Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 903
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(41): e2408719121, 2024 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-39352930

RESUMO

As ambush-hunting predators that consume large prey after long intervals of fasting, Burmese pythons evolved with unique adaptations for modulating organ structure and function. Among these is cardiac hypertrophy that develops within three days following a meal (Andersen et al., 2005, Secor, 2008), which we previously showed was initiated by circulating growth factors (Riquelme et al., 2011). Postprandial cardiac hypertrophy in pythons also rapidly regresses with subsequent fasting (Secor, 2008); however, the molecular mechanisms that regulate the dynamic cardiac remodeling in pythons during digestion are largely unknown. In this study, we employed a multiomics approach coupled with targeted molecular analyses to examine remodeling of the python ventricular transcriptome and proteome throughout digestion. We found that forkhead box protein O1 (FoxO1) signaling was suppressed prior to hypertrophy development and then activated during regression, which coincided with decreased and then increased expression, respectively, of FoxO1 transcriptional targets involved in proteolysis. To define the molecular mechanistic role of FoxO1 in hypertrophy regression, we used cultured mammalian cardiomyocytes treated with postfed python plasma. Hypertrophy regression both in pythons and in vitro coincided with activation of FoxO1-dependent autophagy; however, the introduction of a FoxO1-specific inhibitor prevented both regression of cell size and autophagy activation. Finally, to determine whether FoxO1 activation could induce regression, we generated an adenovirus expressing a constitutively active FoxO1. FoxO1 activation was sufficient to prevent and reverse postfed plasma-induced hypertrophy, which was partially prevented by autophagy inhibition. Our results indicate that modulation of FoxO1 activity contributes to the dynamic ventricular remodeling in postprandial Burmese pythons.


Assuntos
Boidae , Cardiomegalia , Proteína Forkhead Box O1 , Período Pós-Prandial , Animais , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Cardiomegalia/metabolismo , Cardiomegalia/genética , Cardiomegalia/patologia , Miócitos Cardíacos/metabolismo , Autofagia , Transdução de Sinais , Transcriptoma
2.
Nat Commun ; 15(1): 7879, 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39251582

RESUMO

Naïve pluripotency is sustained by a self-reinforcing gene regulatory network (GRN) comprising core and naïve pluripotency-specific transcription factors (TFs). Upon exiting naïve pluripotency, embryonic stem cells (ESCs) transition through a formative post-implantation-like pluripotent state, where they acquire competence for lineage choice. However, the mechanisms underlying disengagement from the naïve GRN and initiation of the formative GRN are unclear. Here, we demonstrate that phosphorylated AKT acts as a gatekeeper that prevents nuclear localisation of FoxO TFs in naïve ESCs. PTEN-mediated reduction of AKT activity upon exit from naïve pluripotency allows nuclear entry of FoxO TFs, enforcing a cell fate transition by binding and activating formative pluripotency-specific enhancers. Indeed, FoxO TFs are necessary and sufficient for the activation of the formative pluripotency-specific GRN. Our work uncovers a pivotal role for FoxO TFs in establishing formative post-implantation pluripotency, a critical early embryonic cell fate transition.


Assuntos
Redes Reguladoras de Genes , Células-Tronco Pluripotentes , Animais , Camundongos , Células-Tronco Pluripotentes/metabolismo , Diferenciação Celular/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/genética , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição Forkhead/genética , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Fosforilação , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento
3.
Sci Rep ; 14(1): 20773, 2024 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-39237585

RESUMO

The study of cooperating genes in cancer can lead to mechanistic understanding and identifying potential therapeutic targets. To facilitate these types of studies, we developed a new dual-inducible system utilizing the tetracycline- and cumate-inducible systems driving HES3 and the PAX3::FOXO1 fusion-oncogene, respectively, as cooperating genes from fusion-positive rhabdomyosarcoma. With this model, we can independently induce expression of either HES3 or PAX3::FOXO1, as well as simultaneously induce expression of both genes. This new model will allow us to further investigate the cooperation between HES3 and PAX3::FOXO1 including the temporal requirements for genetic cooperation. Functionally, we show that dual-induction of PAX3::FOXO1 and HES3 modifies sphere formation in a HEK293T-based system. More broadly, this lentiviral dual-inducible system can be adapted for any cooperating genes (overexpression or knockdown), allowing for independent, simultaneous, or temporally controlled gene expression.


Assuntos
Regulação Neoplásica da Expressão Gênica , Humanos , Células HEK293 , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Rabdomiossarcoma/genética , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Fator de Transcrição PAX3/genética , Fator de Transcrição PAX3/metabolismo
4.
Int J Mol Sci ; 25(18)2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39337411

RESUMO

Sirtuin 1 (SIRT1) is a key upstream regulator of lipid metabolism; however, the molecular mechanisms by which SIRT1 regulates milk fat synthesis in dairy goats remain unclear. This study aimed to investigate the regulatory roles of SIRT1 in modulating lipid metabolism in goat mammary epithelial cells (GMECs) and its impact on the adipose triglyceride lipase (ATGL) promoter activity using RNA interference (RNAi) and gene overexpression techniques. The results showed that SIRT1 is significantly upregulated during lactation compared to the dry period. Additionally, SIRT1 knockdown notably increased the expressions of genes related to fatty acid synthesis (SREBP1, SCD1, FASN, ELOVL6), triacylglycerol (TAG) production (DGAT2, AGPAT6), and lipid droplet formation (PLIN2). Consistent with the transcriptional changes, SIRT1 knockdown significantly increased the intracellular contents of TAG and cholesterol and the lipid droplet abundance in the GMECs, while SIRT1 overexpression had the opposite effects. Furthermore, the co-overexpression of SIRT1 and Forkhead box protein O1 (FOXO1) led to a more pronounced increase in ATGL promoter activity, and the ability of SIRT1 to enhance ATGL promoter activity was nearly abolished when the FOXO1 binding sites (FKH1 and FKH2) were mutated, indicating that SIRT1 enhances the transcriptional activity of ATGL via the FKH element in the ATGL promoter. Collectively, our data reveal that SIRT1 enhances the transcriptional activity of ATGL through the FOXO1 binding sites located in the ATGL promoter, thereby regulating lipid metabolism. These findings provide novel insights into the role of SIRT1 in fatty acid metabolism in dairy goats.


Assuntos
Células Epiteliais , Ácidos Graxos , Proteína Forkhead Box O1 , Cabras , Lipase , Glândulas Mamárias Animais , Regiões Promotoras Genéticas , Sirtuína 1 , Animais , Sirtuína 1/metabolismo , Sirtuína 1/genética , Lipase/metabolismo , Lipase/genética , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/citologia , Feminino , Ácidos Graxos/metabolismo , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Metabolismo dos Lipídeos , Lactação , Triglicerídeos/metabolismo , Triglicerídeos/biossíntese , Regulação da Expressão Gênica
5.
Nutrients ; 16(18)2024 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-39339684

RESUMO

BACKGROUND: Sarcopenic obesity, which is associated with a poorer prognosis than that of sarcopenia alone, may be positively affected by soy isoflavones, known inhibitors of muscle atrophy. Herein, we hypothesize that these compounds may prevent sarcopenic obesity by upregulating the gut metabolites with anti-inflammatory effects. METHODS: To explore the effects of soy isoflavones on sarcopenic obesity and its mechanisms, we employed both in vivo and in vitro experiments. Mice were fed a high-fat, high-sucrose diet with or without soy isoflavone supplementation. Additionally, the mouse C2C12 myotube cells were treated with palmitic acid and daidzein in vitro. RESULTS: The isoflavone considerably reduced muscle atrophy and the expression of the muscle atrophy genes in the treated group compared to the control group (Fbxo32, p = 0.0012; Trim63, p < 0.0001; Foxo1, p < 0.0001; Tnfa, p = 0.1343). Elevated levels of daidzein were found in the muscles and feces of the experimental group compared to the control group (feces, p = 0.0122; muscle, p = 0.0020). The real-time PCR results demonstrated that the daidzein decreased the expression of the palmitate-induced inflammation and muscle atrophy genes in the C2C12 myotube cells (Tnfa, p = 0.0201; Il6, p = 0.0008; Fbxo32, p < 0.0001; Hdac4, p = 0.0002; Trim63, p = 0.0114; Foxo1, p < 0.0001). Additionally, it reduced the palmitate-induced protein expression related to the muscle atrophy in the C2C12 myotube cells (Foxo1, p = 0.0078; MuRF1, p = 0.0119). CONCLUSIONS: The daidzein suppressed inflammatory cytokine- and muscle atrophy-related gene expression in the C2C12 myotubes, thereby inhibiting muscle atrophy.


Assuntos
Citocinas , Isoflavonas , Atrofia Muscular , Isoflavonas/farmacologia , Animais , Camundongos , Atrofia Muscular/tratamento farmacológico , Atrofia Muscular/metabolismo , Atrofia Muscular/prevenção & controle , Masculino , Citocinas/metabolismo , Citocinas/genética , Linhagem Celular , Camundongos Endogâmicos C57BL , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Sarcopenia/prevenção & controle , Sarcopenia/metabolismo , Sarcopenia/tratamento farmacológico , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Dieta Hiperlipídica/efeitos adversos , Obesidade/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/genética , Glycine max/química , Modelos Animais de Doenças , Ácido Palmítico/farmacologia
6.
PLoS One ; 19(9): e0310999, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39348384

RESUMO

Regulation of visual system function demands precise gene regulation. Dysregulation of miRNAs, as key regulators of gene expression in retinal cells, contributes to different eye disorders such as diabetic retinopathy (DR), macular edema, and glaucoma. MIR-96, a member of the MIR-183 cluster family, is widely expressed in the retina, and its alteration is associated with neovascular eye diseases. MIR-96 regulates protein cascades in inflammatory and insulin signaling pathways, but further investigation is required to understand its potential effects on related genes. For this purpose, we identified a series of key target genes for MIR-96 based on gene and protein interaction networks and utilized text-mining resources. To examine the MIR-96 impact on candidate gene expression, we overexpressed MIR-96 via adeno-associated virus (AAV)-based plasmids in human retinal pigment epithelial (RPE) cells. Based on Real-Time PCR results, the relative expression of the selected genes responded differently to overexpressed MIR-96. While the expression levels of IRS2, FOXO1, and ERK2 (MAPK1) were significantly decreased, the SERPINF1 gene exhibited high expression simultaneously. pAAV-delivered MIR-96 had no adverse effect on the viability of human RPE cells. The data showed that changes in insulin receptor substrate-2 (IRS2) expression play a role in disrupted retinal insulin signaling and contribute to the development of diabetic complications. Considered collectively, our findings suggest that altered MIR-96 and its impact on IRS/PI3K/AKT/VEGF axis regulation contribute to DR progression. Therefore, further investigation of the IRS/PI3K/AKT/VEGF axis is recommended as a potential target for DR treatment.


Assuntos
Retinopatia Diabética , Proteínas Substratos do Receptor de Insulina , MicroRNAs , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Epitélio Pigmentado da Retina , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular , MicroRNAs/genética , MicroRNAs/metabolismo , Humanos , Retinopatia Diabética/genética , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Linhagem Celular , Regulação da Expressão Gênica
7.
Front Immunol ; 15: 1389387, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39247196

RESUMO

Introduction: The role of zinc (Zn) in tumor development and immune modulation has always been paradoxical. This study redefines our understanding of the impact of Zn on cancer progression and therapeutic strategies. Methods: We investigated the effects of dietary Zn levels on tumor progression and immune responses. This included examining the impact of both high and deficient dietary Zn, as well as Zn chelation, on tumor growth and immune cell populations. Specifically, we analyzed the frequency of Foxp3+ regulatory T-cells (Tregs) and identified the role of FOXO1 in Zn-mediated effects on Tregs. Additionally, we explored the therapeutic potential of clioquinol (CQ) in enhancing α-PD-1 immunotherapy responses, particularly in melanoma. Results: Our findings show that high dietary Zn promotes tumor progression by fostering a protumorigenic environment mediated by T cells. Increased Zn intake was found to facilitate tumor progression by increasing Foxp3+ Treg frequency. In contrast, deficiency in dietary Zn and chelation of tissue Zn emerged as potent drivers of antitumor immunity. We pinpointed FOXO1 as the master regulator governing the influence of Zn on Tregs. Discussion: These results reveal a novel mechanistic insight into how Zn influences tumor progression and immune regulation. The identification of FOXO1 as a key regulator opens new avenues for understanding the role of Zn in cancer biology. Furthermore, we introduce a promising therapeutic approach by showing that administering clioquinol (CQ) significantly enhances α-PD-1 immunotherapy response, particularly in melanoma. These revelations transform our comprehension of the multifaceted role of Zn in tumorigenesis and immune regulation, highlighting innovative possibilities for cancer therapy.


Assuntos
Fatores de Transcrição Forkhead , Linfócitos T Reguladores , Zinco , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Zinco/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Camundongos , Clioquinol/farmacologia , Camundongos Endogâmicos C57BL , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Melanoma Experimental/imunologia , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/metabolismo , Melanoma/imunologia , Melanoma/tratamento farmacológico , Humanos , Linhagem Celular Tumoral , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos dos fármacos , Imunoterapia/métodos , Feminino
8.
Int J Nanomedicine ; 19: 8751-8768, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39220194

RESUMO

Purpose: Periodontitis is a chronic infectious disease characterized by progressive inflammation and alveolar bone loss. Forkhead box O1 (FoxO1), an important regulator, plays a crucial role in maintaining bone homeostasis and regulating macrophage energy metabolism and osteogenic differentiation of mesenchymal stem cells (MSCs). In this study, FoxO1 was overexpressed into small extracellular vesicles (sEV) using engineering technology, and effects of FoxO1-overexpressed sEV on periodontal tissue regeneration as well as the underlying mechanisms were investigated. Methods: Human periodontal ligament stem cell (hPDLSCs)-derived sEV (hPDLSCs-sEV) were isolated using ultracentrifugation. They were then characterized using transmission electron microscopy, Nanosight, and Western blotting analyses. hPDLSCs were treated with hPDLSCs-sEV in vitro after stimulation with lipopolysaccharide, and osteogenesis was evaluated. The effect of hPDLSCs-sEV on the polarization phenotype of THP-1 macrophages was also evaluated. In addition, we measured the reactive oxygen species (ROS) levels, adenosine triphosphate (ATP) production, mitochondrial characteristics, and metabolism of hPDLSCs and THP-1 cells. Experimental periodontitis was established in vivo in mice. HPDLSCs-sEV or phosphate-buffered saline (PBS) were injected into periodontal tissues for four weeks, and the maxillae were collected and assessed by micro-computed tomography, histological staining, and small animal in vivo imaging. Results: In vitro, FoxO1-overexpressed sEV promoted osteogenic differentiation of hPDLSCs in the inflammatory environment and polarized THP-1 cells from the M1 phenotype to the M2 phenotype. Furthermore, FoxO1-overexpressed sEV regulated the ROS level, ATP production, mitochondrial characteristics, and metabolism of hPDLSCs and THP-1 cells in the inflammatory environment. In the in vivo analyses, FoxO1-overexpressed sEV effectively promoted bone formation and inhibited inflammation. Conclusion: FoxO1-overexpressed sEV can regulate osteogenesis and immunomodulation. The ability of FoxO1-overexpressed sEV to regulate inflammation and osteogenesis can pave the way for the establishment of a therapeutic approach for periodontitis.


Assuntos
Vesículas Extracelulares , Proteína Forkhead Box O1 , Mitocôndrias , Osteogênese , Ligamento Periodontal , Periodontite , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Osteogênese/efeitos dos fármacos , Animais , Humanos , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Mitocôndrias/metabolismo , Periodontite/terapia , Periodontite/metabolismo , Camundongos , Ligamento Periodontal/citologia , Células THP-1 , Espécies Reativas de Oxigênio/metabolismo , Inflamação/metabolismo , Masculino , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Macrófagos/metabolismo , Regeneração , Células Cultivadas
9.
Acta Neuropathol ; 148(1): 43, 2024 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-39283487

RESUMO

Amyotrophic Lateral Sclerosis (ALS) is a multisystemic neurodegenerative disorder, with accumulating evidence indicating metabolic disruptions in the skeletal muscle preceding disease symptoms, rather than them manifesting as a secondary consequence of motor neuron (MN) degeneration. Hence, energy homeostasis is deeply implicated in the complex physiopathology of ALS and skeletal muscle has emerged as a key therapeutic target. Here, we describe intrinsic abnormalities in ALS skeletal muscle, both in patient-derived muscle cells and in muscle cell lines with genetic knockdown of genes related to familial ALS, such as TARDBP (TDP-43) and FUS. We found a functional impairment of myogenesis that parallels defects of glucose oxidation in ALS muscle cells. We identified FOXO1 transcription factor as a key mediator of these metabolic and functional features in ALS muscle, via gene expression profiling and biochemical surveys in TDP-43 and FUS-silenced muscle progenitors. Strikingly, inhibition of FOXO1 mitigated the impaired myogenesis in both the genetically modified and the primary ALS myoblasts. In addition, specific in vivo conditional knockdown of TDP-43 or FUS orthologs (TBPH or caz) in Drosophila muscle precursor cells resulted in decreased innervation and profound dysfunction of motor nerve terminals and neuromuscular synapses, accompanied by motor abnormalities and reduced lifespan. Remarkably, these phenotypes were partially corrected by foxo inhibition, bolstering the potential pharmacological management of muscle intrinsic abnormalities associated with ALS. The findings demonstrate an intrinsic muscle dysfunction in ALS, which can be modulated by targeting FOXO factors, paving the way for novel therapeutic approaches that focus on the skeletal muscle as complementary target tissue.


Assuntos
Esclerose Lateral Amiotrófica , Proteína Forkhead Box O1 , Músculo Esquelético , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Humanos , Animais , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Masculino , Proteína FUS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/metabolismo , Feminino , Drosophila , Desenvolvimento Muscular/fisiologia , Pessoa de Meia-Idade , Idoso , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Mioblastos/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 44(9): 2004-2023, 2024 09.
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
11.
Development ; 151(18)2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39166965

RESUMO

The microvascular system consists of two cell types: endothelial and mural (pericytes and vascular smooth muscle cells; VSMCs) cells. Communication between endothelial and mural cells plays a pivotal role in the maintenance of vascular homeostasis; however, in vivo molecular and cellular mechanisms underlying mural cell development remain unclear. In this study, we found that macrophages played a crucial role in TGFß-dependent pericyte-to-VSMC differentiation during retinal vasculature development. In mice with constitutively active Foxo1 overexpression, substantial accumulation of TGFß1-producing macrophages and pericytes around the angiogenic front region was observed. Additionally, the TGFß-SMAD pathway was activated in pericytes adjacent to macrophages, resulting in excess ectopic α-smooth muscle actin-positive VSMCs. Furthermore, we identified endothelial SEMA3C as an attractant for macrophages. In vivo neutralization of SEMA3C rescued macrophage accumulation and ectopic VSMC phenotypes in the mice, as well as drug-induced macrophage depletion. Therefore, macrophages play an important physiological role in VSMC development via the FOXO1-SEMA3C pathway.


Assuntos
Proteína Forkhead Box O1 , Macrófagos , Músculo Liso Vascular , Miócitos de Músculo Liso , Semaforinas , Animais , Macrófagos/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citologia , Camundongos , Semaforinas/metabolismo , Semaforinas/genética , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/citologia , Pericitos/metabolismo , Pericitos/citologia , Diferenciação Celular , Transdução de Sinais , Vasos Retinianos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição Forkhead/genética , Fator de Crescimento Transformador beta1/metabolismo , Camundongos Endogâmicos C57BL
12.
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 , Animais , Humanos , Camundongos , Apoptose/genética , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Sobrevivência Celular/genética , Progressão da Doença , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/patologia , RNA Circular/genética , RNA Circular/metabolismo
13.
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
14.
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
15.
J Dig Dis ; 25(7): 453-462, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39211938

RESUMO

OBJECTIVE: We aimed to investigate the role of forkhead box O1 (FoxO1) inhibitor AS1842856 (AS) in nonalcoholic steatohepatitis (NASH) mice and the potential mechanisms. METHODS: Mice were given methionine-choline-sufficient (MCS), or methionine- and choline-deficient (MCD) diet for 5 weeks, along with AS (60 mg/kg) or vehicle gavage treatment (0.2 mL/day). Body and liver weight, serum triglyceride (TG), low-density lipoprotein-cholesterol (LDL-C), high-density lipoprotein-cholesterol (HDL-C), alanine aminotransferase (ALT), aspartate aminotransferase (AST), fasting glucose and insulin levels were measured. Liver macrophage infiltration and ileal ZO-1 protein expression were also detected. Interleukin (IL)-6, IL-1ß, and tumor necrosis factor (TNF)-α, sterol regulatory element binding protein (SREBP)-1c, phosphoenolpyruvate carboxykinase (PEPCK), and glucose-6-phosphatase (G6Pase), α-smooth muscle actin (SMA), recombinant collagen type III α1 (Col3a1), and connective tissue growth factor (Ctgf) expressions were measured. Stool samples were collected for 16S rDNA sequencing. RESULTS: Compared to the MCD group, AS attenuated liver weight, reduced serum TG, ALT, and AST levels, increased HDL-C levels, mitigated hepatic steatosis, decreased macrophage infiltration, and augmented ileal ZO-1 proteins in NASH mice. It also reduced the levels of IL-6, IL-1ß, and TNF-α, alongside with the Srebp-1c mRNA expression. However, no significant effects on Pepck, G6Pase, α-SMA, Col3a1, or Ctgf were observed. Furthermore, AS promoted diversity and altered gut microbiota composition in NASH mice, causing increased beneficial bacteria like Akkermansia muciniphila, Parabacteroides distasonis, and Prevotellamassilia, which were associated with metabolic functions. CONCLUSION: FoxO1 inhibitor AS ameliorated hepatic steatosis, inflammation, and intestinal dysbiosis in NASH mice, making it a potentially promising treatment for NASH.


Assuntos
Proteína Forkhead Box O1 , Microbioma Gastrointestinal , Hepatopatia Gordurosa não Alcoólica , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo , Camundongos , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Masculino , Fígado/patologia , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Hepatite/tratamento farmacológico , Hepatite/prevenção & controle
16.
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
17.
Fish Shellfish Immunol ; 153: 109793, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39134230

RESUMO

Microplastic pollution poses challenges for ecosystems worldwide, and nanoplastics (NPs, 1-1000 nm) have been identified as persistent pollutants. However, although some studies have described the hazards of NPs to aquatic organisms, the toxicological processes of NPs in the common carp kidney and the biotoxicity of differently sized NPs remain unclear. In this study, we used juvenile common carp as an in vivo model that were constantly exposed to freshwater at 1000 µg/L polystyrene nanoparticle (PSNP) concentrations (50, 100, and 400 nm) for 28 days. Simultaneously, we constructed an in vitro model utilizing grass fish kidney cells (CIK) to study the toxicological effects of PSNPs of various sizes. We performed RT-PCR and Western blot assays on the genes involved in FOXO1, HMGB1, HIF-1α, endoplasmic reticulum stress, autophagy, and immunoreaction. According to these results, exposure to PSNPs increased reactive oxygen species (ROS) levels, and the carp kidneys experienced endoplasmic reticulum stress. Additionally, PSNPs promoted renal autophagy by activating the ROS/ERS/FOXO1 (ERS: endoplasmic reticulum stress) pathway, and it affected immunological function by stimulating the ROS/HMGB1/HIF-1α signaling pathway. This study provides new insights into the contamination hazards of NPs in freshwater environments, as well as the harm they pose to the human living environments. The relationship between particle size and the degree of damage caused by PSNPs to organisms is a potential future research direction.


Assuntos
Autofagia , Carpas , Rim , Nanopartículas , Tamanho da Partícula , Poliestirenos , Espécies Reativas de Oxigênio , Animais , Carpas/imunologia , Nanopartículas/toxicidade , Nanopartículas/química , Autofagia/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Poliestirenos/toxicidade , Poliestirenos/química , Rim/efeitos dos fármacos , Rim/imunologia , Poluentes Químicos da Água/toxicidade , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Microplásticos/toxicidade , Microplásticos/química
18.
Toxicol Appl Pharmacol ; 491: 117075, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39173720

RESUMO

Artesunate (ART) is a derivative of artemisinin and has anti-inflammatory, anti-tumor, and anti-angiogenic properties. Although ART has been implicated in osteoarthritis (OA), the mechanism needs to be further dissected. Here, we explored the effects of ART on the development of OA and the underlying mechanism using destabilization of the medial meniscus (DMM) surgical instability model. Mice with OA were developed using DMM and treated with ART. The pathological morphology of knee joint tissues was examined, and the degeneration of joint cartilage was assessed. Mouse knee chondrocytes were isolated and induced with IL-1ß, followed by ART treatment. ART alleviates OA in mice by elevating ubiquitin carboxyl-terminal hydrolase 7 (USP7) expression, and USP7 inhibitor (P22077) treatment mitigated the protective effects of ART on chondrocytes. We also showed that USP7 mediated the deubiquitination of forkhead box protein O1 (FoxO1), while FoxO1 alleviated chondrocyte injury. In addition, FoxO1 promoted metastasis-associated protein MTA1 (MTA1) transcription, and downregulation of MTA1 exacerbated chondrocyte injury. Our study identifies that USP7/FoxO1/MTA1 is a key signaling cascade in the treatment of ART on OA.


Assuntos
Artesunato , Condrócitos , Proteína Forkhead Box O1 , Camundongos Endogâmicos C57BL , Osteoartrite , Peptidase 7 Específica de Ubiquitina , Animais , Artesunato/farmacologia , Artesunato/uso terapêutico , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Peptidase 7 Específica de Ubiquitina/metabolismo , Peptidase 7 Específica de Ubiquitina/genética , Camundongos , Masculino , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Osteoartrite/patologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Transativadores/metabolismo , Transativadores/genética , Transdução de Sinais/efeitos dos fármacos , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transcrição Gênica/efeitos dos fármacos
19.
Sci Rep ; 14(1): 19948, 2024 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-39198693

RESUMO

Non-small cell lung cancer (NSCLC), being the most prevalent and lethal malignancy affecting the lungs, poses a significant threat to human health. This research aims at illustrating the precise role and related mechanisms of silent information regulator type-1 (SIRT1) in NSCLC progression. The expression pattern of SIRT1 in NSCLC cell lines was examined using quantitative real-time polymerase chain reaction and western blotting. Functional assays in NSCLC cell lines validated the biological capabilities of SIRT1 on malignant phenotypes, and its impact on tumorigenicity was further evaluated in vivo. In addition, the FOXO1 inhibitor AS1842856 was applied to verify the role of SIRT1 on FOXO pathway in vitro. SIRT1 expression was prominently elevated in NSCLC cell lines. The depletion of SIRT1 retarded the capabilities of proliferation, migration and invasion, while enhancing apoptosis in NSCLC cells. Furthermore, SIRT1 silencing restricted the tumorigenesis of NSCLC in vivo. Additionally, AS1842856 treatment ameliorated the inhibitory effect of SIRT1 deficiency on malignant phenotypes in NSCLC cells. SIRT1 deletion exerted an anti-oncogenic role in NSCLC via activation of FOXO1.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Movimento Celular , Proliferação de Células , Proteína Forkhead Box O1 , Neoplasias Pulmonares , Sirtuína 1 , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Sirtuína 1/metabolismo , Sirtuína 1/genética , Humanos , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Linhagem Celular Tumoral , Animais , Movimento Celular/genética , Camundongos , Apoptose/genética , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Camundongos Nus , Quinolonas
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA