Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Theranostics ; 11(17): 8464-8479, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34373753

RESUMO

As glutamine plays a central role in cancer metabolism, inhibition of glutaminolysis has become an ideal anticancer therapeutic target. However, glutaminolysis inhibition leads to activation of autophagy, which compromises its antitumor effect. Hence, we investigated the mechanism underlying glutaminolysis inhibition-induced pro-survival autophagy. Methods: High-throughput sequencing was performed on colorectal cancer (CRC) cells before and after glutaminolysis inhibition to identify differentially expressed genes. Activating transcription factor 4 (ATF4) pathway enrichment in glutaminolysis inhibited cells was identified through gene set enrichment analysis. ATF4 expression was assessed by quantitative real-time PCR (qRT-PCR) and western blotting. The function of ATF4 on mechanistic target of rapamycin (mTOR) regulation was assessed by western blotting. Luciferase reporter assays and chromatin immunoprecipitation were used to confirm the regulation of DNA damage inducible transcript 4 (DDIT4) by ATF4. mRNA half-life assays, RNA immunoprecipitation, qRT-PCR and western blotting were performed to determine the relationship between FTO alpha-ketoglutarate dependent dioxygenase (FTO), YTH N6-methyladenosine RNA binding protein 2 (YTHDF2), and ATF4. ATF4 regulation of pro-survival autophagy was measured by tandem monomeric red fluorescent protein-green fluorescent protein fluorescence microscopy. Finally, the synergistic effect of autophagy and glutaminolysis inhibition was analyzed in an azoxymethane/dextran sodium sulfate mouse model. Results: The ATF4 pathway was activated in CRC cells upon glutaminolysis inhibition. Functionally, ATF4 transcriptionally upregulated DDIT4 to suppress mTOR, which induced pro-survival autophagy during glutaminolysis inhibition. Interestingly, glutaminolysis inhibition promoted ATF4 mRNA expression by abrogating N6-methyladenosine (m6A) modification and YTHDF2-mediated RNA decay. Finally, inhibition of ATF4-induced autophagy enhanced the antitumor efficacy of glutaminolysis inhibition. Conclusion: Glutaminolysis inhibition upregulated ATF4 expression in an m6A-dependent manner to activate pro-survival autophagy through transcriptional activation of the mTOR inhibitor DDIT4. Targeting ATF4-induced autophagy is a new strategy to synergize glutaminolysis-targeting therapies for cancer treatment.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Autofagia/fisiologia , Glutamina/metabolismo , Fator 4 Ativador da Transcrição/fisiologia , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Células HCT116 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estabilidade de RNA , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Fatores de Transcrição/metabolismo
2.
FASEB J ; 35(6): e21612, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33948996

RESUMO

Lipid overload is intimately connected with the change of endothelial epigenetic status which impacts cellular signaling activities and endothelial function. Activating transcription factor 4 (ATF4) is involved in the regulation of lipid metabolism and meanwhile an epigenetic modifier. However, the role of ATF4 in the angiogenesis under lipid overload is not well understood. Here, to induce lipid overload status, we employed high-fat diet (HFD)-induced obese mouse model in vivo and palmitic acid (PA) to stimulate endothelial cells in vitro. Compared with mice fed with normal chow diet (NCD), HFD-induced obese mice showed angiogenic defects evidenced by decline in (1) blood flow recovery after hind limb ischemia, (2) wound healing speed after skin injury, (3) capillary density in injured tissues and matrigel plugs, and (4) endothelial sprouts of aortic ring. ATF4 deficiency aggravated above angiogenic defects in mice while ATF4 overexpression improved the blunted angiogenic response. Mechanistically, lipid overload lowered the H3K4 methylation levels at the regulatory regions of NOS3 and ERK1 genes, leading to reduced angiogenic signaling activity. Methionine adenosyltransferase 2A (MAT2A) is identified as a target of ATF4 and formed complex with ATF4 to direct lysine methyltransferase 2A (MLL1) to the regulatory regions of both genes for the maintenance of the H3K4 methylation level and angiogenic signaling activity. Here, we uncovered a novel metabolic-epigenetic coupling orchestrated by the ATF4-MAT2A axis for angiogenesis. The ATF4-MAT2A axis links lipid overload milieu to altered epigenetic status of relevant angiogenic signaling in endothelial cells, suggesting a potential therapeutic target for angiogenesis impaired by lipid overload.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Epigênese Genética , Isquemia/patologia , Lipídeos/efeitos adversos , Metionina Adenosiltransferase/metabolismo , Neovascularização Patológica/patologia , Obesidade/complicações , Animais , Dieta Hiperlipídica , Isquemia/etiologia , Isquemia/metabolismo , Masculino , Metionina Adenosiltransferase/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Neovascularização Patológica/etiologia , Neovascularização Patológica/metabolismo
3.
Life Sci ; 275: 119356, 2021 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-33737080

RESUMO

AIMS: Growth and differentiation factor 15 (GDF15) is a stress-related factor, which implicated in various diseases. This study aimed to investigate the role of GDF15 in LPS-mediated inflammation and to explore the potential underlying molecular mechanisms in human nasal epithelial cells (HNEpCs). MAIN METHODS: HNEpCs were treated with LPS. GDF15 loss-of-function and gain-of-function experiments were performed. The expression of GDF15 by quantitative real-time PCR (RT-qPCR). The mRNA levels and secretion of inflammatory cytokines and MUC5AC were assessed by RT-qPCR and ELISA kits. LY294002 (PI3K inhibitor) and 740Y-P (PI3K agonist) were utilized to interfere with PI3k/Akt pathway. The relationship between GDF15 and ATF4 was identified by chromatin immunoprecipitation (ChIP) and luciferase reporter assay. KEY FINDINGS: We observed that LPS triggered GDF15 expression. GDF15 ablation reduced the mRNA levels and secretion of inflammatory cytokines. GDF15 silencing led to the reduction of the MUC5AC mRNA level, protein level and secretion in response to LPS. Enhanced expression of GDF15 showed the opposite results. Furthermore, we found that GDF15 deficiency inhibited activation of the PI3K/Akt pathway, LY294002 treatment further enhanced the role of GDF15 suppression in inflammation and MUC5AC expression, while 740Y-P administration partly reversed the biological activities of GDF15 silencing. ATF4 could bind to the promoter of GDF15 and positively regulate GDF15 expression. Depression of ATF4 diminished the secretion of inflammatory cytokines and MUC5AC via regulation of GDF15. SIGNIFICANCE: Our data suggest that GDF15 is regulated by ATF4 and suppresses LPS-induced inflammation and MUC5AC in human nasal epithelial cells through the PI3K/Akt pathway.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Fator 15 de Diferenciação de Crescimento/fisiologia , Inflamação/metabolismo , Mucina-5AC/metabolismo , Mucosa Nasal/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fator 4 Ativador da Transcrição/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Citocinas/metabolismo , Citocinas/fisiologia , Fator 15 de Diferenciação de Crescimento/metabolismo , Humanos , Inflamação/fisiopatologia , Lipopolissacarídeos/farmacologia , Mucina-5AC/fisiologia , Mucosa Nasal/fisiopatologia , Reação em Cadeia da Polimerase em Tempo Real
4.
J Mol Med (Berl) ; 99(3): 383-402, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33409554

RESUMO

Endoplasmic reticulum (ER) stress is a common threat to photoreceptors during the pathogenesis of chronic retinopathies and often results in irreversible visual impairment. 2,3,5,6-Tetramethylpyrazine (TMP), which possesses many beneficial pharmacological activities, is a potential drug that could be used to protect photoreceptors. In the present study, we found that the cellular growth rate of 661 W cells cultured under low glucose conditions was lower than that of control cells, while the G2/M phase of the cell cycle was longer. We further found that the mitochondrial membrane potential (ΔΨm) was lower and that ER stress factor expression was increased in 661 W cells cultured under low glucose conditions. TMP reversed these trends. Visual function and cell counts in the outer nuclear layer (ONL) were low and the TUNEL-positive rate in the ONL was high in a C3H mouse model of spontaneous retinal degeneration. Similarly, visual function was decreased, and the TUNEL-positive rate in the ONL was increased in fasted C57/BL6j mice compared with control mice. On the other hand, ER stress factor expression was found to be increased in the retinas of both mouse models, as shown by reverse transcription real-time PCR (RT-qPCR) and western blotting. TMP reversed the physiological and molecular biological variations observed in both mouse models, and ATF4 expression was enhanced again. Further investigation by using western blotting illustrated that the proportion of insoluble prion protein (PRP) versus soluble PRP was reduced both in vitro and in vivo. Taken together, these results suggest that TMP increased the functions of photoreceptors by alleviating ER stress in vitro and in vivo, and the intrinsic mechanism was the ATF4-mediated inhibition of PRP aggregation. TMP may potentially be used clinically as a therapeutic agent to attenuate the functional loss of photoreceptors during the pathogenesis of chronic retinopathies. KEY MESSAGES: • Already known: TMP is a beneficial drug mainly used in clinic to enhance organ functions, and the intrinsic mechanism is still worthy of exploring. • New in the study: We discovered that TMP ameliorated retinal photoreceptors function via ER stress alleviation, which was promoted by ATF4-mediated inhibition of PRP aggregation. • Application prospect: In prospective clinical practices, TMP may potentially be used in the clinic as a therapeutic agent to attenuate the photoreceptors functional reduction in chronic retinopathies.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Proteínas Priônicas/efeitos dos fármacos , Agregados Proteicos/efeitos dos fármacos , Agregação Patológica de Proteínas/prevenção & controle , Pirazinas/farmacologia , Células Fotorreceptoras Retinianas Cones/efeitos dos fármacos , Degeneração Retiniana/prevenção & controle , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Eletrorretinografia , Proteínas do Olho/biossíntese , Proteínas do Olho/genética , Jejum , Feminino , Glucose/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Proteínas Priônicas/química , Agregação Patológica de Proteínas/metabolismo , Retina/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Degeneração Retiniana/fisiopatologia , Método Simples-Cego , Solubilidade , Organismos Livres de Patógenos Específicos , Transcrição Gênica/efeitos dos fármacos
5.
Biomed Pharmacother ; 127: 110145, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32361164

RESUMO

Parkinson disease (PD) is the second common neurodegenerative disorder after Alzheimer's disease (AD). The predominant pathological hallmark is progressive loss of dopaminergic (DA) neurones in the substantia nigra (SN) complicated by aggregation of misfolded forms of alpha-synuclein (α-syn). α-syn is a cytosolic synaptic protein localized in the presynaptic neuron under normal circumstances. What drives misfolding of this protein is largely unknown. However, recent studies suggest that autophagy might be an important risk factor for contributing towards PD. Autophagy is an evolutionarily conserved mechanism that causes the clearance or degradation of misfolded, mutated and damaged proteins, organelles etc. However, in an aging individual this process might deteriorate which could possibly lead to the accumulation of damaged proteins. Hence, autophagy modulation might provide some interesting cues for the treatment of PD. Additionally, Fibroblast growth factor 21 (FGF21) which is known for its role as a potent regulator of glucose and energy metabolism has also proved to be neuroprotective in various neurodegenerative conditions possibly via mediation of autophagy.


Assuntos
Autofagia/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Doença de Parkinson/etiologia , Fator 4 Ativador da Transcrição/fisiologia , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/fisiologia , Doença de Parkinson/patologia , Fatores de Transcrição/fisiologia
6.
Cells ; 8(8)2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-31416289

RESUMO

BACKGROUND: Nod-like receptor protein 3 (NLRP3) inflammasome is a crucial contributor in the inflammatory process during cerebral ischemia/reperfusion (I/R) injury. ATF4 plays a pivotal role in the pathogenesis of cerebral I/R injury, however, its function and underlying mechanism are not fully characterized yet. In the current study, we examined whether ATF4 ameliorates cerebral I/R injury by inhibiting NLRP3 inflammasome activation and whether mitophagy is involved in this process. In addition, we explored the role of parkin in ATF4-mediated protective effects. Method: To address these issues, healthy male adult Sprague-Dawley rats were exposed to middle cerebral artery occlusion for 1 h followed by 24 h reperfusion. Adeno-associated virus (AAV) and siRNA were injected into rats to overexpress and knockdown ATF4 expression, respectively. After pretreatment with AAV, mdivi-1(mitochondrial division inhibitor-1) was injected into rats to block mitophagy activity. Parkin expression was knockdown using specific siRNA after AAV pretreatment. Result: Data showed that ATF4 overexpression induced by AAV was protective against cerebral I/R injury, as evidenced by reduced cerebral infraction volume, decreased neurological scores and improved outcomes of HE and Nissl staining. In addition, overexpression of ATF4 gene was able to up-regulate Parkin expression, enhance mitophagy activity and inhibit NLRP3 inflammasome-mediated inflammatory response. ATF4 knockdown induced by siRNA resulted in the opposite effects. Furthermore, ATF4-mediated inhibition of NLRP3 inflammasome activation was strongly affected by mitophagy blockage upon mdivi-1 injection. Besides, ATF4-mediated increase of mitophagy activity and inhibition of NLRP3 inflammasome activation were effectively reversed by Parkin knockdown using siRNA. Conclusion: Our study demonstrated that ATF4 is able to alleviate cerebral I/R injury by suppressing NLRP3 inflammasome activation through parkin-dependent mitophagy activity. These results may provide a new strategy to relieve cerebral I/R injury by modulating mitophagy-NLRP3 inflammasome axis.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Isquemia Encefálica/metabolismo , Inflamassomos/metabolismo , Mitocôndrias/metabolismo , Mitofagia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Traumatismo por Reperfusão/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Masculino , Ratos , Ratos Sprague-Dawley
7.
Oncogene ; 38(35): 6301-6318, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31312022

RESUMO

Cancer cells exploit many of the cellular adaptive responses to support their survival needs. One such critical pathway in eukaryotic cells is the unfolded protein response (UPR) that is important in normal physiology as well as disease states, including cancer. Since UPR can serve as a lever between survival and death, regulated control of its activity is critical for tumor formation and growth although the underlying mechanisms are poorly understood. Here we show that one of the main transcriptional effectors of UPR, activating transcription factor 4 (ATF4), is essential for prostate cancer (PCa) growth and survival. Using systemic unbiased gene expression and proteomic analyses, we identified a novel direct ATF4 target gene, family with sequence similarity 129 member A (FAM129A), which is critical in mediating ATF4 effects on prostate tumorigenesis. Interestingly, FAM129A regulated both PERK and eIF2α in a feedback loop that differentially channeled the UPR output. ATF4 and FAM129A protein expression is increased in patient PCa samples compared with benign prostate. Importantly, in vivo therapeutic silencing of ATF4-FAM129A axis profoundly inhibited tumor growth in a preclinical PCa model. These data support that one of the canonical UPR branches, through ATF4 and its target gene FAM129A, is required for PCa growth and thus may serve as a novel therapeutic target.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Biomarcadores Tumorais/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias da Próstata/metabolismo , Resposta a Proteínas não Dobradas/genética , Animais , Proliferação de Células/genética , Estresse do Retículo Endoplasmático/genética , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Masculino , Camundongos , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Transdução de Sinais/genética , Células Tumorais Cultivadas
8.
Br J Cancer ; 119(12): 1538-1551, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30385822

RESUMO

BACKGROUND: Although NGLY1 is known as a pivotal enzyme that catalyses the deglycosylation of denatured glycoproteins, information regarding the responses of human cancer and normal cells to NGLY1 suppression is limited. METHODS: We examined how NGLY1 expression affects viability, tumour growth, and responses to therapeutic agents in melanoma cells and an animal model. Molecular mechanisms contributing to NGLY1 suppression-induced anticancer responses were revealed by systems biology and chemical biology studies. Using computational and medicinal chemistry-assisted approaches, we established novel NGLY1-inhibitory small molecules. RESULTS: Compared with normal cells, NGLY1 was upregulated in melanoma cell lines and patient tumours. NGLY1 knockdown caused melanoma cell death and tumour growth retardation. Targeting NGLY1 induced pleiotropic responses, predominantly stress signalling-associated apoptosis and cytokine surges, which synergise with the anti-melanoma activity of chemotherapy and targeted therapy agents. Pharmacological and molecular biology tools that inactivate NGLY1 elicited highly similar responses in melanoma cells. Unlike normal cells, melanoma cells presented distinct responses and high vulnerability to NGLY1 suppression. CONCLUSION: Our work demonstrated the significance of NGLY1 in melanoma cells, provided mechanistic insights into how NGLY1 inactivation leads to eradication of melanoma with limited impact on normal cells, and suggested that targeting NGLY1 represents a novel anti-melanoma strategy.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Interferon gama/fisiologia , Melanoma/tratamento farmacológico , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/antagonistas & inibidores , Fator 4 Ativador da Transcrição/fisiologia , Animais , Células Cultivadas , Citocinas/análise , Perfilação da Expressão Gênica , Humanos , Interferon gama/genética , Melanoma/patologia , Camundongos , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/fisiologia , Células-Tronco Pluripotentes/fisiologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Transdução de Sinais/fisiologia , Fator de Transcrição CHOP/fisiologia
9.
Mol Metab ; 17: 39-48, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30174228

RESUMO

OBJECTIVE: Loss of insulin secretion due to failure or death of the insulin secreting ß cells is the central cause of diabetes. The cellular response to stress (endoplasmic reticulum (ER), oxidative, inflammatory) is essential to sustain normal ß cell function and survival. Pancreatic and duodenal homeobox 1 (PDX1), Activating transcription factor 4 (ATF4), and Activating transcription factor 5 (ATF5) are transcription factors implicated in ß cell survival and susceptibility to stress. Our goal was to determine if a PDX1-ATF transcriptional complex or complexes regulate ß cell survival in response to stress and to identify direct transcriptional targets. METHODS: Pdx1, Atf4 and Atf5 were silenced by viral delivery of gRNAs or shRNAs to Min6 insulinoma cells or primary murine islets. Gene expression was assessed by qPCR, RNAseq analysis, and Western blot analysis. Chromatin enrichment was measured in the Min6 ß cell line and primary isolated mouse islets by ChIPseq and ChIP PCR. Immunoprecipitation was used to assess interactions among transcription factors in Min6 cells and isolated mouse islets. Activation of caspase 3 by immunoblotting or by irreversible binding to a fluorescent inhibitor was taken as an indication of commitment to an apoptotic fate. RESULTS: RNASeq identified a set of PDX1, ATF4 and ATF5 co-regulated genes enriched in stress and apoptosis functions. We further identified stress induced interactions among PDX1, ATF4, and ATF5. PDX1 chromatin occupancy peaks were identified over composite C/EBP-ATF (CARE) motifs of 26 genes; assessment of a subset of these genes revealed co-enrichment for ATF4 and ATF5. PDX1 occupancy over CARE motifs was conserved in the human orthologs of 9 of these genes. Of these, Glutamate Pyruvate Transaminase 2 (Gpt2), Cation transport regulator 1 (Chac1), and Solute Carrier Family 7 Member 1 (Slc7a1) induction by stress was conserved in human islets and abrogated by deficiency of Pdx1, Atf4, and Atf5 in Min6 cells. Deficiency of Gpt2 reduced ß cell susceptibility to stress induced apoptosis in both Min6 cells and primary islets. CONCLUSIONS: Our results identify a novel PDX1 stress inducible complex (es) that regulates expression of stress and apoptosis genes to govern ß cell survival.


Assuntos
Fatores Ativadores da Transcrição/fisiologia , Proteínas de Homeodomínio/fisiologia , Células Secretoras de Insulina/citologia , Transativadores/fisiologia , Fator 4 Ativador da Transcrição/metabolismo , Fator 4 Ativador da Transcrição/fisiologia , Fatores Ativadores da Transcrição/metabolismo , Animais , Apoptose/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Diabetes Mellitus/metabolismo , Modelos Animais de Doenças , Retículo Endoplasmático/fisiologia , Regulação da Expressão Gênica/genética , Genes Homeobox , Proteínas de Homeodomínio/metabolismo , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Estresse Fisiológico/fisiologia , Transativadores/metabolismo , Transcriptoma/genética
10.
J Nutr Sci Vitaminol (Tokyo) ; 64(3): 200-208, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29962431

RESUMO

Fibroblast growth factor 21 (FGF21), mainly synthesized and secreted from the liver, is an endocrine FGF that regulates glucose and fatty acid metabolism to maintain whole body energy homeostasis. Gene expression of FGF21 was previously reported to be induced by endoplasmic reticulum (ER) stress through activating transcription factor 4 (ATF4). It has been reported that drug-induced ER stress is reduced by overexpression of FGF21. However, the function of endogenous FGF21 under physiological conditions such as the postprandial state remains unknown. Here, we examined the effects of both endogenous and exogenous FGF21 on postprandial hepatic ER stress. In mice, postprandial and tunicamycin-induced ER stress was significantly reduced by overexpression of FGF21 using a recombinant adenovirus. FGF21-deficient mice exhibited a more considerable increase in drug-induced ER stress target gene expression than wild-type mice. Following refeeding after fasting, FGF21 deficiency caused severe ER stress in the liver. The postprandial ER stress response was significantly reduced when hepatic FGF21 gene expression was increased by feeding a diet containing the soy protein ß-conglycinin which activates ATF4. Together, these results demonstrate that FGF21 reduces the increased expression of a subset of genes in the liver in response to ER stress and may correct metabolic disorders caused by ER stress.


Assuntos
Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/farmacologia , Fator 4 Ativador da Transcrição/efeitos dos fármacos , Fator 4 Ativador da Transcrição/fisiologia , Animais , Antígenos de Plantas/farmacologia , Estresse do Retículo Endoplasmático/genética , Jejum , Fatores de Crescimento de Fibroblastos/deficiência , Fatores de Crescimento de Fibroblastos/genética , Expressão Gênica/efeitos dos fármacos , Globulinas/farmacologia , Fígado/metabolismo , Fígado/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Período Pós-Prandial , Proteínas de Armazenamento de Sementes/farmacologia , Proteínas de Soja/farmacologia , Tunicamicina/farmacologia
11.
Biomed Res Int ; 2018: 3694362, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30003094

RESUMO

INTRODUCTION: Cardiomyocyte remodelling in atrial fibrillation (AF) has been associated with both oxidative stress and endoplasmic reticulum (ER) stress and is accompanied by a complex transcriptional regulation. Here, we investigated the role the oxidative stress and ER stress responsive bZIP transcription factor ATF4 plays in atrial cardiomyocyte viability and AF induced gene expression. METHODS: HL-1 cardiomyocytes were subjected to rapid field stimulation. Forced expression of ATF4 was achieved by adenoviral gene transfer. Using global gene expression analysis and chromatin immunoprecipitation, ATF4 dependent transcriptional regulation was studied, and tissue specimen of AF patients was analysed by immunohistochemistry. RESULTS: Oxidative stress and ER stress caused a significant reduction in cardiomyocyte viability and were associated with an induction of ATF4. Accordingly, ATF4 was also induced by rapid field stimulation mimicking AF. Forced expression of wild type ATF4 promoted cardiomyocyte death. ATF4 was demonstrated to bind to the promoters of several cell stress genes and to induce the expression of a number of ATF4 dependent stress responsive genes. Moreover, immunohistochemical analyses showed that ATF4 is expressed in the nuclei of cardiomyocytes of tissue specimen obtained from AF patients. CONCLUSION: ATF4 is expressed in human atrial cardiomyocytes and is induced in response to different types of cell stress. High rate electrical field stimulation seems to result in ATF4 induction, and forced expression of ATF4 reduces cardiomyocyte viability.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Fibrilação Atrial/genética , Miócitos Cardíacos , Animais , Fibrilação Atrial/fisiopatologia , Morte Celular , Estresse do Retículo Endoplasmático , Regulação da Expressão Gênica , Humanos
12.
BMB Rep ; 51(8): 388-393, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29636121

RESUMO

The activating transcription factor (ATF) 4 belongs to the ATF/CREB (cAMP Response Element Binding bZIP [Basic Leucine Zipper]) transcription factor family, and plays a central role in the UPR (Unfolded Protein Response) process in cells. The induction of ATF4 expression has previously been shown to increase the replication of HIV-1. However, the detailed mechanism underlying this effect and the factors involved in the regulation of ATF4 function are still unknown. Here, we demonstrate first that knocking out ATF4 using siRNA shows a strong negative effect on HIV-1 production, indicating that ATF4 is a functional positive cellular factor in HIV-1 production. To determine the mechanism by which ATF4 regulates the HIV-1 life cycle, we assessed the effect of the overexpression of wild type ATF4 and its various derivatives on HIV-1 LTR-mediated transcriptional activation and the production of HIV-1 particles. This effect was studied through co-transfection experiments with either reporter vectors or proviral DNA. We found that the N-terminal domains of ATF4 are involved in HIV-1 LTR-mediated transcriptional activation, and thus in HIV-1 production. [BMB Reports 2018; 51(8): 388-393].


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , HIV-1/fisiologia , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação Viral da Expressão Gênica , Células HEK293 , Infecções por HIV/genética , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Repetição Terminal Longa de HIV , HIV-1/genética , HIV-1/metabolismo , Humanos , Transcrição Gênica , Ativação Transcricional , Resposta a Proteínas não Dobradas
13.
PLoS One ; 13(2): e0191107, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29420561

RESUMO

The ATF4 transcription factor is a key regulator of the adaptive integrated stress response (ISR) induced by various stresses and pathologies. Identification of novel transcription targets of ATF4 during ISR would contribute to the understanding of adaptive networks and help to identify novel therapeutic targets. We were previously searching for genes that display an inverse regulation mode by the transcription factors ATF4 and p53 in response to mitochondrial respiration chain complex III inhibition. Among the selected candidates the human genes for cytokeratine 16 (KRT16), anti-apoptotic protein Niban (FAM129A) and hexokinase HKDC1 have been found highly responsive to ATF4 overexpression. Here we explored potential roles of the induction of KRT16, FAM129A and HKDC1 genes in ISR. As verified by RT-qPCR, a dysfunction of mitochondrial respiration chain and ER stress resulted in a partially ATF4-dependent stimulation of KRT16, FAM129A and HKDC1 expression in the HCT116 colon carcinoma cell line. ISRIB, a specific inhibitor of ISR, was able to downregulate the ER stress-induced levels of KRT16, FAM129A and HKDC1 transcripts. An inhibition of ATF4 by RNAi attenuated the induction of KRT16, FAM129A and HKDC1 mRNAs in response to ER stress or to a dysfunctional mitochondrial respiration. The similar induction of the three genes was observed in another tumor-derived cervical carcinoma cell line HeLa. However, in HaCaT and HEK293T cells that display transformed phenotypes, but do not originate from patient-derived tumors, the ER stress-inducing treatments resulted in an upregulation of FAM129A and HKDC1, but not KRT16 transcripts, By a luciferase reporter approach we identified a highly active ATF4-responsive element within the upstream region of the KRT16 gene. The results suggest a conditional regulation of KRT16 gene by ATF4 that may be inhibited in normal cells, but engaged during cancer progression. Potential roles of KRT16, FAM129A and HKDC1 genes upregulation in adaptive stress responses and pathologies are discussed.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Biomarcadores Tumorais/genética , Hexoquinase/genética , Queratina-16/genética , Proteínas de Neoplasias/genética , Estresse Fisiológico , Linhagem Celular , Estresse do Retículo Endoplasmático , Regulação da Expressão Gênica/genética , Humanos , Regiões Promotoras Genéticas , Interferência de RNA , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Regulação para Cima
14.
J Orthop Res ; 36(5): 1334-1345, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29080374

RESUMO

Although the number of patients with intervertebral disc (IVD) degeneration is increasing in aging societies, its etiology and pathogenesis remain elusive and there is currently no effective treatment to prevent this undesirable condition. The unfolded protein response (UPR) is a cellular machinery that plays critical roles in handling endoplasmic reticulum (ER) stress, a condition caused by the accumulation of unfolded proteins in the ER lumen. This study aimed to elucidate the potential role of the UPR mediated by pancreatic endoplasmic reticulum kinase (PERK), one of the major ER stress sensors in mammalian cells, in the development of IVD degeneration. IVD degeneration was artificially induced in Wister rats by percutaneously puncturing the coccyx IVDs and human IVDs were collected from patients who underwent spinal surgery. Expression of the UPR target genes was elevated in degenerative IVDs in both humans and rats. The induction of ER stress in annulus fibrosus cells significantly increased the transcripts for tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) in a nuclear factor (NF)-κB pathway-dependent manner. The expression of TNF-α and IL-6 was significantly reduced by treatment with a selective PERK inhibitor, GSK2606414, and by gene silencing against PERK and activating transcription factor 4 (ATF4) transcripts. Our findings indicate that the UPR mediated by the PERK pathway is causally related to the development of IVD degeneration, suggesting that PERK may be a potential molecular target for suppressing the degenerative changes in IVDs. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1334-1345, 2018.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Degeneração do Disco Intervertebral/etiologia , Resposta a Proteínas não Dobradas , eIF-2 Quinase/fisiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Células Cultivadas , Pré-Escolar , Feminino , Humanos , Degeneração do Disco Intervertebral/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , NF-kappa B/fisiologia , Ratos , Ratos Wistar , Transdução de Sinais , eIF-2 Quinase/antagonistas & inibidores
15.
Cancer Lett ; 412: 46-58, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29037867

RESUMO

Colon cancer is one of the leading causes of cancer-related deaths. A natural sesquiterpene lactone, costunolide (CTD), showed inhibition of cancer development. However, the underlying mechanisms are not known. Here, we have examined the therapeutic activity and novel mechanisms of the anti-cancer activities of CTD in colon cancer cells. Using SPR analysis and enzyme activity assay on recombinant TrxR1 protein, our results show that CTD directly binds and inhibits the activity of TrxR1, which caused enhanced generation of ROS and led to ROS-dependent endoplasmic reticulum stress and cell apoptosis in colon cancer cells. Overexpression of TrxR1 in HCT116 cells reversed CTD-induced cell apoptosis and ROS increase. CTD treatment of mice implanted with colon cancer cells showed tumor growth inhibition and reduced TrxR1 activity and ROS level. In addition, it was observed that TrxR1 was significantly up-regulated in existing colon cancer gene database and clinically obtained colon cancer tissues. Our studies have uncovered the mechanism underlying the biological activity of CTD in colon cancer and suggest that targeting TrxR1 may prove to be beneficial as a treatment option.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Sesquiterpenos/farmacologia , Tiorredoxina Redutase 1/antagonistas & inibidores , Fator 4 Ativador da Transcrição/fisiologia , Animais , Antineoplásicos Fitogênicos/farmacologia , Neoplasias do Colo/patologia , Estresse do Retículo Endoplasmático , Feminino , Células HCT116 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Espécies Reativas de Oxigênio/metabolismo , Sesquiterpenos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Trends Endocrinol Metab ; 28(11): 794-806, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28797581

RESUMO

Activating transcription factor 4 (ATF4) is a stress-induced transcription factor that is frequently upregulated in cancer cells. ATF4 controls the expression of a wide range of adaptive genes that allow cells to endure periods of stress, such as hypoxia or amino acid limitation. However, under persistent stress conditions, ATF4 promotes the induction of apoptosis. Recent advances point to a role for post-translational modifications (PTMs) and epigenetic mechanisms in balancing these pro- and anti-survival effects of ATF4. We review here how PTMs and epigenetic modifiers associated with ATF4 may be exploited by cancer cells to cope with cellular stress conditions that are intrinsically associated with tumor growth. Identification of mechanisms that modulate ATF4-mediated transcription and its effects on cellular metabolism may uncover new targets for cancer treatment.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Adaptação Fisiológica , Apoptose/genética , Neoplasias/patologia , Estresse Fisiológico/fisiologia , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Adaptação Fisiológica/genética , Aminoácidos/deficiência , Animais , Sobrevivência Celular/genética , Humanos , Hipóxia/genética , Hipóxia/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Processamento de Proteína Pós-Traducional
17.
Exp Cell Res ; 360(2): 105-112, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28843961

RESUMO

Activating transcription factor 4 (ATF4), an endoplasmic reticulum stress-inducible transcription factor, plays important roles in cancer progression and resistance to therapy. However, no report is available about its roles in endometrial cancer (EC). In this study, we found that ATF4 is commonly expressed in EC cell lines. Loss-of-function studies in two EC cell lines showed that ATF4 knockdown suppresses tumor growth of EC in vivo without influencing cell proliferation in vitro. And xenograft tumors derived from ATF4-knockdown cells had reduced M2 macrophage infiltration. In clinical specimens, ATF4-high expressing tumors indeed contained more macrophage infiltration compared to those with lower ATF4 expression. Moreover, we identified that ATF4-mediated chemokine CCL2 expression ultimately results in macrophage infiltration and tumor growth of EC. Taken together, our findings suggest that ATF4 contributes to tumor growth of EC by promoting CCL2 and subsequent recruitment of macrophage, and that ATF4/CCL2 axis might be a potential therapeutic target for EC.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Carcinoma Endometrioide/genética , Proliferação de Células/genética , Quimiocina CCL2/genética , Quimiotaxia de Leucócito/genética , Neoplasias do Endométrio/genética , Macrófagos/fisiologia , Fator 4 Ativador da Transcrição/antagonistas & inibidores , Fator 4 Ativador da Transcrição/genética , Animais , Carcinoma Endometrioide/patologia , Células Cultivadas , Neoplasias do Endométrio/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Camundongos Nus , Evasão Tumoral/genética
18.
Am J Respir Cell Mol Biol ; 57(2): 193-203, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28363030

RESUMO

Ventilator-induced lung injury (VILI) is a severe complication of mechanical ventilation that can lead to acute respiratory distress syndrome. VILI is characterized by damage to the epithelial barrier with subsequent pulmonary edema and profound hypoxia. Available lung-protective ventilator strategies offer only a modest benefit in preventing VILI because they cannot impede alveolar overdistension and concomitant epithelial barrier dysfunction in the inflamed lung regions. There are currently no effective biochemical therapies to mitigate injury to the alveolar epithelium. We hypothesize that alveolar stretch activates the integrated stress response (ISR) pathway and that the chemical inhibition of this pathway mitigates alveolar barrier disruption during stretch and mechanical ventilation. Using our established rat primary type I-like alveolar epithelial cell monolayer stretch model and in vivo rat mechanical ventilation that mimics the alveolar overdistension seen in acute respiratory distress syndrome, we studied epithelial responses to mechanical stress. Our studies revealed that the ISR signaling pathway is a key modulator of epithelial permeability. We show that prolonged epithelial stretch and injurious mechanical ventilation activate the ISR, leading to increased alveolar permeability, cell death, and proinflammatory signaling. Chemical inhibition of protein kinase RNA-like endoplasmic reticulum kinase, an upstream regulator of the pathway, resulted in decreased injury signaling and improved barrier function after prolonged cyclic stretch and injurious mechanical ventilation. Our results provide new evidence that therapeutic targeting of the ISR can mitigate VILI.


Assuntos
Células Epiteliais Alveolares/patologia , Estresse Fisiológico/fisiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/fisiopatologia , Fator 4 Ativador da Transcrição/antagonistas & inibidores , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/fisiologia , Adenina/análogos & derivados , Adenina/farmacologia , Adenina/uso terapêutico , Células Epiteliais Alveolares/metabolismo , Animais , Morte Celular , Permeabilidade da Membrana Celular , Células Cultivadas , Citocinas/metabolismo , Avaliação Pré-Clínica de Medicamentos , Ativação Enzimática , Regulação da Expressão Gênica , Indóis/farmacologia , Indóis/uso terapêutico , Inibidores de Proteases/farmacologia , Inibidores de Proteases/uso terapêutico , Edema Pulmonar/tratamento farmacológico , Edema Pulmonar/etiologia , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Estresse Mecânico , Fator de Transcrição CHOP/antagonistas & inibidores , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/fisiologia , Resposta a Proteínas não Dobradas , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/genética , eIF-2 Quinase/fisiologia
19.
Curr Opin Clin Nutr Metab Care ; 20(3): 175-180, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28376508

RESUMO

PURPOSE OF REVIEW: The article highlights the recent development of an ATF4 (activating transcription factor) inducible luciferase (LUC) mouse model to monitor the integrated stress response pathway (ISR) in vivo. RECENT FINDING: The ISR pathway plays a key role in cellular adaptation to stress and is dysregulated in numerous diseases. The core event in this pathway is the phosphorylation of eukaryotic translation initiation factor 2 α, which leads to the recruitment of the transcription factor ATF4 to specific CCAAT/enhancer-binding protein-ATF response elements (CAREs) located in the promoters of target genes. To monitor the modulation of this pathway in the whole animal and at tissue and cellular levels, we generated a CARE-driven LUC mouse model. We validated the relevance of this model to study stress-related pathologies and recently observed the correlation between the ISR pathway induction in muscle and the occurrence of stress-induced skeletal muscle atrophy. SUMMARY: The CARE-LUC mouse model represents an innovative tool for investigating the role of the ISR pathway in physiology and disease and opens new avenues for the development of drugs that could modify this important pathway in stress-related human diseases.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Luciferases/fisiologia , Modelos Animais , Transdução de Sinais/fisiologia , Estresse Fisiológico/fisiologia , Animais , Camundongos , Músculo Esquelético/metabolismo , Atrofia Muscular/fisiopatologia , Fosforilação/fisiologia , Regiões Promotoras Genéticas/fisiologia , Proteínas Serina-Treonina Quinases
20.
Cancer Res ; 76(21): 6193-6204, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27651314

RESUMO

Tryptophan degradation is an immune escape strategy shared by many tumors. However, cancer cells' compensatory mechanisms remain unclear. We demonstrate here that a shortage of tryptophan caused by expression of indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) resulted in ATF4-dependent upregulation of several amino acid transporters, including SLC1A5 and its truncated isoforms, which in turn enhanced tryptophan and glutamine uptake. Importantly, SLC1A5 failed to be upregulated in resting human T cells kept under low tryptophan conditions but was enhanced upon cognate antigen T-cell receptor engagement. Our results highlight key differences in the ability of tumor and T cells to adapt to tryptophan starvation and provide important insights into the poor prognosis of tumors coexpressing IDO and SLC1A5. Cancer Res; 76(21); 6193-204. ©2016 AACR.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Sistema ASC de Transporte de Aminoácidos/fisiologia , Sistemas de Transporte de Aminoácidos/genética , Reprogramação Celular , Indolamina-Pirrol 2,3,-Dioxigenase/fisiologia , Antígenos de Histocompatibilidade Menor/fisiologia , Neoplasias/metabolismo , Triptofano/metabolismo , Linhagem Celular Tumoral , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA