitumor effect of metformin and would be a promising strategy to sensitize non-small-cell lung cancer to a combination of metformin with radiation or cisplatin.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Metformina/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Hipoglucemiantes/farmacología , Neoplasias Pulmonares/metabolismo
9.
Sci Rep ; 9(1): 653, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30679694

RESUMEN

Progesterone receptor membrane component1 (PGRMC1) is a heme-binding protein involved in cancers and Alzheimer's disease. PGRMC1 consists of a short N-terminal extracellular or luminal domain, a single membrane-spanning domain, and a long cytoplasmic domain. Previously, we generated two monoclonal antibodies (MAbs) 108-B6 and 4A68 that recognize cell surface-expressed PGRMC1 (csPGRMC1) on human pluripotent stem cells and some cancer cells. In this study, flow cytometric analysis found that an anti-PGRMC1 antibody recognizing the N-terminus of PGRMC1 could not bind to csPGRMC1 on cancer cells, and 108-B6 and 4A68 binding to csPGRMC1 was inhibited by trypsin treatment, suggesting that the epitopes of 108-B6 and 4A68 are trypsin-sensitive. To examine the epitope specificity of 108-B6 and 4A68, glutathione-S-transferase (GST)-fused PGRMC1 mutants were screened to identify the epitopes targeted by the antibodies. The result showed that 108-B6 and 4A68 recognized C-terminal residues 183-195 and 171-182, respectively, of PGRMC1, where trypsin-sensitive sites are located. A polyclonal anti-PGRMC1 antibody raised against the C-terminus of PGRMC1 could also recognized csPGRMC1 in a trypsin-sensitive manner, suggesting that the C-terminus of csPGRMC1 is exposed on the cell surface. This finding reveals that csPGRMC1 has a non-conventional plasma membrane topology, which is different from that of intracellular PGRMC1.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Membrana Celular/metabolismo , Mapeo Epitopo , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Receptores de Progesterona/inmunología , Receptores de Progesterona/metabolismo , Línea Celular , Humanos , Espacio Intracelular/metabolismo , Proteínas de la Membrana/química , Receptores de Progesterona/química
10.
Int J Oncol ; 54(5): 1833-1842, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30816444

RESUMEN

Chemoresistance is one of most critical clinical problems encountered when treating patients with ovarian cancer, due to the fact that the disease is usually diagnosed at advanced stages. Metformin is used as a first­line drug for the treatment of type 2 diabetes; however, drug repositioning studies have revealed its antitumor effects, mainly mediated through AMP­activated protein kinase (AMPK) activation and AKT/mammalian target of rapamycin (mTOR) pathway inhibition in various types of cancer, including drug­resistant cancer cells. The current study revealed that the novel antitumor mechanism of metformin is mediated by regulation of mitochondrial E3 ubiquitin protein ligase 1 (MUL1) expression that negatively regulates AKT. The results demonstrated that metformin decreased the expression of AKT protein levels via MUL1 E3 ligase. In addition, metformin increased both mRNA and protein levels of MUL1 and promoted degradation of AKT in a proteasome­dependent manner. Silencing MUL1 expression suppressed the metformin­mediated AKT degradation and its downstream effects. Cell cycle analysis and a clonogenic assay demonstrated that knockdown of MUL1 significantly diminished the antitumor effects of metformin. Together, these data indicate that MUL1 regulates metformin­mediated AKT degradation and the antitumor effects of metformin in chemoresistant ovarian cancer cell lines.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Metformina/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Reposicionamiento de Medicamentos , Femenino , Humanos , Metformina/farmacología , Ratones , Neoplasias Ováricas/enzimología , Proteolisis , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Microbiol Biotechnol ; 29(11): 1830-1840, 2019 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-31601058

RESUMEN

Loliolide is one of the most ubiquitous monoterpenoid compounds found in algae, and its potential therapeutic effect on various dermatological conditions via agent-induced biological functions, including anti-oxidative and anti-apoptotic properties, was demonstrated. Here, we investigated the effects of loliolide on hair growth in dermal papilla (DP) cells, the main components regulating hair growth and loss conditions. For this purpose, we used a threedimensional (3D) DP spheroid model that mimics the in vivo hair follicle system. Biochemical assays showed that low doses of loliolide increased the viability and size of 3D DP spheroids in a dose-dependent manner. This result correlated with increases in expression levels of hair growth-related autocrine factors including VEGF, IGF-1, and KGF. Immunoblotting and luciferase-reporter assays further revealed that loliolide induced AKT phosphorylation, and this effect led to stabilization of ß-catenin, which plays a crucial role in the hair-inductive properties of DP cells. Further experiments showed that loliolide increased the expression levels of the DP signature genes, ALP, BMP2, VCAN, and HEY1. Furthermore, conditioned media from loliolide-treated DP spheroids significantly enhanced proliferation and the expression of hair growth regulatory genes in keratinocytes. These results suggested that loliolide could function in the hair growth inductivity of DP cells via the AKT/ ß-catenin signaling pathways.


Asunto(s)
Benzofuranos/farmacología , Dermis/efectos de los fármacos , Folículo Piloso/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , beta Catenina/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Dermis/citología , Células HEK293 , Folículo Piloso/crecimiento & desarrollo , Humanos , Queratinocitos/efectos de los fármacos , Monoterpenos/farmacología , Fosforilación/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos
12.
Photochem Photobiol ; 95(2): 595-604, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30266035

RESUMEN

Skin photoaging, which is mainly induced by ultraviolet B (UVB) radiation, is prevented by the application of UV-protective agents. The microalga Nannochloropsis oceanica (N. oceanica) has been primarily reported as a potential biofuel; however, in this study, we investigated whether N. oceanica extracts exerted photoprotective effects against UVB-irradiated human dermal fibroblasts (HDFs) and which single component was responsible for the protective effect of the extracts. Two extracts-pigment and nonpigment-were prepared from N. oceanica biomass. WST-1 assay and expression analysis of interleukin genes showed that the pigment extracts were not significantly cytotoxic to HDFs. Further experiments revealed that treatment with the pigment extract upregulated the expression of collagen genes and significantly blocked UVB-induced damage such as decreased cell viability and increased ROS production. Next, to investigate the pigment composition of the extracts, HPLC analysis was conducted and violaxanthin was identified as the major pigment. The UVB photoprotective effect of the pigment extracts was confirmed in violaxanthin-treated HDFs. In addition, violaxanthin significantly attenuated UVB-induced G1 phase arrest, senescence-associated ß-galactosidase activation, p16 and p21 upregulation, ERK phosphorylation and the downregulation of ECM molecules in HDFs. Therefore, we concluded that violaxanthin was a potential antiphotoaging agent.


Asunto(s)
Algas Marinas/química , Piel/efectos de los fármacos , Piel/efectos de la radiación , Rayos Ultravioleta , Biomasa , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/efectos de la radiación , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Humanos , Piel/citología , Xantófilas/farmacología
13.
J Microbiol Biotechnol ; 28(12): 2121-2132, 2018 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-30415530

RESUMEN

Abnormal melanin synthesis results in several hyperpigmentary disorders such as freckles, melanoderma, age spots, and other related conditions. In this study, we investigated the antimelanogenic effects of an extract from the microalgae Chlamydomonas reinhardtii (CE) and potential mechanisms responsible for its inhibitory effect in B16F10, normal human epidermal melanocyte cells, and human skin-equivalent models. The CE extract showed significant dose-dependent inhibitory effects on α-melanocyte-stimulating, hormone-induced melanin synthesis in cells. Additionally, the CE extract exhibited suppressive effects on the mRNA and protein expression of microphthalmia-associated transcription factor, tyrosinase, tyrosinase-related protein-1, and tyrosinase-related protein-2. The CE extract also inhibited the phosphorylation of protein kinase A and extracellular signal-related kinase, which function as upstream regulators of melanogenesis. Using a three-dimensional, reconstructed pigmented epidermis model, the CE-mediated, anti-pigmentation effects were confirmed by Fontana-Masson staining and melanin content assays. Taken together, CE extract can be used as an anti-pigmentation agent.


Asunto(s)
Chlamydomonas reinhardtii/química , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Pigmentación/efectos de los fármacos , Extractos Vegetales/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Descubrimiento de Drogas , Células Epidérmicas/efectos de los fármacos , Células Epidérmicas/patología , Epidermis/efectos de los fármacos , Epidermis/patología , Quinasas MAP Reguladas por Señal Extracelular , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/farmacología , Oxidorreductasas Intramoleculares/metabolismo , Melaninas/metabolismo , Melanoma , Glicoproteínas de Membrana/metabolismo , Ratones , Factor de Transcripción Asociado a Microftalmía/metabolismo , Monofenol Monooxigenasa/metabolismo , Oxidorreductasas/metabolismo , Fosforilación , ARN Mensajero/efectos de los fármacos , Piel , alfa-MSH/metabolismo
14.
Sci Rep ; 8(1): 3048, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29445107

RESUMEN

Progesterone receptor membrane component 1 (PGRMC1) is a multifunctional heme-binding protein involved in various diseases, including cancers and Alzheimer's disease. Previously, we generated two monoclonal antibodies (MAbs) 108-B6 and 4A68 against surface molecules on human pluripotent stem cells (hPSCs). Here we show that PGRMC1 is the target antigen of both MAbs, and is predominantly expressed on hPSCs and some cancer cells. PGRMC1 is rapidly downregulated during early differentiation of hPSCs. Although PGRMC1 knockdown leads to a spread-out morphology and impaired self-renewal in hPSCs, PGRMC1 knockdown hPSCs do not show apoptosis and autophagy. Instead, PGRMC1 knockdown leads to differentiation of hPSCs into multiple lineage cells without affecting the expression of pluripotency markers. PGRMC1 knockdown increases cyclin D1 expression and decreases Plk1 expression in hPSCs. PGRMC1 knockdown also induces p53 expression and stability, suggesting that PGRMC1 maintains hPSC self-renewal through suppression of p53-dependent pathway. Analysis of signaling molecules further reveals that PGRMC1 knockdown promotes inhibitory phosphorylation of GSK-3ß and increased expression of Wnt3a and ß-catenin, which leads to activation of Wnt/ß-catenin signaling. The results suggest that PGRMC1 suppresses the p53 and Wnt/ß-catenin pathways to promote self-renewal and inhibit early differentiation in hPSCs.


Asunto(s)
Autorrenovación de las Células/fisiología , Proteínas de la Membrana/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Receptores de Progesterona/metabolismo , Vía de Señalización Wnt/fisiología , Apoptosis/fisiología , Diferenciación Celular/fisiología , Linaje de la Célula , Proliferación Celular/fisiología , Regulación hacia Abajo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HEK293 , Células Madre Embrionarias Humanas , Humanos , Fosforilación , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA