• 제목/요약/키워드: Forkhead Transcription Factor O1

검색결과 8건 처리시간 0.025초

Molecular Cloning and Expression of Forkhead Transcription Factor O1 Gene from Pig Sus scrofa

  • Pang, Weijun;Sun, Shiduo;Bai, Liang;Yang, Gongshe
    • Asian-Australasian Journal of Animal Sciences
    • /
    • 제21권4호
    • /
    • pp.499-509
    • /
    • 2008
  • Foxo1 plays an important role in the integration of hormone-activated signaling pathways with the complex transcriptional cascade that promotes preadipocyte differentiation of clonal cell lines from rodents. We isolated the full-length cDNA of porcine FoxO1 gene using RACE, confirmed by visual Northern blotting. The deduced amino acids indicated 94% and 90% identities with the corresponding human and mice aa. Analysis of the aa sequence, showed that it included a Forkhead domain (aa 167-247), a transmembrane structure domain (aa 90-113), a LXXLL motif (aa 469-473), and 51 Ser, 8 Thr, and 4 Tyr phosphorylation sites, indicating a potential important role for FoxO1 transcriptional activity in vivo. Using the IMpRH panel, we mapped FoxO1 gene to chromosome 11p13. Our data provide basic molecular information useful for the further investigation on the function of FoxO1 gene. Time-course analysis of FoxO1 expressions indicated that levels of mRNA and protein gradually increased from day 0 to 3, and it reached almost maximal level at day 3, then decreased from day 5 to 7 in porcine primary preadipocyte differentiation. After induction by IGF-1, GPDH activity and accumulation of lipid increased, however, expressions of FoxO1 mRNA and protein were inhibited in a dose dependent manner. These results suggest that FoxO1 takes part in porcine preadipocyte differentiation and expressions of FoxO1 were regulated by IGF-1.

FoxO3a mediates transforming growth factor-β1-induced apoptosis in FaO rat hepatoma cells

  • Kim, Byung-Chul
    • BMB Reports
    • /
    • 제41권10호
    • /
    • pp.728-732
    • /
    • 2008
  • FoxO3a is a member of the forkhead box class O (FoxO) transcription factor family and an important regulator of apoptosis. This work aimed to elucidate the involvement of FoxO3a in transforming growth factor-${\beta}1$(TGF-${\beta}1$)-induced apoptosis in FaO rat hepatoma cells. TGF-${\beta}1$ caused a time-dependent activation of FoxO3a and a subsequent increase in FoxO response-element-containing luciferase reporter activity, which was Akt-sensitive. The FaO cells stably transfected with a wild type FoxO3a were more susceptible to the formation of apoptotic bodies, populations of sub-G1 apoptotic cells, and collapse of the mitochondrial-membrane potential triggered by TGF-${\beta}1$. In contrast, transfection with small-interfering RNA (siRNA) oligonucleotide specific for FoxO3a significantly inhibited caspase activation in FaO cells treated with TGF-${\beta}1$. It thus appears that FoxO3a plays a crucial mediatory role in the TGF-${\beta}1$ signaling pathway leading to apoptosis.

Exploring the Potential of Natural Products as FoxO1 Inhibitors: an In Silico Approach

  • Anugya Gupta;Rajesh Haldhar;Vipul Agarwal;Dharmendra Singh Rajput;Kyung-Soo Chun;Sang Beom Han;Vinit Raj;Sangkil Lee
    • Biomolecules & Therapeutics
    • /
    • 제32권3호
    • /
    • pp.390-398
    • /
    • 2024
  • FoxO1, a member of the Forkhead transcription factor family subgroup O (FoxO), is expressed in a range of cell types and is crucial for various pathophysiological processes, such as apoptosis and inflammation. While FoxO1's roles in multiple diseases have been recognized, the target has remained largely unexplored due to the absence of cost-effective and efficient inhibitors. Therefore, there is a need for natural FoxO1 inhibitors with minimal adverse effects. In this study, docking, MMGBSA, and ADMET analyses were performed to identify natural compounds that exhibit strong binding affinity to FoxO1. The top candidates were then subjected to molecular dynamics (MD) simulations. A natural product library was screened for interaction with FoxO1 (PDB ID-3CO6) using the Glide module of the Schrödinger suite. In silico ADMET profiling was conducted using SwissADME and pkCSM web servers. Binding free energies of the selected compounds were assessed with the Prime-MMGBSA module, while the dynamics of the top hits were analyzed using the Desmond module of the Schrödinger suite. Several natural products demonstrated high docking scores with FoxO1, indicating their potential as FoxO1 inhibitors. Specifically, the docking scores of neochlorogenic acid and fraxin were both below -6.0. These compounds also exhibit favorable drug-like properties, and a 25 ns MD study revealed a stable interaction between fraxin and FoxO1. Our findings highlight the potential of various natural products, particularly fraxin, as effective FoxO1 inhibitors with strong binding affinity, dynamic stability, and suitable ADMET profiles.

마우스 C2C12 근관세포에서 AICAR로 유도된 근위축에 미치는 오미자 추출물의 영향 (Ethanol Extract of Schisandra chinensis (Turcz.) Baill. Reduces AICAR-induced Muscle Atrophy in C2C12 Myotubes)

  • 강영순;한민호;박철;홍수현;황혜진;김병우;김철민;최영현
    • 생명과학회지
    • /
    • 제25권3호
    • /
    • pp.293-298
    • /
    • 2015
  • 근위축은 근육 단백질 합성의 저하와 근육 단백질의 분해 증가에 따른 근섬유의 감소에 의한 근육량이 감소되는 현상이다. 오미자(Schisandrae Fructus, fruits of Schisandra chinensis (Turcz.) Baillon)는 오랫동안 전통의학에서 강장제로서 널리 사용되어 왔다. 비록 다양한 질병 연관 오미자의 생리활성 효능이 폭넓게 연구되어져 왔으나 근육 질환 관련 연구는 매우 제한적으로 이루어져 왔다. 본 연구에서는 오미자 에탄올 추출물(SF)이 AMPK 활성인자 AICAR 처리에 의한 C2C12 근관세포의 근위축 모델계를 이용하여 근위축 억제 효능을 가지는지의 여부와 관련 기전의 해석을 시도하였다. AICAR 처리는 근단백질 분해 연관 ubiquitin ligase muscle RING finger-1 (MuRF-1)의 발현을 전사 수준에서 증가시켰고, MuRF-1 조절 전사인자의 하나인 forkhead box O3a (FoxO3a) 단백질의 인산화를 증가시켰으며, 이러한 변화는 근위축과 연관된 C2C12 근관세포의 형태적 변형과 동반된 현상이었다. 그러나 SF의 전처리에 의하여, AICAR에 의하여 유도된 근위축성 형태변화를 억제하였으며, MuRF-1의 발현과 FoxO3a의 활성화를 억제시켰다. 본 연구의 결과는 SF가 AICAR 처리에 의한 C2C12 근관세포의 근위축을 AMPK 및 FoxO3a 신호전달계 조절을 통하여 억제하였음을 보여주는 것으로 오미자는 근기능 향상을 위한 식의 약 소재로서의 개발 가능성이 매우 높음을 시사하여 준다.

Human Norovirus Replication in Temperature-Optimized MDCK Cells by Forkhead Box O1 Inhibition

  • Jeong, Eun-Hye;Cho, Se-Young;Vaidya, Bipin;Ha, Sang Hoon;Jun, Sangmi;Ro, Hyun-Joo;Lee, Yujeong;Lee, Juhye;Kwon, Joseph;Kim, Duwoon
    • Journal of Microbiology and Biotechnology
    • /
    • 제30권9호
    • /
    • pp.1412-1419
    • /
    • 2020
  • Human noroviruses (HuNoVs) are a leading cause of gastroenteritis outbreaks worldwide. However, the paucity of appropriate cell culture models for HuNoV replication has prevented developing effective anti-HuNoV therapies. In this study, first, the replication of the virus at various temperatures in different cells was compared, which showed that lowering the culture temperature from 37℃ significantly increased virus replication in Madin-Darby canine kidney (MDCK) cells. Second, the expression levels of autophagy-, immune-, and apoptosis-related genes at 30℃ and 37℃ were compared to explore factors affecting HuNoV replication. HuNoV cultured at 37℃ showed significantly increased autophagy-related genes (ATG5 and ATG7) and immune-related genes (IFNA, IFNB, ISG15, and NFKB) compared to mock. However, the virus cultured at 30℃ showed significantly decreased expression of autophagy-related genes (ATG5 and ATG7), but not significantly different major immune-related genes (IFNA, ISG15, and NFKB) compared to mock. Importantly, expression of the transcription factor FOXO1, which controls autophagy- and immune-related gene expression, was significantly lower at 30℃. Moreover, FOXO1 inhibition in temperature-optimized MDCK cells enhanced HuNoV replication, highlighting FOXO1 inhibition as an approach for successful virus replication. In the temperature-optimized cells, various HuNoV genotypes were successfully replicated, with GI.8 showing the highest replication levels followed by GII.1, GII.3, and GII.4. Furthermore, ultrastructural analysis of the infected cells revealed functional HuNoV replication at low temperature, with increased cellular apoptosis and decreased autophagic vacuoles. In conclusion, temperature-optimized MDCK cells can be used as a convenient culture model for HuNoV replication by inhibiting FOXO1 and providing adaptability to different genotypes.

Role of stearyl-coenzyme A desaturase 1 in mediating the effects of palmitic acid on endoplasmic reticulum stress, inflammation, and apoptosis in goose primary hepatocytes

  • Tang, Bincheng;Qiu, Jiamin;Hu, Shenqiang;Li, Liang;Wang, Jiwen
    • Animal Bioscience
    • /
    • 제34권7호
    • /
    • pp.1210-1220
    • /
    • 2021
  • Objective: Unlike mammals, goose fatty liver shows a strong tolerance to fatty acids without obvious injury. Stearyl-coenzyme A desaturase 1 (SCD1) serves crucial role in desaturation of saturated fatty acids (SAFs), but its role in the SAFs tolerance of goose hepatocytes has not been reported. This study was conducted to explore the role of SCD1 in regulating palmitic acid (PA) tolerance of goose primary hepatocytes. Methods: 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide was examined to reflect the effect of PA on hepatocytes viability, and quantitative polymerase chain reaction was used to detect the mRNA levels of several genes related to endoplasmic reticulum (ER) stress, inflammation, and apoptosis, and the role of SCD1 in PA tolerance of goose hepatocytes was explored using RNA interfere. Results: Our results indicated that goose hepatocytes exhibited a higher tolerant capacity to PA than human hepatic cell line (LO2 cells). In goose primary hepatocytes, the mRNA levels of fatty acid desaturation-related genes (SCD1 and fatty acid desaturase 2) and fatty acid elongate enzyme-related gene (elongase of very long chain fatty acids 6) were significantly upregulated with 0.6 mM PA treatment. However, in LO2 cells, expression of ER stress-related genes (x box-binding protein, binding immunoglobulin protein, and activating transcription factor 6), inflammatory response-related genes (interleukin-6 [IL-6], interleukin-1β [IL-1β], and interferon-γ) and apoptosis-related genes (bcl-2-associated X protein, b-cell lymphoma 2, Caspase-3, and Caspase-9) was significantly enhanced with 0.6 mM PA treatment. Additionally, small interfering RNA (siRNA) mediated downregulation of SCD1 significantly reduced the PA tolerance of goose primary hepatocytes under the treatment of 0.6 mM PA; meanwhile, the mRNA levels of inflammatory-related genes (IL-6 and IL-1β) and several key genes involved in the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT), forkhead box O1 (FoxO1), mammalian target of rapamycin and AMPK pathways (AKT1, AKT2, FoxO1, and sirtuin 1), as well as the protein expression of cytochrome C and the apoptosis rate were upregulated. Conclusion: In conclusion, our data suggested that SCD1 was involved in enhancing the PA tolerance of goose primary hepatocytes by regulating inflammation- and apoptosis-related genes expression.

Novel SIRT Inhibitor, MHY2256, Induces Cell Cycle Arrest, Apoptosis, and Autophagic Cell Death in HCT116 Human Colorectal Cancer Cells

  • Kim, Min Jeong;Kang, Young Jung;Sung, Bokyung;Jang, Jung Yoon;Ahn, Yu Ra;Oh, Hye Jin;Choi, Heejeong;Choi, Inkyu;Im, Eunok;Moon, Hyung Ryong;Chung, Hae Young;Kim, Nam Deuk
    • Biomolecules & Therapeutics
    • /
    • 제28권6호
    • /
    • pp.561-568
    • /
    • 2020
  • We examined the anticancer effects of a novel sirtuin inhibitor, MHY2256, on HCT116 human colorectal cancer cells to investigate its underlying molecular mechanisms. MHY2256 significantly suppressed the activity of sirtuin 1 and expression levels of sirtuin 1/2 and stimulated acetylation of forkhead box O1, which is a target protein of sirtuin 1. Treatment with MHY2256 inhibited the growth of the HCT116 (TP53 wild-type), HT-29 (TP53 mutant), and DLD-1 (TP53 mutant) human colorectal cancer cell lines. In addition, MHY2256 induced G0/G1 phase arrest of the cell cycle progression, which was accompanied by the reduction of cyclin D1 and cyclin E and the decrease of cyclin-dependent kinase 2, cyclin-dependent kinase 4, cyclin-dependent kinase 6, phosphorylated retinoblastoma protein, and E2F transcription factor 1. Apoptosis induction was shown by DNA fragmentation and increase in late apoptosis, which were detected using flow cytometric analysis. MHY2256 downregulated expression levels of procaspase-8, -9, and -3 and led to subsequent poly(ADP-ribose) polymerase cleavage. MHY2256-induced apoptosis was involved in the activation of caspase-8, -9, and -3 and was prevented by pretreatment with Z-VAD-FMK, a pan-caspase inhibitor. Furthermore, the autophagic effects of MHY2256 were observed as cytoplasmic vacuolation, green fluorescent protein-light-chain 3 punctate dots, accumulation of acidic vesicular organelles, and upregulated expression level of light-chain 3-II. Taken together, these results suggest that MHY2256 could be a potential novel sirtuin inhibitor for the chemoprevention or treatment of colorectal cancer or both.

C2C12 근관세포에서 상엽에 의한 AMPK의 불활성화와 AICAR로 유도된 근위축 억제의 연관성에 관한 연구 (Ethanol Extract of Mori Folium Inhibits AICAR-induced Muscle Atrophy Through Inactivation of AMPK in C2C12 Myotubes)

  • 이유성;김홍재;정진우;한민호;홍수현;최영현;박철
    • 생명과학회지
    • /
    • 제28권4호
    • /
    • pp.435-443
    • /
    • 2018
  • AMPK는 세포 내 에너지 균형을 조절하는 조절자 및 에너지 센서이며, 특히 골격근에서는 muscle-specific ubiquitin ligases의 조절을 통한 근육 단백질 분해를 증가시키는 것으로 알려져 있다. 한편 상엽은 다양한 약리학적 효능을 가지는 전통약재 중 하나이지만 근위축과 관련된 효능에 대해서는 거의 알려져 있지 않다. 본 연구에서는 C2C12 myotubes에서 AMPK 활성제인 AICAR가 유발하는 근위축 및 관련 유전자의 발현과 함께 상엽 에탄올 추출물(ethanol extracts of Mori Folium, EEMF)이 유발하는 근위축 억제 효능에 대해서 조사하였다. 먼저 C2C12 myoblasts에 AICAR를 처리하였을 경우 AMPK 활성화가 유발되었으며, 하위 단계에 있는 FoxO3a의 발현 증가와 함께 muscle-specific ubiquitin ligases인 MAFbx/atrogin-1 및 MuRF1의 발현 증가와 muscle-specific transcription factors인 MyoD 및 myogenin의 발현 감소가 유발되었다. 또한 분화가 유발된 C2C12 myotubes에 세포독성이 없는 조건의 AICAR를 처리하였을 경우 근위축이 유발되었으며, EEMF는 AMPK 불활성화 및 FoxO3a 발현 억제를 유발함으로서 AICAR 처리에 의한 근위축을 억제하는 것으로 나타났다. 본 연구 결과에서 AICAR에 의한 AMPK 활성화가 근위축을 유발한다는 것을 알 수 있었으며, EEMF는 AMPK signaling pathway를 통하여 AICAR에 의한 근위축을 억제한다는 것을 알 수 있었다.