Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 319
Filtrar
1.
ACS Appl Mater Interfaces ; 16(17): 21383-21399, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38626424

RESUMO

Osteoarthritis (OA) progression is highly associated with chondrocyte mitochondrial dysfunction and disorders of catabolism and anabolism of the extracellular matrix (ECM) in the articular cartilage. The mitochondrial unfolded protein response (UPRmt), which is an integral component of the mitochondrial quality control (MQC) system, is essential for maintaining chondrocyte homeostasis. We successfully validated the pivotal role of activating transcription factor 5 (ATF5) in upregulating the UPRmt, mitigating IL-1ß-induced inflammation and mitochondrial dysfunction, and promoting balanced metabolism in articular cartilage ECM, proving its potential as a promising therapeutic target for OA. Modified mRNAs (modRNAs) have emerged as novel and efficient gene delivery vectors for nucleic acid therapeutic approaches. In this study, we combined Atf5-modRNA (modAtf5) with engineered exosomes derived from bone mesenchymal stem cells (ExmodAtf5) to exert cytoprotective effects on chondrocytes in articular cartilage via Atf5. However, the rapid localized metabolization of ExmodAtf5 limits its application. PLGA-PEG-PLGA (Gel), an injectable thermosensitive hydrogel, was used as a carrier of ExmodAtf5 (Gel@ExmodAtf5) to achieve a sustained release of ExmodAtf5. In vitro and in vivo, the use of Gel@ExmodAtf5 was shown to be a highly effective strategy for OA treatment. The in vivo therapeutic effect of Gel@ExmodAtf5 was evidenced by the preservation of the intact cartilage surface, low OARSI scores, fewer osteophytes, and mild subchondral bone sclerosis and cystic degeneration. Consequently, the combination of ExmodAtf5 and PLGA-PEG-PLGA could significantly enhance the therapeutic efficacy and prolong the exosome release. In addition, the mitochondrial protease ClpP enhanced chondrocyte autophagy by modulating the mTOR/Ulk1 pathway. As a result of our research, Gel@ExmodAtf5 can be considered to be effective at alleviating the progression of OA.


Assuntos
Fatores Ativadores da Transcrição , Condrócitos , Exossomos , Mitocôndrias , Osteoartrite , RNA Mensageiro , Resposta a Proteínas não Dobradas , Osteoartrite/patologia , Osteoartrite/metabolismo , Osteoartrite/terapia , Exossomos/metabolismo , Exossomos/química , Animais , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Fatores Ativadores da Transcrição/metabolismo , Fatores Ativadores da Transcrição/química , Fatores Ativadores da Transcrição/genética , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Condrócitos/metabolismo , Condrócitos/efeitos dos fármacos , Humanos , Células-Tronco Mesenquimais/metabolismo , Hidrogéis/química , Masculino , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Cartilagem Articular/efeitos dos fármacos
2.
Cancer Res Commun ; 3(12): 2518-2530, 2023 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-38014922

RESUMO

MYCN-amplified neuroblastoma often presents as a highly aggressive metastatic disease with a poor prognosis. Activating transcription factor 5 (ATF5) is implicated in neural cell differentiation and cancer cell survival. Here, we show that ATF5 is highly expressed in patients with stage 4 high-risk neuroblastoma, with increased expression correlating with a poorer prognosis. We demonstrated that ATF5 promotes the metastasis of neuroblastoma cell lines in vivo. Functionally, ATF5 depletion significantly reduced xenograft tumor growth and metastasis of neuroblastoma cells to the bone marrow and liver. Mechanistically, ATF5 endows tumor cells with resistance to anoikis, thereby increasing their survival in systemic circulation and facilitating metastasis. We identified the proapoptotic BCL-2 modifying factor (BMF) as a critical player in ATF5-regulated neuroblastoma anoikis. ATF5 suppresses BMF under suspension conditions at the transcriptional level, promoting anoikis resistance, whereas BMF knockdown significantly prevents ATF5 depletion-induced anoikis. Therapeutically, we showed that a cell-penetrating dominant-negative ATF5 peptide, CP-d/n-ATF5, inhibits neuroblastoma metastasis to the bone marrow and liver by inducing anoikis sensitivity in circulating tumor cells. Our study identified ATF5 as a metastasis promoter and CP-d/n-ATF5 as a potential antimetastatic therapeutic agent for neuroblastoma. SIGNIFICANCE: This study shows that resistance to anoikis in neuroblastoma is mediated by ATF5 and offers a rationale for targeting ATF5 to treat metastatic neuroblastoma.


Assuntos
Antineoplásicos , Neuroblastoma , Humanos , Anoikis/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Neuroblastoma/tratamento farmacológico , Antineoplásicos/farmacologia , Fatores Ativadores da Transcrição
3.
Fish Physiol Biochem ; 49(6): 1115-1128, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37855969

RESUMO

Glucose-regulated protein 78 (grp78) and activating transcription factor 6α (atf6α) are considered vital endoplasmic reticulum (ER) molecular chaperones and ER stress (ERS) sensors, respectively. In the present study, the full cDNA sequences of these two ERS-related genes were first cloned and characterized from black seabream (Acanthopagrus schlegelii). The grp78 cDNA sequence is 2606 base pair (bp) encoding a protein of 654 amino acids (aa). The atf6α cDNA sequence is 2168 base pair (bp) encoding a protein of 645 aa. The predicted aa sequences of A. schlegelii grp78 and atf6α indicated that the proteins contain all the structural features, which were characteristic of the two genes in other species. Tissues transcript abundance analysis revealed that the mRNAs of grp78 and atf6α were expressed in all measured tissues, but the highest expression of these two genes was all recorded in the gill followed by liver/ brain. Moreover, in vivo experiment found that fish intake of a high lipid diet (HLD) can trigger ERS by activating grp78/Grp78 and atf6α/Atf6α. However, it can be alleviated by dietary betaine supplementation, similar results were also obtained by in vitro experiment using primary hepatocytes of A. schlegelii. These findings will be beneficial for us to evaluate the regulator effects of HLD supplemented with betaine on ERS at the molecular level, and thus provide some novel insights into the functions of betaine in marine fish fed with an HLD.


Assuntos
Perciformes , Dourada , Animais , Chaperona BiP do Retículo Endoplasmático , Dourada/genética , Betaína , DNA Complementar/genética , Perciformes/genética , Estresse do Retículo Endoplasmático , Fatores Ativadores da Transcrição/genética , Clonagem Molecular
4.
Nat Commun ; 14(1): 2886, 2023 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-37210583

RESUMO

Cisplatin kills proliferating cells via DNA damage but also has profound effects on post-mitotic cells in tumors, kidneys, and neurons. However, the effects of cisplatin on post-mitotic cells are still poorly understood. Among model systems, C. elegans adults are unique in having completely post-mitotic somatic tissues. The p38 MAPK pathway controls ROS detoxification via SKN-1/NRF and immune responses via ATF-7/ATF2. Here, we show that p38 MAPK pathway mutants are sensitive to cisplatin, but while cisplatin exposure increases ROS levels, skn-1 mutants are resistant. Cisplatin exposure leads to phosphorylation of PMK-1/MAPK and ATF-7 and the IRE-1/TRF-1 signaling module functions upstream of the p38 MAPK pathway to activate signaling. We identify the response proteins whose increased abundance depends on IRE-1/p38 MAPK activity as well as cisplatin exposure. Four of these proteins are necessary for protection from cisplatin toxicity, which is characterized by necrotic death. We conclude that the p38 MAPK pathway-driven proteins are crucial for adult cisplatin resilience.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Cisplatino/toxicidade , Cisplatino/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sistema de Sinalização das MAP Quinases , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Fatores Ativadores da Transcrição
5.
Cell Rep ; 42(5): 112479, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37178686

RESUMO

CREB/ATF transcription factor OASIS/CREB3L1 is upregulated in long-term-cultured astrocytes undergoing cell-cycle arrest due to loss of DNA integrity by repeated replication. However, the roles of OASIS in the cell cycle remain unexplored. We find that OASIS arrests the cell cycle at G2/M phase after DNA damage via direct induction of p21. Cell-cycle arrest by OASIS is dominant in astrocytes and osteoblasts, but not in fibroblasts, which are dependent on p53. In a brain injury model, Oasis-/- reactive astrocytes surrounding the lesion core show sustained growth and inhibition of cell-cycle arrest, resulting in prolonged gliosis. We find that some glioma patients exhibit low expression of OASIS due to high methylation of its promoter. Specific removal of this hypermethylation in glioblastomas transplanted into nude mice by epigenomic engineering suppresses the tumorigenesis. These findings suggest OASIS as a critical cell-cycle inhibitor with potential to act as a tumor suppressor.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Proteína Supressora de Tumor p53 , Camundongos , Animais , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Camundongos Nus , Pontos de Checagem do Ciclo Celular , Fatores Ativadores da Transcrição/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo
6.
Biomed Pharmacother ; 161: 114544, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36934558

RESUMO

Activating transcription factors, ATFs, is a family of transcription factors that activate gene expression and transcription by recognizing and combining the cAMP response element binding proteins (CREB). It is present in various viruses as a cellular gene promoter. ATFs is involved in regulating the mammalian gene expression that is associated with various cell physiological processes. Therefore, ATFs play an important role in maintaining the intracellular homeostasis. ATF2 and ATF3 is mostly involved in mediating stress responses. ATF4 regulates the oxidative metabolism, which is associated with the survival of cells. ATF5 is presumed to regulate apoptosis, and ATF6 is involved in the regulation of endoplasmic reticulum stress (ERS). ATFs is actively studied in oncology. At present, there has been an increasing amount of research on ATFs for the treatment of neurological diseases. Here, we have focused on the different types of ATFs and their association with Alzheimer's disease (AD). The level of expression of different ATFs have a significant difference in AD patients when compared to healthy control. Recent studies have suggested that ATFs are implicated in the pathogenesis of AD, such as neuronal repair, maintenance of synaptic activity, maintenance of cell survival, inhibition of apoptosis, and regulation of stress responses. In this review, the potential role of ATFs for the treatment of AD has been highlighted. In addition, we have systematically reviewed the progress of research on ATFs in AD. This review will provide a basic and innovative understanding on the pathogenesis and treatment of AD.


Assuntos
Doença de Alzheimer , Animais , Humanos , Doença de Alzheimer/tratamento farmacológico , Fatores Ativadores da Transcrição/genética , Fatores Ativadores da Transcrição/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Estresse do Retículo Endoplasmático/fisiologia , Mamíferos
7.
Cells ; 12(4)2023 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-36831248

RESUMO

Developing novel therapeutics often follows three steps: target identification, design of strategies to suppress target activity and drug development to implement the strategies. In this review, we recount the evidence identifying the basic leucine zipper transcription factors ATF5, CEBPB, and CEBPD as targets for brain and other malignancies. We describe strategies that exploit the structures of the three factors to create inhibitory dominant-negative (DN) mutant forms that selectively suppress growth and survival of cancer cells. We then discuss and compare four peptides (CP-DN-ATF5, Dpep, Bpep and ST101) in which DN sequences are joined with cell-penetrating domains to create drugs that pass through tissue barriers and into cells. The peptide drugs show both efficacy and safety in suppressing growth and in the survival of brain and other cancers in vivo, and ST101 is currently in clinical trials for solid tumors, including GBM. We further consider known mechanisms by which the peptides act and how these have been exploited in rationally designed combination therapies. We additionally discuss lacunae in our knowledge about the peptides that merit further research. Finally, we suggest both short- and long-term directions for creating new generations of drugs targeting ATF5, CEBPB, CEBPD, and other transcription factors for treating brain and other malignancies.


Assuntos
Peptídeos Penetradores de Células , Neoplasias , Humanos , Peptídeos Penetradores de Células/farmacologia , Encéfalo/metabolismo , Desenvolvimento de Medicamentos , Proteína delta de Ligação ao Facilitador CCAAT , Fatores Ativadores da Transcrição/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT
8.
Int J Mol Sci ; 24(4)2023 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-36834738

RESUMO

The highly dynamic changes in microglia necessary to achieve a rapid neuroinflammatory response require a supply of energy from mitochondrial respiration, which leads to the accumulation of unfolded mitochondrial proteins. We previously reported that microglial activation is correlated with the mitochondrial unfolded protein response (UPRmt) in a kaolin-induced hydrocephalus model, but we still do not know the extent to which these changes in microglia are involved in cytokine release. Here, we investigated the activation of BV-2 cells and found that treatment with lipopolysaccharide (LPS) for 48 h increased the secretion of pro-inflammatory cytokines. This increase was accompanied by a concurrent decrease in oxygen consumption rate (OCR) and mitochondrial membrane potential (MMP), in association with the up-regulation of the UPRmt. Inhibition of the UPRmt by knockdown of ATF5, a key upstream regulator of the UPRmt, using small-interfering RNA against ATF5 (siATF5) not only increased production of the pro-inflammatory cytokines, interleukin-6 (IL-6), IL-1ß and tumor necrosis factor-α (TNF-α), but also decreased MMP. Our results suggest that ATF5-dependent induction of the UPRmt in microglia acts as a protective mechanism during neuroinflammation and may be a potential therapeutic target for reducing neuroinflammation.


Assuntos
Citocinas , Microglia , Fatores Ativadores da Transcrição/metabolismo , Citocinas/metabolismo , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Microglia/metabolismo , Doenças Neuroinflamatórias , Fator de Necrose Tumoral alfa/metabolismo
9.
Exp Mol Med ; 55(2): 413-425, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36765266

RESUMO

Glutathione (GSH), an abundant nonprotein thiol antioxidant, participates in several biological processes and determines the functionality of stem cells. A detailed understanding of the molecular network mediating GSH dynamics is still lacking. Here, we show that activating transcription factor-2 (ATF2), a cAMP-response element binding protein (CREB), plays a crucial role in maintaining the level and activity of GSH in human mesenchymal stem cells (MSCs) by crosstalking with nuclear factor erythroid-2 like-2 (NRF2), a well-known master regulator of cellular redox homeostasis. Priming with ascorbic acid 2-glucoside (AA2G), a stable vitamin C derivative, increased the expression and activity of ATF2 in MSCs derived from human embryonic stem cells and umbilical cord. Subsequently, activated ATF2 crosstalked with the CREB1-NRF2 pathway to preserve the GSH dynamics of MSCs through the induction of genes involved in GSH synthesis (GCLC and GCLM) and redox cycling (GSR and PRDX1). Accordingly, shRNA-mediated silencing of ATF2 significantly impaired the self-renewal, migratory, proangiogenic, and anti-inflammatory capacities of MSCs, and these defects were rescued by supplementation of the cells with GSH. In addition, silencing ATF2 attenuated the ability of MSCs to alleviate airway inflammatory responses in an ovalbumin-induced mouse model of allergic asthma. Consistently, activation of ATF2 by overexpression or the AA2G-based priming procedure enhanced the core functions of MSCs, improving the in vivo therapeutic efficacy of MSCs for treating asthma. Collectively, our findings suggest that ATF2 is a novel modulator of GSH dynamics that determines the core functionality and therapeutic potency of MSCs used to treat allergic asthma.


Assuntos
Asma , Células-Tronco Mesenquimais , Animais , Humanos , Camundongos , Fatores Ativadores da Transcrição/metabolismo , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Asma/metabolismo , Glutationa/metabolismo , Fatores Imunológicos , Inflamação/metabolismo , Células-Tronco Mesenquimais/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo
10.
Life Sci ; 315: 121339, 2023 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-36621538

RESUMO

AIMS: Liver is a pivotal organ for sepsis-induced injury and approximately 40 % of liver injury results from sepsis. During hepatic injury, monocyte-to-macrophage differentiation is a key event because it results in the regulation of immune response. Asialoglycoprotein receptor 1 (ASGR1) is enriched in classical monocyte of peripheral blood mononuclear cells (PBMCs). We aimed to explore the effect of ASGR1 on monocyte-to-macrophage differentiation and the modulation of sepsis-induced liver injury. MAIN METHODS: ASGR1-knockdown/overexpression THP-1 cells and mice bone marrow-derived macrophages (BMDMs) induced by PMA and 30 % L929-cell conditioned medium were utilized to test the impact of ASGR1 on monocyte-to-macrophage differentiation and molecular mechanism respectively. Expression of differentiation specific factors were assessed via flow cytometry and real-time quantitative PCR. RNA-sequencing (RNA-seq) analysis revealed the effect of ASGR1 on monocyte-to-macrophage differentiation. Further, differentiation specific factors ATF5 and NF-κB pathways were examined via Western blot. The interaction between ASGR1 and ATF5 was further examined by co-IP. Finally, LPS-induced ASGR1-knockdown mice sepsis was used to investigate the effect of ASGR1 on monocyte-to-macrophage differentiation, liver injury and survival. KEY FINDINGS: ASGR1 promoted monocyte-to-macrophage differentiation via up-regulating CD68, F4/80 and CD86. Additionally, inhibited-ASGR1 decreased ATF5 expression by suppressing phosphorylation of NF-κB and IKBa in vitro and in vivo. ASGR1-knockdown mice suppressed Ly6Chi inflammatory monocytes in PBMCs, and restrained CD45+CD11bhiF4/80+Ly6Clo monocyte-derived macrophages and CD45+CD11b+F4/80+Ly6C+ inflammatory macrophages in livers. It also suppressed the level of IL-1ß, IL-6, TNF-α and alleviated liver injury and improved survival after sepsis. SIGNIFICANCE: ASGR1 is a negative regulator for sepsis-induced liver injury and survival.


Assuntos
Doença Hepática Crônica Induzida por Substâncias e Drogas , Sepse , Camundongos , Animais , Monócitos/metabolismo , NF-kappa B/metabolismo , Leucócitos Mononucleares/metabolismo , Doença Hepática Crônica Induzida por Substâncias e Drogas/metabolismo , Macrófagos/metabolismo , Diferenciação Celular , Sepse/complicações , Sepse/metabolismo , Camundongos Endogâmicos C57BL , Fatores Ativadores da Transcrição/metabolismo
11.
Cancer Immunol Immunother ; 72(4): 815-826, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36063172

