Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.363
Filtrar
1.
Sci Rep ; 14(1): 19456, 2024 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-39169157

RESUMO

Approximately 80% of pediatric tumors occur in low- and middle-income countries (LMIC), where diagnostic tools essential for treatment decisions are often unavailable or incomplete. Development of cost-effective molecular diagnostics will help bridge the cancer diagnostic gap and ultimately improve pediatric cancer outcomes in LMIC settings. We investigated the feasibility of using nanopore whole transcriptome sequencing on formalin-fixed paraffin embedded (FFPE)-derived RNA and a composite machine learning model for pediatric solid tumor diagnosis. Transcriptome cDNA sequencing was performed on a heterogenous set of 221 FFPE and 32 fresh frozen pediatric solid tumor and lymphoma specimens on Oxford Nanopore Technologies' sequencing platforms. A composite machine learning model was then used to classify transcriptional profiles into clinically actionable tumor types and subtypes. In total, 95.6% and 89.7% of pediatric solid tumors and lymphoma specimens were correctly classified, respectively. 71.5% of pediatric solid tumors had prediction probabilities > 0.8 and were classified with 100% accuracy. Similarly, for lymphomas, 72.4% of samples that had prediction probabilities > 0.6 were classified with 97.6% accuracy. Additionally, FOXO1 fusion status was predicted accurately for 97.4% of rhabdomyosarcomas and MYCN amplification was predicted with 88% accuracy in neuroblastoma. Whole transcriptome sequencing from FFPE-derived pediatric solid tumor and lymphoma samples has the potential to provide clinical classification of both tissue lineage and core genomic classification. Further expansion, refinement, and validation of this approach is necessary to explore whether this technology could be part of the solution of addressing the diagnostic limitations in LMIC.


Assuntos
Perfilação da Expressão Gênica , Linfoma , Humanos , Criança , Linfoma/genética , Linfoma/diagnóstico , Linfoma/classificação , Perfilação da Expressão Gênica/métodos , Transcriptoma , Aprendizado de Máquina , Neoplasias/genética , Neoplasias/diagnóstico , Neoplasias/classificação , Pré-Escolar , Masculino , Feminino , Proteína Forkhead Box O1/genética , Rabdomiossarcoma/genética , Rabdomiossarcoma/diagnóstico , Rabdomiossarcoma/classificação , Biomarcadores Tumorais/genética , Adolescente , Lactente
2.
Aging (Albany NY) ; 16(15): 11568-11576, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-39103205

RESUMO

Osteosarcoma is a highly metastatic, aggressive bone cancer that occurs in children and young adults worldwide. Circular RNAs (circRNAs) are crucial molecules for osteosarcoma progression. In this study, we aimed to investigate the impact of circMRPS35 overexpression and its interaction with FOXO1 via evaluating apoptosis, cell cycle, and bioinformatic analyses on the malignant development of osteosarcoma in MG63 and MNNG/HOS cells. We found that circMRPS35 overexpression reduced osteosarcoma cell viability and inhibited tumor growth in vivo. It increased the apoptosis rate and induced cell cycle arrest in osteosarcoma cells. We identified a potential interaction between circMRPS35 and FOXO1 with miR-105-5p using bioinformatics analysis. Overexpression of circMRPS35 decreased miR-105-5p expression, whereas miR-105-5p mimic treatment increased its expression. This mimic also suppressed the luciferase activity of circMRPS35 and FOXO1 and reduced FOXO1 expression. Overexpression of circMRPS35 elevated FOXO1 protein levels, but this effect was reversed by co-treatment with the miR-105-5p mimic. We demonstrated that inhibiting miR-105-5p decreased viability and induced apoptosis. Overexpression of FOXO1 or treatment with a miR-105-5p inhibitor could counteract the effects of circMRPS35 on viability and apoptosis in osteosarcoma cells. Therefore, we concluded that circMRPS35 suppressed the malignant progression of osteosarcoma via targeting the miR-105-5p/FOXO1 axis.


Assuntos
Apoptose , Neoplasias Ósseas , Proteína Forkhead Box O1 , Regulação Neoplásica da Expressão Gênica , MicroRNAs , Osteossarcoma , RNA Circular , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/patologia , RNA Circular/genética , RNA Circular/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Humanos , Linhagem Celular Tumoral , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Apoptose/genética , Camundongos , Progressão da Doença , Proliferação de Células/genética , Camundongos Nus , Sobrevivência Celular/genética
3.
Cell Mol Life Sci ; 81(1): 360, 2024 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-39158700

RESUMO

Iron deficiency is a prevalent nutritional deficit associated with organ damage and dysfunction. Recent research increasingly associates iron deficiency with bone metabolism dysfunction, although the precise underlying mechanisms remain unclear. Some studies have proposed that iron-dependent methylation-erasing enzyme activity regulates cell proliferation and differentiation under physiological or pathological conditions. However, it remains uncertain whether iron deficiency inhibits the activation of quiescent mesenchymal stem cells (MSCs) by affecting histone demethylase activity. In our study, we identified KDM4D as a key player in the activation of quiescent MSCs. Under conditions of iron deficiency, the H3K9me3 demethylase activity of KDM4D significantly decreased. This alteration resulted in increased heterochromatin with H3K9me3 near the PIK3R3 promoter, suppressing PIK3R3 expression and subsequently inhibiting the activation of quiescent MSCs via the PI3K-Akt-Foxo1 pathway. Iron-deficient mice displayed significantly impaired bone marrow MSCs activation and decreased bone mass compared to normal mice. Modulating the PI3K-Akt-Foxo1 pathway could reverse iron deficiency-induced bone loss.


Assuntos
Proteína Forkhead Box O1 , Ferro , Histona Desmetilases com o Domínio Jumonji , Células-Tronco Mesenquimais , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Animais , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Camundongos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Ferro/metabolismo , Camundongos Endogâmicos C57BL , Proliferação de Células , Diferenciação Celular , Masculino , Deficiências de Ferro , Humanos
4.
Pol J Pathol ; 75(2): 83-96, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39166517

RESUMO

There is growing evidence that the KDM5 family of histone demethylases plays a causal role in human cancer. However, few studies have been reported on the KDM5 family in endometrial carcinoma (EC). Moreover, it was found that there was some correlation between the KDM5 family and FOXO1 in EC. The current study was performed to explore the expressions of KDM5A, KDM5B, and FOXO1 in endometrioid adenocarcinoma detected by immunohistochemistry; paracancer endometrium, simple hyperplastic endometrium, and normal endometrium were used as control groups to explore the possible diagnostic value of KDM5A and KDM5B expression in endometrioid adenocarcinoma, with the aim of evaluating the potential of this marker in predicting the prognosis of endometrioid adenocarcinoma.


Assuntos
Biomarcadores Tumorais , Carcinoma Endometrioide , Neoplasias do Endométrio , Proteína Forkhead Box O1 , Imuno-Histoquímica , Histona Desmetilases com o Domínio Jumonji , Humanos , Neoplasias do Endométrio/patologia , Neoplasias do Endométrio/metabolismo , Feminino , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/metabolismo , Pessoa de Meia-Idade , Histona Desmetilases com o Domínio Jumonji/metabolismo , Carcinoma Endometrioide/patologia , Carcinoma Endometrioide/metabolismo , Adulto , Idoso , Prognóstico , Proteína 2 de Ligação ao Retinoblastoma/metabolismo , Proteína 2 de Ligação ao Retinoblastoma/análise , Relevância Clínica , Proteínas Nucleares , Proteínas Repressoras
5.
J Cell Mol Med ; 28(16): e70021, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39153212

RESUMO

Accumulating evidence has demonstrated that F-box protein 22 (FBXO22) participates in tumour development and progression in various types of human malignancies. However, the functions and detailed molecular mechanisms of FBXO22 in osteosarcoma tumorigenesis and progression remain elusive. In this study, we aimed to determine the effects of FBXO22 on the cell proliferation, migration and invasion of osteosarcoma cells using cell counting kit-8 and Matrigel Transwell approaches. Moreover, we explored the molecular mechanisms by which FBXO22 mediated oncogenesis and progression in osteosarcoma via Western blotting, immunoprecipitation and ubiquitination. We found that FBXO22 depletion inhibited the proliferation, migration and invasion of osteosarcoma cells, whereas FBXO22 overexpression increased the proliferation and motility of osteosarcoma cells. Mechanistically, FBXO22 promoted the ubiquitination and degradation of FoxO1 in osteosarcoma cells. FBXO22 depletion reduced cell proliferation and motility via regulation of FoxO1. Taken together, our findings provide new insight into FBXO22-induced osteosarcoma tumorigenesis. The inhibition of FBXO22 could be a promising strategy for the treatment of osteosarcoma.


Assuntos
Movimento Celular , Proliferação de Células , Proteínas F-Box , Proteína Forkhead Box O1 , Regulação Neoplásica da Expressão Gênica , Osteossarcoma , Ubiquitinação , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Osteossarcoma/genética , Humanos , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Proteínas F-Box/metabolismo , Proteínas F-Box/genética , Movimento Celular/genética , Linhagem Celular Tumoral , Proteólise , Progressão da Doença , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Neoplasias Ósseas/genética , Invasividade Neoplásica , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Receptores Citoplasmáticos e Nucleares
6.
Stem Cell Res Ther ; 15(1): 261, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-39148121

RESUMO

BACKGROUND: Human adipose-derived stem cells (ADSCs) exert a strong anti-inflammatory effect, and synovium-derived stem cells (SDSCs) have high chondrogenic potential. Thus, this study aims to investigate whether a combination of human ADSCs and SDSCs will have a synergistic effect that will increase the chondrogenic potential of osteoarthritis (OA) chondrocytes in vitro and attenuate the cartilage degeneration of early and advanced OA in vitro. METHODS: ADSCs, SDSCs, and chondrocytes were isolated from OA patients who underwent total knee arthroplasty. The ADSCs-SDSCs mixed cell ratios were 1:0 (ADSCs only), 8:2, 5:5 (5A5S), 2:8, and 0:1 (SDSCs only). The chondrogenic potential of the OA chondrocytes was evaluated in vitro with a transwell assay or pellet culture with various mixed cell groups. The mixed cell group with the highest chondrogenic potential was then selected and injected into the knee joints of nude rats of early and advanced OA stages in vivo. The animals were then evaluated 12 and 20 weeks after surgery through gait analysis, von frey test, microcomputed tomography, MRI, and immunohistochemical and histological analyses. Finally, the mechanisms underlying these findings were investigated through the RNA sequencing of tissue samples in vivo and Western blot of the OA chondrocyte autophagy pathway. RESULTS: Among the MSCs treatment groups, 5A5S had the greatest synergistic effect that increased the chondrogenic potential of OA chondrocytes in vitro and inhibited early and advanced OA in vivo. The 5A5S group significantly reduced cartilage degeneration, synovial inflammation, pain sensation, and nerve invasion in subchondral nude rat OA, outperforming both single-cell treatments. The underlying mechanism was the activation of chondrocyte autophagy via the FoxO1 signaling pathway. CONCLUSION: A combination of human ADSCs and SDSCs demonstrated higher potential than a single type of stem cell, demonstrating potential as a novel treatment for OA.


Assuntos
Autofagia , Condrócitos , Proteína Forkhead Box O1 , Células-Tronco Mesenquimais , Osteoartrite , Transdução de Sinais , Humanos , Condrócitos/metabolismo , Animais , Ratos , Osteoartrite/terapia , Osteoartrite/metabolismo , Osteoartrite/patologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Proteína Forkhead Box O1/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Masculino , Ratos Nus , Condrogênese , Membrana Sinovial/metabolismo , Membrana Sinovial/citologia , Pessoa de Meia-Idade , Feminino
7.
Sci Adv ; 10(33): eado1533, 2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-39151008

RESUMO

Chronic stress-induced epinephrine (EPI) accelerates breast cancer progression and metastasis, but the molecular mechanisms remain unclear. Herein, we found a strong positive correlation between circulating EPI levels and the tumoral expression of ubiquitin-specific peptidase 22 (USP22) in patients with breast cancer. USP22 facilitated EPI-induced breast cancer progression and metastasis by enhancing adipose triglyceride lipase (ATGL)-mediated lipolysis. Targeted USP22 deletion decreased ATGL expression and lipolysis, subsequently inhibiting EPI-mediated breast cancer lung metastasis. USP22 acts as a bona fide deubiquitinase for the Atgl gene transcription factor FOXO1, and EPI architects a lipolysis signaling pathway to stabilize USP22 through AKT-mediated phosphorylation. Notably, USP22 phosphorylation levels are positively associated with EPI and with downstream pathways involving both FOXO1 and ATGL in breast cancers. Pharmacological USP22 inhibition synergized with ß-blockers in treating preclinical xenograft breast cancer models. This study reveals a molecular pathway behind EPI's tumor-promoting effects and provides a strong rationale for combining USP22 inhibition with ß-blockers to treat aggressive breast cancer.


Assuntos
Neoplasias da Mama , Epinefrina , Lipólise , Ubiquitina Tiolesterase , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/tratamento farmacológico , Lipólise/efeitos dos fármacos , Feminino , Ubiquitina Tiolesterase/metabolismo , Ubiquitina Tiolesterase/genética , Epinefrina/metabolismo , Humanos , Animais , Camundongos , Linhagem Celular Tumoral , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Lipase/metabolismo , Lipase/genética , Transdução de Sinais/efeitos dos fármacos , Metástase Neoplásica , Fosforilação , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Aciltransferases
8.
J Transl Med ; 22(1): 782, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39175012

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease worldwide. Osteocalcin plays an important role in energy metabolism. In this study, we investigated the mechanism of action of chemically synthesized osteocalcin (csOCN) in ameliorating NAFLD. We demonstrated for the first time that csOCN attenuates lipid accumulation in the liver and hepatocytes by modulating CD36 protein expression. In addition, we found that the expression of p-AMPK, FOXO1 and BCL6 decreased and the expression of CD36 increased after OA/PA induction compared to the control group, and these effects were reversed by the addition of csOCN. In contrast, the therapeutic effect of csOCN was inhibited by the addition of AMPK inhibitors and BCL6 inhibitors. This finding suggested that csOCN regulates CD36 expression via the AMPK-FOXO1/BCL6 axis. In NAFLD mice, oral administration of csOCN also activated the AMPK pathway and reduced CD36 expression. Molecular docking revealed that osteocalcin has a docking site with CD36. Compared to oleic acid and palmitic acid, osteocalcin bound more strongly to CD36. Laser confocal microscopy results showed that osteocalcin colocalized with CD36 at the cell membrane. In conclusion, we demonstrated the regulatory role of csOCN in fatty acid uptake pathways for the first time; it regulates CD36 expression via the AMPK-FOXO1/BCL6 axis to reduce fatty acid uptake, and it affects fatty acid transport by may directly binding to CD36. There are indications that csOCN has potential as a CD36-targeted drug for the treatment of NAFLD.


Assuntos
Proteínas Quinases Ativadas por AMP , Antígenos CD36 , Proteína Forkhead Box O1 , Hepatopatia Gordurosa não Alcoólica , Osteocalcina , Proteínas Proto-Oncogênicas c-bcl-6 , Transdução de Sinais , Animais , Humanos , Masculino , Camundongos , Proteínas Quinases Ativadas por AMP/metabolismo , Antígenos CD36/metabolismo , Proteína Forkhead Box O1/metabolismo , Fígado/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Osteocalcina/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Arterioscler Thromb Vasc Biol ; 44(9): 2004-2023, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39087350

RESUMO

BACKGROUND: Lymphatic valves are specialized structures in collecting lymphatic vessels and are crucial for preventing retrograde lymph flow. Mutations in valve-forming genes have been clinically implicated in the pathology of congenital lymphedema. Lymphatic valves form when oscillatory shear stress from lymph flow signals through the PI3K/AKT pathway to promote the transcription of valve-forming genes that trigger the growth and maintenance of lymphatic valves. Conventionally, in many cell types, AKT is phosphorylated at Ser473 by the mTORC2 (mammalian target of rapamycin complex 2). However, mTORC2 has not yet been implicated in lymphatic valve formation. METHODS: In vivo and in vitro techniques were used to investigate the role of Rictor, a critical component of mTORC2, in lymphatic endothelium. RESULTS: Here, we showed that embryonic and postnatal lymphatic deletion of Rictor, a critical component of mTORC2, led to a significant decrease in lymphatic valves and prevented the maturation of collecting lymphatic vessels. RICTOR knockdown in human dermal lymphatic endothelial cells not only reduced the level of activated AKT and the expression of valve-forming genes under no-flow conditions but also abolished the upregulation of AKT activity and valve-forming genes in response to oscillatory shear stress. We further showed that the AKT target, FOXO1 (forkhead box protein O1), a repressor of lymphatic valve formation, had increased nuclear activity in Rictor knockout mesenteric lymphatic endothelial cells in vivo. Deletion of Foxo1 in Rictor knockout mice restored the number of valves to control levels in lymphatic vessels of the ear and mesentery. CONCLUSIONS: Our work identifies a novel role for RICTOR in the mechanotransduction signaling pathway, wherein it activates AKT and prevents the nuclear accumulation of the valve repressor, FOXO1, which ultimately enables the formation and maintenance of lymphatic valves.


Assuntos
Proteínas de Transporte , Proteína Forkhead Box O1 , Linfangiogênese , Vasos Linfáticos , Alvo Mecanístico do Complexo 2 de Rapamicina , Mecanotransdução Celular , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt , Proteína Companheira de mTOR Insensível à Rapamicina , Transdução de Sinais , Animais , Proteína Companheira de mTOR Insensível à Rapamicina/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Vasos Linfáticos/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/genética , Humanos , Proteínas de Transporte/metabolismo , Proteínas de Transporte/genética , Células Endoteliais/metabolismo , Células Cultivadas , Serina-Treonina Quinases TOR/metabolismo , Fosforilação , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição Forkhead/genética , Camundongos , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/genética , Camundongos Endogâmicos C57BL , Interferência de RNA , Transfecção
10.
Int J Immunopathol Pharmacol ; 38: 3946320241272642, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39096175

RESUMO

Prolonged exposure to different occupational or environmental toxicants triggered oxidative stress and inflammatory reactions mediated lung damage. This study was designed to explore the influence and protective impact of flavone on lung injury in rats intoxicated with nicotine (NIC) and exposed to radiation (IR). Forty rats were divided into four groups; group I control, group II flavone; rats were administered with flavone (25 mg/kg/day), group III NIC + IR; rats were injected intraperitoneally with NIC (1 mg/kg/day) and exposed to γ-IR (3.5 Gy once/week for 2 weeks) while group IV NIC + IR + flavone; rats were injected with NIC, exposed to IR and administered with flavone. Redox status parameters and histopathological changes in lung tissue were evaluated. Nuclear factor-kappa B (NF-κB), forkhead box O-class1 (FoxO1) and nucleotide-binding domain- (NOD-) like receptor pyrin domain-containing-3 (NLRP3) gene expression were measured in lung tissues. Moreover, nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and phosphatidylinositol three kinase (PI3K) were measured using ELISA kits. Our data demonstrates, for the first time, that flavone protects the lung from NIC/IR-associated cytotoxicity, by attenuating the disrupted redox status and aggravating the antioxidant defence mechanism via activation of the PI3K/Nrf2. Moreover, flavone alleviates pulmonary inflammation by inhibiting the inflammatory signaling pathway FOXO1/NF-κB/NLRP3- Inflammasome. Collectively, the obtained results exhibited a notable efficiency of flavone in alleviating lung injury induced by NIC and IR via modulating PI3K/Nrf2 and FoxO1/NLRP3 Inflammasome.


Assuntos
Flavonas , Inflamassomos , Lesão Pulmonar , Nicotina , Animais , Masculino , Ratos , Flavonas/farmacologia , Proteína Forkhead Box O1 , Raios gama , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/efeitos da radiação , Lesão Pulmonar/metabolismo , Lesão Pulmonar/prevenção & controle , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Nicotina/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
11.
Cell Commun Signal ; 22(1): 396, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39138534

RESUMO

BACKGROUND: Secreted frizzled-related proteins (SFRPs) comprise a family of WNT signaling antagonists whose roles in the ovary are poorly understood. Sfrp4-null mice were previously found to be hyperfertile due to an enhanced granulosa cell response to gonadotropins, leading to decreased antral follicle atresia and enhanced ovulation rates. The present study aimed to elucidate the mechanisms whereby SFRP4 antagonizes FSH action. METHODS: Primary cultures of granulosa cells from wild-type mice were treated with FSH and/or SFRP4, and effects of treatment on gene expression were evaluated by RT-qPCR and RNAseq. Bioinformatic analyses were conducted to analyse the effects of SFRP4 on the transcriptome, and compare them to those of FSH or a constitutively active mutant of FOXO1. Additional granulosa cell cultures from wild-type or Sfrp4-null mice, some pretreated with pharmacologic inhibitors of specific signaling effectors, were used to examine the effects of FSH and/or SFRP4 on signaling pathways, autophagy and apoptosis by western blotting and TUNEL. RESULTS: Treatment of cultured granulosa cells with recombinant SFRP4 was found to decrease basal and FSH-stimulated mRNA levels of FSH target genes. Unexpectedly, this effect was found to occur neither via a canonical (CTNNB1-dependent) nor non-canonical WNT signaling mechanism, but was found to be GSK3ß-dependent. Rather, SFRP4 was found to antognize AKT activity via a mechanism involving AMPK. This lead to the hypophosphorylation of FOXO1 and a decrease in the expression of a portion of the FSH and FOXO1 transcriptomes. Conversely, FSH-stimulated AMPK, AKT and FOXO1 phosphorylation levels were found to be increased in the granulosa cells of Sfrp4-null mice relative to wild-type controls. SFRP4 treatement of granulosa cells also induced autophagy by signaling via AKT-mTORC1-ULK1, as well as apoptosis. CONCLUSIONS: This study identifies a novel GSK3ß-AMPK-AKT signaling mechanism through which SFPR4 antagonizes FSH action, and further identifies SFRP4 as a novel regulator of granulosa cell autophagy. These findings provide a mechanistic basis for the phenotypic changes previously observed in Sfrp4-null mice, and broaden our understanding of the physiological roles of WNT signaling processes in the ovary.


Assuntos
Autofagia , Hormônio Foliculoestimulante , Células da Granulosa , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Animais , Células da Granulosa/metabolismo , Células da Granulosa/efeitos dos fármacos , Feminino , Proteínas Proto-Oncogênicas c-akt/metabolismo , Autofagia/efeitos dos fármacos , Hormônio Foliculoestimulante/farmacologia , Hormônio Foliculoestimulante/metabolismo , Camundongos , Transdução de Sinais/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Camundongos Endogâmicos C57BL , Células Cultivadas , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Camundongos Knockout
12.
Mol Biol Rep ; 51(1): 807, 2024 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-39002036

RESUMO

BACKGROUND: Acute Myeloid Leukemia (AML) is a fast-developing invading cancer that impacts the blood and bone marrow, marked by the rapid proliferation of abnormal white blood cells. Chemotherapeutic agents, a primary treatment for AML, encounter clinical limitations such as poor solubility and low bioavailability. Previous studies have highlighted antibiotics as effective in inducing cancer cell death and potentially preventing metastasis. Besides, insulin is known to activate the PI3K/Akt pathway, often disrupted in cancers, leading to enhanced cell survival and resistance to apoptosis. In light of the above-mentioned points, we examined the anti-cancer impact of antibiotics Ciprofloxacin (CP) and Salinomycin (SAL) and their combination on KG1-a cells in the presence and absence of insulin. METHODS: This was accomplished by exposing KG1-a cells to different doses of CP and SAL alone, in combination, and with or without insulin for 24-72 h. Cell viability was evaluated using the MTT assay. Besides, apoptotic effects were examined using Hoechst staining and Annexin-V/PI flow cytometry. The expression levels of Bax, p53, BIRC5, Akt, PTEN, and FOXO1 were analyzed through Real-Time PCR. RESULTS: CP and SAL demonstrated cytotoxic and notable pro-apoptotic impact on KG1-a cells by upregulating Bax and p53 and downregulating BIRC5, leading to G0/G1 cell cycle arrest and prevention of the PI3K-Akt signaling pathway. Our findings demonstrated that combination of CP and SAL promote apoptosis in the KG1-a cell line by down-regulating BIRC5 and Akt, as well as up-regulating Bax, p53, PTEN, and FOXO1. Additionally, the findings strongly indicated that insulin effectively mitigates apoptosis by enhancing Akt expression and reducing FOXO1 and PTEN gene expression in the cells treated with CP and SAL. CONCLUSION: Our findings showed that the combined treatment of CP and SAL exhibit a strong anti-cancer effect on leukemia KG1-a cells. Moreover, it was discovered that the PI3K-Akt signaling can be a promising target in leukemia treatment particularly in hyperinsulinemia condition.


Assuntos
Apoptose , Sobrevivência Celular , Ciprofloxacina , Insulina , Piranos , Humanos , Ciprofloxacina/farmacologia , Apoptose/efeitos dos fármacos , Piranos/farmacologia , Linhagem Celular Tumoral , Insulina/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Forkhead Box O1/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proliferação de Células/efeitos dos fármacos , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/genética , Leucemia/tratamento farmacológico , Leucemia/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/genética , Policetídeos de Poliéter
13.
J Mol Model ; 30(8): 260, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38981921

RESUMO

CONTEXT: Diabetes mellitus (DM) is a metabolic disorder disease that causes hyperglycemia conditions and associated with various chronic complications leading to mortality. Due to high toxicity of conventional diabetic drugs, the exploration of natural compounds as alternative diabetes treatments has been widely carried out. Previous in silico studies have highlighted berberine, a natural compound, as a promising alternative in antidiabetic therapy, potentially acting through various pathways, including the inhibition of the FOXO1 transcription factor in the gluconeogenesis pathway. However, the specific mechanism by which berberine interacts with FOXO1 remains unclear, and research in this area is relatively limited. Therefore, this study aims to determine the stability of berberine structure with FOXO1 based on RMSD, RMSF, binding energy, and trajectory analysis to determine the potential of berberine to inhibit the gluconeogenesis pathway. This research was conducted by in silico method with molecular docking using AutoDock4.2 and molecular dynamics study using Amber20, then visualized by VMD. METHODS: Docking between ligand and FOXO1 receptor was carried out with Autodock4.2. For molecular dynamics simulations, the force fields of DNA.OL15, protein.ff14SB, gaff2, and tip3p were used.


Assuntos
Berberina , Proteína Forkhead Box O1 , Hipoglicemiantes , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Berberina/química , Berberina/farmacologia , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/química , Humanos , Ligação Proteica , Sítios de Ligação , Ligantes
14.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167333, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38960054

RESUMO

Periodontitis, a common chronic inflammatory disease, epitomizes a significant impairment in the host immune system and an imbalance of bone metabolism. Macrophage polarization, a dynamic process dictated by the microenvironment, intricately contributes to the interplay between the immune system and bone remodeling, namely the osteoimmune system. Forkhead box protein O1 (FoxO1) has been shown to play a dramatic role in mediating oxidative stress, bone mass, as well as cellular metabolism. Nevertheless, the function and underlying mechanisms of FoxO1 in regulating macrophage polarization-mediated osteogenesis in periodontitis remain to be further elucidated. Here, we found that FoxO1 expression was closely linked to periodontitis, accompanied by aggravated inflammation. Notably, FoxO1 knockdown skewed macrophage polarization from M1 to the antiinflammatory M2 phenotype under inflammatory conditions, which rescued the impaired osteogenic potential. Mechanistically, we revealed that the enhancement of the transcription of peroxisome proliferator-activated receptor (PPAR) signaling in FoxO1-knockdown macrophages. In agreement with this contention, GW9662, a specific inhibitor of PPAR-γ signaling, greatly aggravated macrophage polarization from M2 to the M1 phenotype and attenuated osteogenic potential under inflammatory conditions. Additionally, PPAR-γ signaling agonist rosiglitazone (RSG) was applied to address ligature-induced periodontitis with attenuated inflammation. Our data lend conceptual credence to the function of FoxO1 in mediating macrophage polarization-regulated osteogenesis which serves as a novel therapeutic target for periodontitis.


Assuntos
Proteína Forkhead Box O1 , Macrófagos , Osteogênese , PPAR gama , Periodontite , Transdução de Sinais , PPAR gama/metabolismo , PPAR gama/genética , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Animais , Camundongos , Macrófagos/metabolismo , Periodontite/metabolismo , Periodontite/patologia , Periodontite/genética , Masculino , Camundongos Endogâmicos C57BL , Células RAW 264.7 , Rosiglitazona/farmacologia , Ativação de Macrófagos
15.
Biomed Pharmacother ; 178: 117158, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39042963

RESUMO

Triple-negative breast cancer (TNBC) still one of the most challenging sub-type in breast cancer clinical. Caffeic acid (CA) derived from effective components of traditional Chinese herbal medicine has been show potential against TNBCs. Our research has found that CA can inhibit the proliferation of TNBC cells while also suppressing the size of cancer stem cell spheres. Additionally, it reduces reactive oxygen species (ROS) levels and disruption of mitochondrial membrane potential. Simultaneously, CA influences the stemness of TNBC cells by reducing the expression of the stem cell marker protein CD44. Furthermore, we have observed that CA can modulate the FOXO1/FIS signaling pathway, disrupting mitochondrial function, inducing mitochondrial autophagy, and exerting anti-tumor activity. Additionally, changes in the immune microenvironment were detected using a mass cytometer, we found that CA can induce M1 polarization of macrophages, enhancing anti-tumor immune responses to exert anti-tumor activity. In summary, CA can be considered as a lead compound for further research in targeting TNBC.


Assuntos
Ácidos Cafeicos , Proliferação de Células , Proteína Forkhead Box O1 , Transdução de Sinais , Neoplasias de Mama Triplo Negativas , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/metabolismo , Humanos , Proteína Forkhead Box O1/metabolismo , Ácidos Cafeicos/farmacologia , Linhagem Celular Tumoral , Transdução de Sinais/efeitos dos fármacos , Feminino , Proliferação de Células/efeitos dos fármacos , Animais , Microambiente Tumoral/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Camundongos , Autofagia/efeitos dos fármacos , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo
16.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(6): 1173-1181, 2024 Jun 20.
Artigo em Chinês | MEDLINE | ID: mdl-38977348

RESUMO

OBJECTIVES: To investigate the regulatory role of miRNA-224-5p in hypoxia/reoxygenation (H/R) -induced H9c2 cardiomyocyte injury. METHODS: Plasma samples were collected from 160 patients with acute myocardial infarction and 80 healthy controls(HC) to measure miRNA-224-5p levels and other biochemical parameters. In cultured H9c2 cells with H/R injury, the effects of transfection with miR-224-5p mimics or a negative control sequence on cell viability, malondialdehyde (MDA) content, and superoxide dismutase 2 (SOD2) and lactate dehydrogenase (LDH) activities were tested. Dual luciferase reporter gene assay was performed to verify the targeting relationship between miR-224-5p and PTEN. Bioinformatics methods were used to analyze the potential mechanisms of the target genes. The expression of miRNA-224-5p in the treated cells was detected with qRT-PCR, the protein expressions of PTEN, Bcl-2, Bax, cleaved caspase-3, SOD2, p-PI3K/PI3K, p-Akt/Ak and p-FoxO1/FoxO1 were determined using Western blotting, and cell apoptosis was analysed with flow cytometry. RESULTS: The levels of blood glucose, C-reactive protein, CK, CK-MB and cTnI were significantly higher in the AMI group compared with the HC group (P < 0.05). The expression level of miR-224-5p was significantly lowered in patients with STEMI and NSTEMI and in H9c2 cells with H/R injury. The viability of H9c2 cells decreased time-dependently following H/R injury. PTEN was a target gene of miR-224-5p, and the PI3K/Akt pathway was the most significantly enriched pathway. H9c2 cells with H/R injury showed significantly decreased SOD2 activity, increased LDH activity and MDA content, increased cell apoptosis, decreased protein expression levels of p-PI3K, p-Akt, p-FoxO1, SOD2, and Bcl-2, and increased expressions of PTEN, Bax, and cleaved caspase-3. These changes were obviously attenuated by trasnfection of the cells with miR-224-5p mimics prior to H/R exposure. CONCLUSION: MiR-224-5p overexpression upregulates the expression of the antioxidant gene SOD2 through the PI3K/Akt/FoxO1 axis to relieve H/R-induced oxidative stress and reduce apoptosis of H9c2 cells.


Assuntos
Apoptose , Proteína Forkhead Box O1 , MicroRNAs , Miócitos Cardíacos , Estresse Oxidativo , PTEN Fosfo-Hidrolase , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , MicroRNAs/genética , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Humanos , Ratos , Proteína Forkhead Box O1/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Animais , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Transdução de Sinais , Linhagem Celular , Hipóxia Celular , Superóxido Dismutase/metabolismo , Sobrevivência Celular
17.
Parasit Vectors ; 17(1): 299, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38987795

RESUMO

BACKGROUND: Toxoplasma gondii infection causes adverse pregnancy outcomes by affecting the expression of immunotolerant molecules in decidual immune cells. Galectin-9 (Gal-9) is widely expressed in decidual macrophages (dMφ) and is crucial for maintaining normal pregnancy by interacting with the immunomodulatory protein T-cell immunoglobulin and mucin domain-containing molecule 3 (Tim-3). However, the effects of T. gondii infection on Gal-9 expression in dMφ, and the impact of altered Gal-9 expression levels on the maternal-fetal tolerance function of decidual natural killer (dNK) cells, are still unknown. METHODS: Pregnancy outcomes of T. gondii-infected C57BL/6 and Lgals9-/- pregnant mice models were recorded. Expression of Gal-9, c-Jun N-terminal kinase (JNK), phosphorylated JNK (p-JNK), and Forkhead box protein O1 (FOXO1) was detected by western blotting, flow cytometry or immunofluorescence. The binding of FOXO1 to the promoter of Lgals9 was determined by chromatin immunoprecipitation-polymerase chain reaction (ChIP-PCR). The expression of extracellular signal-regulated kinase (ERK), phosphorylated ERK (p-ERK), cAMP-response element binding protein (CREB), phosphorylated CREB (p-CREB), T-box expressed in T cells (T-bet), interleukin 10 (IL-10), and interferon gamma (IFN-γ) in dNK cells was assayed by western blotting. RESULTS: Toxoplasma gondii infection increased the expression of p-JNK and FOXO1 in dMφ, resulting in a reduction in Gal-9 due to the elevated binding of FOXO1 with Lgals9 promoter. Downregulation of Gal-9 enhanced the phosphorylation of ERK, inhibited the expression of p-CREB and IL-10, and promoted the expression of T-bet and IFN-γ in dNK cells. In the mice model, knockout of Lgals9 aggravated adverse pregnancy outcomes caused by T. gondii infection during pregnancy. CONCLUSIONS: Toxoplasma gondii infection suppressed Gal-9 expression in dMφ by activating the JNK/FOXO1 signaling pathway, and reduction of Gal-9 contributed to dysfunction of dNK via Gal-9/Tim-3 interaction. This study provides new insights for the molecular mechanisms of the adverse pregnancy outcomes caused by T. gondii.


Assuntos
Galectinas , Células Matadoras Naturais , Macrófagos , Camundongos Endogâmicos C57BL , Toxoplasma , Toxoplasmose , Animais , Feminino , Gravidez , Galectinas/genética , Galectinas/metabolismo , Camundongos , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Decídua/imunologia , Camundongos Knockout , Receptor Celular 2 do Vírus da Hepatite A/genética , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Resultado da Gravidez , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo
18.
BMC Genomics ; 25(1): 737, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39080526

RESUMO

OBJECTIVES: Serum/glucocorticoid-inducible kinase 1 (SGK1) gene encodes a serine/threonine protein kinase that plays an essential role in cellular stress response and regulation of multiple metabolic processes. However, its role in bovine adipogenesis remains unknown. In this study, we aimed to clarify the role of SGK1 in bovine lipid accumulation and improvement of meat quality. METHODS: Preadipocytes were induced to differentiation to detect the temporal expression pattern of SGK1. Heart, liver, lung, spleen, kidney, muscle and fat tissues were collected to detect its tissue expression profile. Recombinant adenovirus and the lentivirus were packaged for overexpression and knockdown. Oil Red O staining, quantitative real-time PCR, Western blot analysis, Yeast two-hybrid assay, luciferase assay and RNA-seq were performed to study the regulatory mechanism of SGK1. RESULTS: SGK1 showed significantly higher expression in adipose and significantly induced expression in differentiated adipocytes. Furthermore, overexpression of SGK1 greatly promoted adipogenesis and inhibited proliferation, which could be shown by the remarkable increasement of lipid droplet, and the expression levels of adipogenic marker genes and cell cycle-related genes. Inversely, its knockdown inhibited adipogenesis and facilitated proliferation. Mechanistically, SGK1 regulates the phosphorylation and expression of two critical proteins of FoxO family, FOXO1/FOXO3. Importantly, SGK1 attenuates the transcriptional repression role of FOXO1 for PPARγ via phosphorylating the site S256, then promoting the bovine fat deposition. CONCLUSIONS: SGK1 is a required epigenetic regulatory factor for bovine preadipocyte proliferation and differentiation, which contributes to a better understanding of fat deposition and meat quality improvement in cattle.


Assuntos
Adipócitos , Adipogenia , Proteína Forkhead Box O1 , Proteínas Imediatamente Precoces , Metabolismo dos Lipídeos , Proteínas Serina-Treonina Quinases , Animais , Bovinos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Imediatamente Precoces/metabolismo , Proteínas Imediatamente Precoces/genética , Adipócitos/metabolismo , Adipócitos/citologia , Adipogenia/genética , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Diferenciação Celular , Proliferação de Células , Regulação da Expressão Gênica
19.
Int J Biol Macromol ; 276(Pt 2): 133987, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39032875

RESUMO

This paper aims to investigate the current situation of cancer related fibroblasts promoting malignant development of cancer through FOXO1 protein/LIF signal, and explore the strategy of cancer treatment. Recent studies have shown that the expression of the protein forkhead box O1 (FOXO1) is increased in CAFsCAFs (Cancer-associated fibroblasts). This led researchers to investigate whether FOXO1 is involved in the role of CAFs in lung cancer. The results of the study revealed that FOXO1 is indeed upregulated in CAFs, and it positively regulates the transcription of another protein called LIF. Notably, LIF is also upregulated in both CAFs and lung cancer cells. These changes in protein expression were associated with the overexpression of FOXO1 in CAFs. Conversely, silencing FOXO1 in CAFs suppressed their effects on cancer cells and transplanted tumors. The study revealed that the downregulation of LIFR in cancer cells abolished the impact of CAFs overexpressing FOXO1 on cancer cell behavior. This suggests that the FOXO1/LIF signaling pathway is involved in mediating the malignant development of lung cancer induced by CAFs.


Assuntos
Fibroblastos Associados a Câncer , Proteína Forkhead Box O1 , Regulação Neoplásica da Expressão Gênica , Fator Inibidor de Leucemia , Neoplasias Pulmonares , Transdução de Sinais , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/genética , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Fator Inibidor de Leucemia/metabolismo , Animais , Linhagem Celular Tumoral , Camundongos , Proliferação de Células
20.
Toxicon ; 247: 107857, 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-38996976

RESUMO

Fluoride is a double-edged sword. It was widely used for early caries prevention while excessive intake caused a toxicology effect, affected enamel development, and resulted in dental fluorosis. The study aimed to evaluate the protective effect and mechanism of Epigallocatechin-3-gallate (EGCG) on the apoptosis induced by fluoride in ameloblast-like cells. We observed that NaF triggered apoptotic alterations in cell morphology, excessive NaF arrested cell cycle at the G1, and induced apoptosis by up-regulating Bax and down-regulating Bcl-2. NaF activated the insulin-like growth factor receptor (IGFR), and phosphatidylinositol-3-hydroxylase (p-PI3K), while dose-dependently down-regulating the expression of Forkhead box O1 (FoxO1). EGCG supplements reversed the changes in LS8 morphology, the cell cycle, and apoptosis induced by fluoride. These results indicated that EGCG possesses a protective effect against fluoride toxicity. Furthermore, EGCG suppressed the activation of p-PI3K and the down-regulation of FoxO1 caused by fluoride. Collectively, our findings suggested that EGCG attenuated fluoride-induced apoptosis by inhibiting the PI3K/FoxO1 signaling pathway. EGCG may serve as a new alternative method for dental fluorosis prevention, control, and treatment.


Assuntos
Ameloblastos , Apoptose , Catequina , Fluoretos , Fosfatidilinositol 3-Quinases , Transdução de Sinais , Catequina/análogos & derivados , Catequina/farmacologia , Apoptose/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Fluoretos/toxicidade , Fluoretos/farmacologia , Ameloblastos/efeitos dos fármacos , Ameloblastos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Forkhead Box O1/metabolismo , Linhagem Celular , Camundongos , Fluoreto de Sódio/toxicidade , Fluorose Dentária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...