RESUMO

Immune suppressive factors of the tumor microenvironment (TME) undermine viability and exhaust the activities of the intratumoral cytotoxic CD8 + T lymphocytes (CTL) thereby evading anti-tumor immunity and decreasing the benefits of immune therapies. To counteract this suppression and improve the efficacy of therapeutic regimens, it is important to identify and understand the critical regulators within CD8 + T cells that respond to TME stress and tumor-derived factors. Here we investigated the regulation and importance of activating transcription factor-4 (ATF4) in CTL using a novel Atf4ΔCD8 mouse model lacking ATF4 specifically in CD8 + cells. Induction of ATF4 in CD8 + T cells occurred in response to antigenic stimulation and was further increased by exposure to tumor-derived factors and TME conditions. Under these conditions, ATF4 played a critical role in the maintenance of survival and activities of CD8 + T cells. Conversely, selective ablation of ATF4 in CD8 + T cells in mice rendered these Atf4ΔCD8 hosts prone to accelerated growth of implanted tumors. Intratumoral ATF4-deficient CD8 + T cells were under-represented compared to wild-type counterparts and exhibited impaired activation and increased apoptosis. These findings identify ATF4 as an important regulator of viability and activity of CD8 + T cells in the TME and argue for caution in using agents that could undermine these functions of ATF4 for anti-cancer therapies.


Assuntos
Linfócitos do Interstício Tumoral , Neoplasias , Camundongos , Animais , Linfócitos T CD8-Positivos , Linfócitos T Citotóxicos , Fatores Ativadores da Transcrição , Microambiente Tumoral
12.
Front Immunol ; 14: 1282996, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38223508

RESUMO

Backgrounds: Cutaneous T cell lymphoma (CTCL) is a non-Hodgkin lymphoma characterized by skin infiltration of malignant T cells. The biological overlap between malignant T cells and their normal counterparts has brought obstacles in identifying tumor-specific features and mechanisms, limiting current knowledge of CTCL pathogenesis. Transcriptional dysregulation leading to abnormal gene expression profiles contributes to the initiation, progression and drug resistance of cancer. Therefore, we aimed to identify tumor-specific transcription factor underlying CTCL pathology. Methods: We analyzed and validated the differentially expressed genes (DEGs) in malignant T cells based on single-cell sequencing data. Clinical relevance was evaluated based on progression-free survival and time to next treatment. To determine the functional importance, lentivirus-mediated gene knockdown was conducted in two CTCL cell lines Myla and H9. Cell survival was assessed by examining cell viability, colony-forming ability, in-vivo tumor growth in xenograft models, apoptosis rate and cell-cycle distribution. RNA sequencing was employed to investigate the underlying mechanisms. Results: Activating transcription factor 5 (ATF5) was overexpressed in malignant T cells and positively correlated with poor treatment responses in CTCL patients. Mechanistically, ATF5 promoted the survival of malignant T cells partially through the PI3K/AKT/mTOR pathway, and imparted resistance to endoplasmic reticulum (ER) stress-induced apoptosis. Conclusions: These findings revealed the tumor-specific overexpression of the transcription factor ATF5 with its underlying mechanisms in promoting tumor survival in CTCL, providing new insight into the understanding of CTCL's pathology.


Assuntos
Fatores Ativadores da Transcrição , Linfoma Cutâneo de Células T , Neoplasias Cutâneas , Humanos , Fatores Ativadores da Transcrição/genética , Sobrevivência Celular/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Linfoma Cutâneo de Células T/patologia , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Linfócitos T/metabolismo , Serina-Treonina Quinases TOR
13.
Phytomedicine ; 107: 154428, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36115171

RESUMO

BACKGROUND: 24-epibrassinolide (EBR) is a ubiquitous steroidal phytohormone with anticancer activity. Yet the cytotoxic effects and mechanism of EBR on hepatocarcinoma (HCC) cells remain elusive. METHODS: Cell counting kit-8 (CCK-8) assay was performed to evaluate cell viability. Real-time cell analysis (RTCA) technology and colony formation assays were used to evaluate cell proliferation. The apoptosis ratio was measured by flow cytometry. Seahorse XFe96 was applied to detect the effects of EBR on cellular bioenergetics. RNA-seq analysis was performed to investigate differences in gene expression profiles. Western blot and qRT-PCR were used to detect the changes in target molecules. RESULTS: EBR induced apoptosis and caused energy restriction in HCC, both of which were related to insulin-like growth factor-binding protein 1 (IGFBP1). EBR rapidly and massively induced IGBFP1, part of which was transcribed by activating transcription factor-4 (ATF4). The accumulation of secreted and cellular IGFBP1 had different important roles, in which secreted IGFBP1 affected cell energy metabolism by inhibiting the phosphorylation of Akt, while intracellular IGFBP1 acted as a pro-survival factor to resist apoptosis. Interestingly, the extracellular signal-regulated kinase (ERK) inhibitor SCH772984 and MAP/ERK kinase (MEK) inhibitor PD98059 not only attenuated the EBR-induced IGFBP1 expression but also the basal expression of IGFBP1. Thus, the treatment of cells with these inhibitors further enhances the cytotoxicity of EBR. CONCLUSION: Taken together, these findings suggested that EBR can be considered as a potential therapeutic compound for HCC due to its pro-apoptosis, restriction of energy metabolism, and other anti-cancer properties. Meanwhile, the high expression of IGFBP1 induced by EBR in HCC contributes to our understanding of the role of IGFBP1 in drug resistance.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Somatomedinas , Fatores Ativadores da Transcrição/farmacologia , Apoptose , Brassinosteroides , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular , Quinases de Proteína Quinase Ativadas por Mitógeno , Reguladores de Crescimento de Plantas/farmacologia , Somatomedinas/farmacologia , Esteroides Heterocíclicos
14.
Ecotoxicol Environ Saf ; 245: 114087, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-36122457

RESUMO

Automobile exhaust-derived particulate matter 2.5 (PM2.5) can cause spermatogenic cell damage, potentially resulting in male infertility. This study uses male prepubertal Sprague Dawley (SD) rats to explore the molecular mechanisms by which automobile exhaust-derived PM2.5 causes spermatogenic cell damage and induces spermatogenesis dysfunction during sexual maturity by disrupting the mitochondrial unfolded protein response (UPRmt) in spermatogenic cells. Male prepubertal SD rats were randomly divided into four groups: control (intratracheal instillation of normal saline), low-dose PM2.5 (5 mg/kg), high-dose PM2.5 (10 mg/kg), and PM2.5 10 mg/kg +Vit (100 mg/kg of vitamin C and 50 mg/kg of vitamin E). The rats were treated for four weeks, with five consecutive treatment days and two non-treatment days, followed by cohabitation. Testicular and epididymal tissues were harvested for analysis. The mitochondria in spermatogenic cells were observed under an electron microscope. UPRmt-, oxidative stress-, and apoptosis-related markers in spermatogenic cells were examined. Spermatogenic cell numbers and conception rate declined significantly with increasing PM2.5 dose, with their mitochondria becoming vacuolated, swollen, and degenerated to varying degrees. The apoptosis of spermatogenic cells was abnormally enhanced in PM2.5 exposed groups compared to the control group. Spermatogenic cell numbers of conception rate gradually recovered, mitochondrial damage in spermatogenic cells was alleviated, and spermatogenic cell apoptosis was significantly reduced after vitamin intervention. In addition, protein levels of superoxide dismutase 1 (Sod1), nuclear factor erythroid 2-related factor 2 (Nrf2), and B-cell lymphoma 2 (Bcl-2) were significantly lower, while those of Bcl2-associated X apoptosis regulator (Bax), cleaved caspase 3 (Casp3), and cytochrome c (Cyt-c) and malondialdehyde (MDA) levels were significantly higher in the high-dose PM2.5 group than in the control group. The levels of UPRmt-related proteins C/EBP homologous protein (Chop), heat shock protein 60 (Hsp60), and activating transcription factors 4 (Atf4) and 5 (Atf5) were higher in the low-dose PM2.5 group, lower in the high-dose PM2.5 group, and gradually recovered in PM2.5 10 mg/kg +Vit group. Our results show that exposure to automobile exhaust-derived PM2.5 induces oxidative stress responses, leads to post-sexual maturation UPRmt dysfunction and mitochondrial impairment, and abnormally enhances spermatogenic cell apoptosis in prepubertal rats, resulting in male infertility.


Assuntos
Infertilidade Masculina , Emissões de Veículos , Fatores Ativadores da Transcrição , Animais , Apoptose , Ácido Ascórbico , Caspase 3/metabolismo , Chaperonina 60 , Citocromos c , Humanos , Masculino , Malondialdeído/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Material Particulado/toxicidade , Ratos , Ratos Sprague-Dawley , Solução Salina , Espermatogênese , Superóxido Dismutase-1 , Emissões de Veículos/toxicidade , Vitamina E/farmacologia , Vitaminas , Proteína X Associada a bcl-2/metabolismo
15.
Int J Mol Sci ; 23(13)2022 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-35806136

RESUMO

Activating transcription factor 5 (ATF5) belongs to the activating transcription factor/cyclic adenosine monophosphate (cAMP) response element-binding protein family of basic region leucine zipper transcription factors. ATF5 plays an important role in cell stress regulation and is involved in cell differentiation and survival, as well as centrosome maintenance and development. Accumulating evidence demonstrates that ATF5 plays an oncogenic role in cancer by regulating gene expressions involved in tumorigenesis and tumor survival. Recent studies have indicated that ATF5 may also modify the gene expressions involved in other diseases. This review explores in detail the regulation of ATF5 expression and signaling pathways and elucidates the role of ATF5 in cancer biology. Furthermore, an overview of putative therapeutic strategies that can be used for restoring aberrant ATF5 activity in different cancer types is provided.


Assuntos
Fatores Ativadores da Transcrição , Neoplasias , Fatores Ativadores da Transcrição/genética , Fatores Ativadores da Transcrição/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Humanos , Neoplasias/genética
16.
Int J Mol Sci ; 23(9)2022 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-35563423

RESUMO

With the advancement of science and technology, humans are chronically exposed to ionizing radiation. It is crucial to look for efficient and low-toxic anti-radiation agents. Through preliminary screening, we found that Acanthopanax senticosus polysaccharide (ASPS) played a major role in regulating immune damage caused by radiation. The objective of this study was to apply the Caenorhabditis elegans-P. aeruginosa (PA14) infection model to illuminate the mechanism of ASPS increasing the pathogen resistance of radiation-damaged nematodes. Results indicated that ASPS (1 mg/mL) significantly enhanced the pathogen resistance of radiation-damaged nematodes by directly elevating the immune response of nematodes rather than by affecting the bacterial activity. Through further research on the p38 MAPK signaling pathway and related mutants, we found that ASPS functioned by the p38 MAPK pathway in the intestine, and SKN-1, ATF-7 as the downstream targets of PMK-1 participated the regulation of ASPS. In addition, ASPS markedly alleviated the stress status of damaged nematodes by regulating oxidative stress. Collectively, our findings suggest that ASPS enhances the pathogen resistance of radiation-damaged nematodes through the intestinal p38MAPK-SKN-1/ATF-7 pathway and stress response.


Assuntos
Fatores Ativadores da Transcrição , Proteínas de Caenorhabditis elegans , Polissacarídeos , Fatores de Transcrição , Proteínas Quinases p38 Ativadas por Mitógeno , Fatores Ativadores da Transcrição/genética , Fatores Ativadores da Transcrição/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/efeitos da radiação , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/efeitos da radiação , Proteínas de Ligação a DNA/metabolismo , Eleutherococcus , Imunidade Inata/genética , Intestinos/metabolismo , Polissacarídeos/metabolismo , Polissacarídeos/farmacologia , Estresse Fisiológico/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Genomics ; 114(3): 110350, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35346781

RESUMO

Robust protocols to examine 3D chromatin structure have greatly advanced knowledge of gene regulatory mechanisms. Here we focus on the cystic fibrosis transmembrane conductance regulator (CFTR) gene, which provides a paradigm for validating models of gene regulation built upon genome-wide analysis. We examine the mechanisms by which multiple cis-regulatory elements (CREs) at the CFTR gene coordinate its expression in intestinal epithelial cells. Using CRISPR/Cas9 to remove CREs, individually and in tandem, followed by assays of gene expression and higher-order chromatin structure (4C-seq), we reveal the cross-talk and dependency of two cell-specific intronic enhancers. The results suggest a mechanism whereby the locus responds when CREs are lost, which may involve activating transcription factors such as FOXA2. Also, by removing the 5' topologically-associating domain (TAD) boundary, we illustrate its impact on CFTR gene expression and architecture. These data suggest a multi-layered regulatory hierarchy that is highly sensitive to perturbations.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística , Elementos Facilitadores Genéticos , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Cromatina , Regulação da Expressão Gênica , Fatores Ativadores da Transcrição/genética , Fatores Ativadores da Transcrição/metabolismo
18.
J Cancer Res Ther ; 17(5): 1192-1201, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34850767

RESUMO

BACKGROUND: Receptor tyrosine-protein kinase erbB-2 (human epidermal growth factor receptor 2 [HER2])-based therapies can improve the prognosis of HER2-positive breast cancer (BRCA) patients; however, HER2-positive patients with distal metastasis do not gain significant clinical benefit from molecular targeted therapy. MATERIALS AND METHODS: A database analysis, immunohistochemistry, and quantitative real-time polymerase chain reaction were used to evaluate the expression of activating transcription factor 7 (ATF7) and its clinical value. A transwell chamber assay was used to assess migration and cell signaling was assessed by immunoblotting. RESULTS: ATF7 was expressed at a low level in HER2-enriched BRCA specimens compared with normal or HER2-negative specimens, which was corroborated in HER2-positive tissue chips and cultured cells. ATF7 gradually decreased with increased tumor stage and low ATF7 was associated with poor prognosis in HER2-positive BRCA patients. ATF7-upregulation inhibited, whereas ATF7-knockdown promoted migration, activity of matrix metalloproteinase 9 (MMP9), MMP2, and uridylyl phosphate adenosine and plasminogen activator inhibitor-1 (PAI-1) expression in HER2-positive cells. HER2 overexpression markedly reduced ATF7 expression in MCF-10A mammary epithelial cells, along with decreased E-cadherin, and increased N-cadherin and migration, which were abrogated by exogenous ATF7 transfection. Mechanistically, HER2 upregulation mediated the decline of ATF7 and activated histone lysine demethylase 1 (LSD1), followed by elevation of histone H3K9 dimethylation (H3K9me2) and H3K4me2. However, the enhanced effects on LSD1 and H3K9me2, excluding H3K4me2, were abrogated by exogenous ATF7. ATF7 was negatively associated with KDM1A (encoding LSD1 protein) expression. CONCLUSIONS: ATF7 may be a useful diagnostic and prognostic marker for metastatic HER2-positive BRCA. The ATF7/LSD1/H3K9me2 axis may be responsible for metastasis in HER2-positive cells.


Assuntos
Fatores Ativadores da Transcrição/metabolismo , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/patologia , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases/metabolismo , Receptor ErbB-2/metabolismo , Fatores Ativadores da Transcrição/genética , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Movimento Celular , Bases de Dados Factuais , Feminino , Histona Desmetilases/genética , Humanos , Prognóstico , Receptor ErbB-2/genética , Células Tumorais Cultivadas
19.
Bioengineered ; 12(2): 12070-12086, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34787047

RESUMO

Long non-coding RNA (lncRNA) prostate cancer-associated transcript 18 (PCAT18) is a potential diagnostic target for adenocarcinoma. However, its role in triple-negative breast cancer (TNBC) remains largely unknown. Based on data from an online database, a significant decline in lncRNA PCAT18 was observed in patients with TNBC subtype compared to a population with normal breast tissue. Patients with TNBC with high PCAT18 levels presented good outcomes. Patients with TNBC with high PCAT18 had a lower rate of lymph node-positive metastasis than those with low PCAT18. PCAT18-upregulation inhibited, while PCAT18-downregulation promoted, migration and expression of matrix metalloproteinases 9/2 (MMP9/MMP2) and uridylyl phosphate adenosine (uPA) in TNBC cells. Activating transcription factor 7 (ATF7) was positively associated with PCAT18, and ATF7-inhibition abrogated the anti-migration effects of PCAT18 on TNBC cells. Mechanistically, miR-103a-3p directly targeted and inhibited ATF7 expression. PCAT18 competitively sponges miR-103a-3p, promoting the expression of ATF7. Exogenous PCAT18 was associated with lower incidence of lung metastasis followed by the upregulation of ATF7, which was prevented by the treatment of miR-103a-3p mimics. Collectively, PCAT18 was expressed at low levels in TNBC, and PCAT18 could sponge miR-103a-3p and promote ATF7 expression, resulting in prevention of TNBC metastasis. Thus, PCAT18 can serve as a predictive factor for patients with metastatic TNBC.


Assuntos
Fatores Ativadores da Transcrição/genética , Neoplasias Pulmonares/secundário , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Regulação para Cima/genética , Fatores Ativadores da Transcrição/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Movimento Celular/genética , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , RNA Longo não Codificante/genética
20.
Sci Rep ; 11(1): 19796, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34611283

RESUMO

We previously reported that growth promoter-induced skeletal muscle hypertrophy co-ordinately upregulated expression of genes associated with an integrated stress response (ISR), as well as potential ISR regulators. We therefore used Adeno-Associated Virus (AAV)-mediated overexpression of these genes, individually or in combination, in mouse skeletal muscle to test whether they induced muscle hypertrophy. AAV of each target gene was injected into mouse Tibialis anterior (TA) and effects on skeletal muscle growth determined 28 days later. Individually, AAV constructs for Arginase-2 (Arg2) and Activating transcription factor-5 (Atf5) reduced hindlimb muscle weights and upregulated expression of genes associated with an ISR. AAV-Atf5 also decreased Myosin heavy chain (MyHC)-IIB mRNA, but increased MyHC-IIA and isocitrate dehydrogenase-2 (Idh2) mRNA, suggesting ATF5 is a novel transcriptional regulator of Idh2. AAV-Atf5 reduced the size of both TA oxidative and glycolytic fibres, without affecting fibre-type proportions, whereas Atf5 combined with Cebpg (CCAAT enhancer binding protein-gamma) only reduced the size of glycolytic fibres and tended to increase the proportion of oxidative fibres. It is likely that persistent Atf5 overexpression maintains activation of the ISR, thereby reducing protein synthesis and/or increasing protein degradation and possibly apoptosis, resulting in inhibition of muscle growth, with overexpression of Arg2 having a similar effect.


Assuntos
Fatores Ativadores da Transcrição/genética , Dependovirus/genética , Expressão Gênica , Vetores Genéticos/genética , Músculo Esquelético/metabolismo , Estresse Fisiológico , Transdução Genética , Fatores Ativadores da Transcrição/metabolismo , Animais , Metabolismo Energético , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Camundongos , RNA Mensageiro/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA