Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Blood ; 139(26): 3752-3770, 2022 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-35439288

RESUMO

Differentiation blockade is a hallmark of acute myeloid leukemia (AML). A strategy to overcome such a blockade is a promising approach against the disease. The lack of understanding of the underlying mechanisms hampers development of such strategies. Dysregulated ribonucleotide reductase (RNR) is considered a druggable target in proliferative cancers susceptible to deoxynucleoside triphosphate (dNTP) depletion. Herein, we report an unanticipated discovery that hyperactivating RNR enables differentiation and decreases leukemia cell growth. We integrate pharmacogenomics and metabolomics analyses to identify that pharmacologically (eg, nelarabine) or genetically upregulating RNR subunit M2 (RRM2) creates a dNTP pool imbalance and overcomes differentiation arrest. Moreover, R-loop-mediated DNA replication stress signaling is responsible for RRM2 activation by nelarabine treatment. Further aggravating dNTP imbalance by depleting the dNTP hydrolase SAM domain and HD domain-containing protein 1 (SAMHD1) enhances ablation of leukemia stem cells by RRM2 hyperactivation. Mechanistically, excessive activation of extracellular signal-regulated kinase (ERK) signaling downstream of the imbalance contributes to cellular outcomes of RNR hyperactivation. A CRISPR screen identifies a synthetic lethal interaction between loss of DUSP6, an ERK-negative regulator, and nelarabine treatment. These data demonstrate that dNTP homeostasis governs leukemia maintenance, and a combination of DUSP inhibition and nelarabine represents a therapeutic strategy.


Assuntos
Leucemia Mieloide Aguda , Ribonucleotídeo Redutases , Replicação do DNA , Homeostase , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Polifosfatos , Ribonucleotídeo Redutases/genética , Ribonucleotídeo Redutases/metabolismo
2.
J Pineal Res ; 76(1): e12918, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37814536

RESUMO

Endometrial cancer (EC) is a reproductive system disease that occurs in perimenopausal and postmenopausal women. However, its etiology is unclear. Melatonin (MT) has been identified as a therapeutic agent for EC; however, its exact mechanism remains unclear. In the present study, we determined that GATA-binding protein 2 (GATA2) is expressed at low levels in EC and regulated by MT. MT upregulates the expression of GATA2 through MT receptor 1A (MTNR1A), whereas GATA2 can promote the expression of MTNR1A by binding to its promoter region. In addition, in vivo and in vitro experiments showed that MT inhibited the proliferation and metastasis of EC cells by upregulating GATA2 expression. The protein kinase B (AKT) pathway was also affected. In conclusion, these findings suggest that MT and GATA2 play significant roles in EC development.


Assuntos
Neoplasias do Endométrio , Melatonina , Humanos , Feminino , Melatonina/farmacologia , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Proliferação de Células , Linhagem Celular Tumoral
3.
Environ Sci Technol ; 58(13): 6049-6057, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38525996

RESUMO

High Cl- concentration in saline wastewater (e.g., landfill leachate) limits wastewater purification. Catalytic Cl- conversion into reactive chlorine species (RCS) arises as a sustainable strategy, making the salinity profitable for efficient wastewater treatment. Herein, aiming to reveal the structure-property relationship in Cl- utilization, bismuth oxychloride (BiOCl) photocatalysts with coexposed {001} and {110} facets are synthesized. With an increasing {001} ratio, the RCS production efficiency increases from 75.64 to 96.89 µg L-1 min-1. Mechanism investigation demonstrates the fast release of lattice Cl- as an RCS and the compensation of ambient Cl-. Correlation analysis between the internal electric field (IEF, parallel to [001]) and normalized efficiency on {110} (kRCS/S{110}, perpendicular to [001]) displays a coefficient of 0.86, validating that the promoted carrier dynamics eventually affects Cl- conversion on the open layered structure. The BiOCl photocatalyst is well behaved in ammonium (NH4+-N) degradation ranging from 20 to 800 mg N L-1 with different chlorinity (3-12 g L-1 NaCl). The sustainable Cl- conversion into RCS also realizes 85.4% of NH4+-N removal in the treatment of realistic landfill leachate (662 mg of N L-1 NH4+-N). The structure-property relationship provides insights into the design of efficient catalysts for environment remediation using ambient Cl-.


Assuntos
Compostos de Amônio , Bismuto , Poluentes Químicos da Água , Águas Residuárias , Poluentes Químicos da Água/química , Salinidade
4.
Angew Chem Int Ed Engl ; 63(23): e202403585, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38565432

RESUMO

In spite of the competitive performance at room temperature, the development of sodium-ion batteries (SIBs) is still hindered by sluggish electrochemical reaction kinetics and unstable electrode/electrolyte interphase under subzero environments. Herein, a low-concentration electrolyte, consisting of 0.5M NaPF6 dissolving in diethylene glycol dimethyl ether solvent, is proposed for SIBs working at low temperature. Such an electrolyte generates a thin, amorphous, and homogeneous cathode/electrolyte interphase at low temperature. The interphase is monolithic and rich in organic components, reducing the limitation of Na+ migration through inorganic crystals, thereby facilitating the interfacial Na+ dynamics at low temperature. Furthermore, it effectively blocks the unfavorable side reactions between active materials and electrolytes, improving the structural stability. Consequently, Na0.7Li0.03Mg0.03Ni0.27Mn0.6Ti0.07O2//Na and hard carbon//Na cells deliver a high capacity retention of 90.8 % after 900 cycles at 1C, a capacity over 310 mAh g-1 under -30 °C, respectively, showing long-term cycling stability and great rate capability at low temperature.

5.
Blood ; 134(15): 1257-1268, 2019 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-31395602

RESUMO

Relapse remains the main cause of MLL-rearranged (MLL-r) acute lymphoblastic leukemia (ALL) treatment failure resulting from persistence of drug-resistant clones after conventional chemotherapy treatment or targeted therapy. Thus, defining mechanisms underlying MLL-r ALL maintenance is critical for developing effective therapy. PRMT1, which deposits an asymmetric dimethylarginine mark on histone/non-histone proteins, is reportedly overexpressed in various cancers. Here, we demonstrate elevated PRMT1 levels in MLL-r ALL cells and show that inhibition of PRMT1 significantly suppresses leukemic cell growth and survival. Mechanistically, we reveal that PRMT1 methylates Fms-like receptor tyrosine kinase 3 (FLT3) at arginine (R) residues 972 and 973 (R972/973), and its oncogenic function in MLL-r ALL cells is FLT3 methylation dependent. Both biochemistry and computational analysis demonstrate that R972/973 methylation could facilitate recruitment of adaptor proteins to FLT3 in a phospho-tyrosine (Y) residue 969 (Y969) dependent or independent manner. Cells expressing R972/973 methylation-deficient FLT3 exhibited more robust apoptosis and growth inhibition than did Y969 phosphorylation-deficient FLT3-transduced cells. We also show that the capacity of the type I PRMT inhibitor MS023 to inhibit leukemia cell viability parallels baseline FLT3 R972/973 methylation levels. Finally, combining FLT3 tyrosine kinase inhibitor PKC412 with MS023 treatment enhanced elimination of MLL-r ALL cells relative to PKC412 treatment alone in patient-derived mouse xenografts. These results indicate that abolishing FLT3 arginine methylation through PRMT1 inhibition represents a promising strategy to target MLL-r ALL cells.


Assuntos
Histona-Lisina N-Metiltransferase/genética , Proteína de Leucina Linfoide-Mieloide/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo , Animais , Apoptose , Proliferação de Células , Sobrevivência Celular , Rearranjo Gênico , Humanos , Camundongos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Células Tumorais Cultivadas
6.
Blood ; 134(6): 548-560, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31217189

RESUMO

The presence of FMS-like receptor tyrosine kinase-3 internal tandem duplication (FLT3-ITD) mutations in patients with acute myeloid leukemia (AML) is associated with poor clinical outcome. FLT3 tyrosine kinase inhibitors (TKIs), although effective in kinase ablation, do not eliminate primitive FLT3-ITD+ leukemia cells, which are potential sources of relapse. Thus, understanding the mechanisms underlying FLT3-ITD+ AML cell persistence is essential to devise future AML therapies. Here, we show that expression of protein arginine methyltransferase 1 (PRMT1), the primary type I arginine methyltransferase, is increased significantly in AML cells relative to normal hematopoietic cells. Genome-wide analysis, coimmunoprecipitation assay, and PRMT1-knockout mouse studies indicate that PRMT1 preferentially cooperates with FLT3-ITD, contributing to AML maintenance. Genetic or pharmacological inhibition of PRMT1 markedly blocked FLT3-ITD+ AML cell maintenance. Mechanistically, PRMT1 catalyzed FLT3-ITD protein methylation at arginine 972/973, and PRMT1 promoted leukemia cell growth in an FLT3 methylation-dependent manner. Moreover, the effects of FLT3-ITD methylation in AML cells were partially due to cross talk with FLT3-ITD phosphorylation at tyrosine 969. Importantly, FLT3 methylation persisted in FLT3-ITD+ AML cells following kinase inhibition, indicating that methylation occurs independently of kinase activity. Finally, in patient-derived xenograft and murine AML models, combined administration of AC220 with a type I PRMT inhibitor (MS023) enhanced elimination of FLT3-ITD+ AML cells relative to AC220 treatment alone. Our study demonstrates that PRMT1-mediated FLT3 methylation promotes AML maintenance and suggests that combining PRMT1 inhibition with FLT3 TKI treatment could be a promising approach to eliminate FLT3-ITD+ AML cells.


Assuntos
Arginina/metabolismo , Duplicação Gênica , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo , Animais , Biomarcadores Tumorais , Catálise , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Leucemia Mieloide Aguda/mortalidade , Leucemia Mieloide Aguda/patologia , Metilação , Camundongos , Camundongos Knockout , Modelos Moleculares , Prognóstico , Ligação Proteica , Conformação Proteica , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Proteína-Arginina N-Metiltransferases/química , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/química , Ensaios Antitumorais Modelo de Xenoenxerto , Tirosina Quinase 3 Semelhante a fms/química
7.
Bioorg Med Chem ; 28(7): 115372, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32088124

RESUMO

Histone-lysine N-methyltransferase SET7 emerged as a potential target for multiple cancers. In a virtual screening program used to explore new and potent inhibitors of SET7, compound 16 was discovered as a top hit with an IC50 value of 6.02 µM. A further similarity search afforded a new compound 23, which exhibited better activity against SET7 with an IC50 value of 1.96 µM. Importantly, compound 23 selectively inhibited the proliferation of MV4-11 cells. Comprehensively, compound 23 can serve as a lead for further identification and development of more potent SET7 inhibitors.


Assuntos
Antineoplásicos/farmacologia , Descoberta de Drogas , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Antineoplásicos/química , Domínio Catalítico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Química Computacional , Simulação por Computador , Escherichia coli/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Estrutura Molecular , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
8.
J Environ Manage ; 261: 110226, 2020 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-32148296

RESUMO

Dye decolorization is of crucial concern for effectively treating dye wastewater. In this study, rapid and effective decolorization of malachite green cationic dye was achieved by tea polyphenols and ferric ions under moderate conditions. Approximately 96.2% of decolorization efficiency could be obtained within the first 10 min at the initial dye concentration of 50 mg/L. The proposed method can perform excellently in a wide pH range of 5-9 and decolorization kinetics of malachite green under different solution pH were well fitted by the pseudo-second-order model. After the decolorization, only a slight reduction of tea polyphenols was observed, while the strength of peaks assigned to nitrogen-containing groups was significantly weakened, indicating that the N-demethylation reaction might occur during the decolorization process. The nucleophilic attack of deprotonated hydroxyl groups of tea polyphenols was proposed as the decolorization mechanism. The presence of ferric ions at an appropriate dosage could promote the deprotonation process and therefore enhance decolorization efficiency, while excess ferric ions in solution might compete with malachite green dye towards reductive sites on tea polyphenols. The findings from this study provided an economical and environmentally friendly technique for the effective decolorization of dye wastewater.


Assuntos
Polifenóis , Corantes de Rosanilina , Corantes , Concentração de Íons de Hidrogênio , Íons , Chá
9.
Angiogenesis ; 22(2): 251-262, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30465119

RESUMO

The miR-200 family, consisting of miR-200a/b/c, miR-141, and miR-429, is well known to inhibit epithelial-to-mesenchymal transition (EMT) in cancer invasion and metastasis. Among the miR-200 family members, miR-200a/b/c and miR-429 have been reported to inhibit angiogenesis. However, the role of miR-141 in angiogenesis remains elusive, as contradicting results have been found in different cancer types and tumor models. Particularly, the effect of miR-141 in vascular endothelial cells has not been defined. In this study, we used several in vitro and in vivo models to demonstrate that miR-141 in endothelial cells inhibits angiogenesis. Additional mechanistic studies showed that miR-141 suppresses angiogenesis through multiple targets, including NRP1, GAB1, CXCL12ß, TGFß2, and GATA6, and bioinformatics analysis indicated that miR-141 and its targets comprise a powerful and precise regulatory network to modulate angiogenesis. Taken together, these data not only demonstrate an anti-angiogenic effect of miR-141, further strengthening the critical role of miR-200 family in the process of angiogenesis, but also provides a valuable cancer therapeutic target to control both angiogenesis and EMT, two essential steps in tumor growth and metastasis.


Assuntos
Redes Reguladoras de Genes/fisiologia , MicroRNAs/fisiologia , Neovascularização Fisiológica/genética , Animais , Células Cultivadas , Regulação para Baixo/genética , Transição Epitelial-Mesenquimal/genética , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes/genética , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética
10.
Nucleic Acids Res ; 42(16): 10720-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25159612

RESUMO

Follistatin (FST) performs several vital functions in the cells, including protection from apoptosis during stress. The expression of FST is up-regulated in response to glucose deprivation by an unknown mechanism. We herein showed that the induction of FST by glucose deprivation was due to an increase in the half-life of its mRNA. We further identified an AU-rich element (ARE) in the 3'UTR of FST mRNA that mediated its decay. The expression of FST was elevated after knocking down AUF1 and reduced when AUF1 was further expressed. In vitro binding assays and RNA pull-down assays revealed that AUF1 interacted with FST mRNA directly via its ARE. During glucose deprivation, a majority of AUF1 shuttled from cytoplasm to nucleus, resulting in dissociation of AUF1 from FST mRNA and thus stabilization of FST mRNA. Finally, knockdown of AUF1 decreased whereas overexpression of AUF1 increased glucose deprivation-induced apoptosis. The apoptosis promoting effect of AUF1 was eliminated in FST expressing cells. Collectively, this study provided evidence that AUF1 is a negative regulator of FST expression and participates in the regulation of cell survival under glucose deprivation.


Assuntos
Apoptose , Folistatina/genética , Glucose/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/metabolismo , Estabilidade de RNA , RNA Mensageiro/metabolismo , Regiões 3' não Traduzidas , Proteínas Quinases Ativadas por AMP/metabolismo , Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , Folistatina/metabolismo , Células HeLa , Ribonucleoproteína Nuclear Heterogênea D0 , Humanos
11.
Int J Cancer ; 137(10): 2310-22, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25991585

RESUMO

Colorectal cancer (CRC) is one of the most common cancers worldwide and its metastasis accounts for the majority of deaths. However, the molecular mechanisms underlying CRC progression are not well characterized. In this study, we identified miR-409-3p as a tumor suppressor of CRC. MiR-409-3p expression was significantly downregulated in CRC tissue compared to adjacent non-tumor tissue, and reduced miR-409-3p expression was correlated with CRC metastasis. In vitro and in vivo studies revealed that miR-409-3p negatively regulated CRC metastatic capacities, including suppressing cancer cell migration, invasion and metastasis. To explore the mechanism of action of miR-409-3p, we adopted a pathway and pathophysiological event-based target screening and validation approach, and found nine known metastasis-related genes as potential targets. The 3'-UTR binding assays between the candidates and miR-409-3p suggested that only GAB1, NR4A2 and LMO4 were directly regulated by the miRNA. However, endogenous expression analysis revealed that only GAB1 was modulated by miR-409-3p in CRC cells at both the mRNA and protein levels. Furthermore, we provided evidence to conclude that GAB1 was partially responsible for miR-409-3p-mediated metastasis. Taken together, our data demonstrate that miR-409-3p is a metastatic suppressor, and post-transcriptional inhibition of the oncoprotein GAB1 is one of the mechanisms of action of this miRNA. Our finding suggests miR-409-3p might be a novel target for CRC metastasis treatment.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias Colorretais/patologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , MicroRNAs/genética , Regiões 3' não Traduzidas , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/genética , Feminino , Células HCT116 , Humanos , Neoplasias Pulmonares/genética , Camundongos , Transplante de Neoplasias
13.
Mol Metab ; 84: 101953, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38710444

RESUMO

OBJECTIVE: Lipid metabolism plays an important role in early pregnancy, but its effects on decidualization are poorly understood. Fatty acids (FAs) must be esterified by fatty acyl-CoA synthetases to form biologically active acyl-CoA in order to enter the anabolic and/or catabolic pathway. Long-chain acyl-CoA synthetase 4 (ACSL4) is associated with female reproduction. However, whether it is involved in decidualization is unknown. METHODS: The expression of ACSL4 in human and mouse endometrium was detected by immunohistochemistry. ACSL4 levels were regulated by the overexpression of ACSL4 plasmid or ACSL4 siRNA, and the effects of ACSL4 on decidualization markers and morphology of endometrial stromal cells (ESCs) were clarified. A pregnant mouse model was established to determine the effect of ACSL4 on the implantation efficiency of mouse embryos. Modulation of ACSL4 detects lipid anabolism and catabolism. RESULTS: Through examining the expression level of ACSL4 in human endometrial tissues during proliferative and secretory phases, we found that ACSL4 was highly expressed during the secretory phase. Knockdown of ACSL4 suppressed decidualization and inhibited the mesenchymal-to-epithelial transition induced by MPA and db-cAMP in ESCs. Further, the knockdown of ACSL4 reduced the efficiency of embryo implantation in pregnant mice. Downregulation of ACSL4 inhibited FA ß-oxidation and lipid droplet accumulation during decidualization. Interestingly, pharmacological and genetic inhibition of lipid droplet synthesis did not affect FA ß-oxidation and decidualization, while the pharmacological and genetic inhibition of FA ß-oxidation increased lipid droplet accumulation and inhibited decidualization. In addition, inhibition of ß-oxidation was found to attenuate the promotion of decidualization by the upregulation of ACSL4. The decidualization damage caused by ACSL4 knockdown could be reversed by activating ß-oxidation. CONCLUSIONS: Our findings suggest that ACSL4 promotes endometrial decidualization by activating the ß-oxidation pathway. This study provides interesting insights into our understanding of the mechanisms regulating lipid metabolism during decidualization.


Assuntos
Coenzima A Ligases , Endométrio , Ácidos Graxos , Gotículas Lipídicas , Oxirredução , Feminino , Coenzima A Ligases/metabolismo , Coenzima A Ligases/genética , Animais , Camundongos , Humanos , Endométrio/metabolismo , Ácidos Graxos/metabolismo , Gravidez , Gotículas Lipídicas/metabolismo , Decídua/metabolismo , Adulto , Metabolismo dos Lipídeos , Implantação do Embrião , Células Estromais/metabolismo
14.
Nat Cancer ; 5(4): 601-624, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38413714

RESUMO

Current anticancer therapies cannot eliminate all cancer cells, which hijack normal arginine methylation as a means to promote their maintenance via unknown mechanisms. Here we show that targeting protein arginine N-methyltransferase 9 (PRMT9), whose activities are elevated in blasts and leukemia stem cells (LSCs) from patients with acute myeloid leukemia (AML), eliminates disease via cancer-intrinsic mechanisms and cancer-extrinsic type I interferon (IFN)-associated immunity. PRMT9 ablation in AML cells decreased the arginine methylation of regulators of RNA translation and the DNA damage response, suppressing cell survival. Notably, PRMT9 inhibition promoted DNA damage and activated cyclic GMP-AMP synthase, which underlies the type I IFN response. Genetically activating cyclic GMP-AMP synthase in AML cells blocked leukemogenesis. We also report synergy of a PRMT9 inhibitor with anti-programmed cell death protein 1 in eradicating AML. Overall, we conclude that PRMT9 functions in survival and immune evasion of both LSCs and non-LSCs; targeting PRMT9 may represent a potential anticancer strategy.


Assuntos
Arginina , Leucemia Mieloide Aguda , Células-Tronco Neoplásicas , Nucleotidiltransferases , Proteína-Arginina N-Metiltransferases , Proteína-Arginina N-Metiltransferases/metabolismo , Humanos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/metabolismo , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/tratamento farmacológico , Nucleotidiltransferases/metabolismo , Arginina/metabolismo , Metilação/efeitos dos fármacos , Animais , Camundongos , Interferon Tipo I/metabolismo , Dano ao DNA , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos
15.
Cell Physiol Biochem ; 32(6): 1695-706, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24356419

RESUMO

BACKGROUND: Angiogenin (ANG) can translocate to the target cell nucleus and accumulate in the nucleolus to enhance rRNA transcription, thus promoting cell proliferation. However, the regulation of ANG-enhanced rRNA transcription remains unknown. Previously we identified phospholipid scramblase 1 (PLSCR1) as a potential ANG-interacting protein in yeast two-hybrid screening. METHODS: The interaction was re-confirmed in yeast cells and further verified by in vitro pull down, in vivo co-immunoprecipitation (Co-IP), fluorescent resonance energy transfer (FRET) and immunofluorescence analyses. The rRNA transcription level was determined by real-time quantitative PCR and Northern blot. RESULTS: PLSCR1 was identified as a novel ANG-interacting protein. Notably, PLSCR1 interacted with ANG in the cell nucleus and regulated rRNA transcription. Furthermore, depletion of cellular ANG expression abolished PLSCR1-enhanced rRNA transcription, which could be rescued by exogenous ANG. CONCLUSION: Our data suggest that PLSCR1 positively regulates rRNA transcription through interacting with ANG, thus deepening our understanding on rRNA transcription regulation.


Assuntos
Proteínas de Transferência de Fosfolipídeos/metabolismo , RNA Ribossômico/metabolismo , Ribonuclease Pancreático/metabolismo , Núcleo Celular/metabolismo , Células HeLa , Humanos , Imunoprecipitação , Proteínas de Transferência de Fosfolipídeos/genética , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , RNA Ribossômico/genética , RNA Interferente Pequeno/metabolismo , Ribonuclease Pancreático/antagonistas & inibidores , Ribonuclease Pancreático/genética , Transcrição Gênica , Técnicas do Sistema de Duplo-Híbrido
16.
Front Mol Biosci ; 10: 1203269, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37251080

RESUMO

In 2012, researchers proposed a non-apoptotic, iron-dependent form of cell death caused by lipid peroxidation called ferroptosis. During the past decade, a comprehensive understanding of ferroptosis has emerged. Ferroptosis is closely associated with the tumor microenvironment, cancer, immunity, aging, and tissue damage. Its mechanism is precisely regulated at the epigenetic, transcriptional, and post-translational levels. O-GlcNAc modification (O-GlcNAcylation) is one of the post-translational modifications of proteins. Cells can modulate cell survival in response to stress stimuli, including apoptosis, necrosis, and autophagy, through adaptive regulation by O-GlcNAcylation. However, the function and mechanism of these modifications in regulating ferroptosis are only beginning to be understood. Here, we review the relevant literature within the last 5 years and present the current understanding of the regulatory function of O-GlcNAcylation in ferroptosis and the potential mechanisms that may be involved, including antioxidant defense system-controlled reactive oxygen species biology, iron metabolism, and membrane lipid peroxidation metabolism. In addition to these three areas of ferroptosis research, we examine how changes in the morphology and function of subcellular organelles (e.g., mitochondria and endoplasmic reticulum) involved in O-GlcNAcylation may trigger and amplify ferroptosis. We have dissected the role of O-GlcNAcylation in regulating ferroptosis and hope that our introduction will provide a general framework for those interested in this field.

17.
J Med Chem ; 66(10): 6849-6868, 2023 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-37141440

RESUMO

Osimertinib resistance is an unmet clinical need for the treatment of non-small cell lung cancer (NSCLC), and the main mechanism is tertiary C797S mutation of epidermal growth factor receptor (EGFR). To date, there is no inhibitor approved for the treatment of Osimertinib-resistant NSCLC. Herein, we reported a series of Osimertinib derivatives as fourth-generation inhibitors which were rationally designed. Top candidate D51 potently inhibited the EGFRL858R/T790M/C797S mutant with an IC50 value of 14 nM and suppressed the proliferation of H1975-TM cells with an IC50 value of 14 nM, which show over 500-fold selectivity against wild-type forms. Moreover, D51 inhibited the EGFRdel19/T790M/C797S mutant and the proliferation of the PC9-TM cell line with IC50 values of 62 and 82 nM. D51 also exhibited favorable in vivo druggability, including PK parameters, safety properties, in vivo stability, and antitumor activity.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , Receptores ErbB/metabolismo , Mutação , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Compostos de Anilina/farmacologia , Compostos de Anilina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Linhagem Celular Tumoral
18.
ACS Appl Mater Interfaces ; 15(46): 53623-53631, 2023 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-37955137

RESUMO

Fierce phase transformation and limited sodium ion diffusion dynamics are critical obstacles that hinder the practical energy storage applications of P2-type layered sodium transition metal oxides (NaxTMO2). Herein, a synergistic strategy of electronic state tailoring and pillar effect was carefully implemented by substituting divalent Mg2+ into Na0.67Ni0.33Mn0.67O2 material with unique oriented hollow rodlike structures. Mg2+substitution can not only facilitate the anionic oxygen redox reactions and electronic conductivity through increasing the electronic states at Femi energy but also act as pillars within TMO2 layers to alleviate the severe phase transformation to improve structure stability. Moreover, the oriented hollow structure incorporating sufficient buffer spaces and rationally exposed electrochemically active facets effectively alleviates the stresses induced by low volume changes of 8% and provides more open channels for Na+ ion diffusion without crossing multiple grain boundaries. Hence, the Na0.67Mg0.08Ni0.25Mn0.67O2 cathode showed a superior rate capability with high energy density and cycling stability for sodium-ion storage. The underlying mechanisms of these achievements were deciphered through diversified dynamic analysis and the first principle calculations, providing new insights into P2-type NaxTMO2 cathodes for the infinite prospect as an alternative to lithium-ion batteries.

19.
J Med Chem ; 66(21): 14633-14652, 2023 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-37885208

RESUMO

Epidermal growth factor receptor (EGFR)-activating mutation is an important oncogenic driver of nonsmall cell lung cancer (NSCLC) patients. Osimertinib has been the first-line treatment for EGFR-mutated NSCLC. However, the tertiary C797S mutation leads to Osimertinib resistance by blocking the covalent binding of Cys797 to Osimertinib. To date, there are no approved inhibitors for the treatment of Osimertinib resistance. Herein, we identified a novel lead compound S8 targeting EGFRL858R/T790M/C797S by structure-based virtual screening and synthesized a series of novel compounds. Representative compound C34 showed potent inhibitory activity against EGFRL858R/T790M/C797S with an IC50 of 5.1 nM and significantly inhibited the proliferation of the H1975-TM cell line harboring EGFRL858R/T790M/C797S with an IC50 of 0.05 µM. Additionally, compound C34 demonstrated good pharmacokinetic properties with an oral bioavailability of 30.72% and significantly inhibited tumor growth in the H1975-TM xenograft tumor model. This study provides a novel thiazole derivative as an EGFR inhibitor to overcome C797S-mediated resistance.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/metabolismo , Receptores ErbB/metabolismo , Mutação , Resistencia a Medicamentos Antineoplásicos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Inibidores de Proteínas Quinases/química , Compostos de Anilina/farmacologia , Linhagem Celular Tumoral
20.
Front Mol Biosci ; 9: 837971, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35463945

RESUMO

Angiogenin (ANG) is the first human tumor-derived angiogenic protein, which can promote angiogenesis and tumor growth. In a previous study, we identified alpha-actinin 2 (ACTN2), a cytoskeletal protein, as a direct interacting protein with angiogenin. However, the interaction between ANG and ACTN2 was not characterized in detail, which may provide information on the molecular mechanisms of ANG functions. In this study, we mapped the accurate binding domain and sites in ANG and ACTN2, respectively. In ANG, the residues from 83 to 105 are the smallest motif that can bind to ACTN2. We then use site mutation analysis to identify the precise binding sites of ANG in the interaction and found that the 101st residue arginine (R101) represents the critical residue involved in the ANG-ACTN2 interaction. In ACTN2, the residues from 383 to 632, containing two spectrin domains in the middle of the rod structure of ACTN2, play an important role in the interaction. Furthermore, we validated the interaction of ACTN2-383-632 to ANG by glutathione-S-transferase (GST) pull-down assay. In functional analysis, overexpressed ACTN2-383-632 could impair tumor cell motility observably, including cell migration and invasion. Meanwhile, ACTN2-383-632 overexpression inhibited tumor cell proliferation and survival as well. These data suggest that an excess expression of ACTN2 segment ACTN2-383-632 can inhibit tumor cell motility and proliferation by interfering with the interaction between ANG and ACTN2, which provides a potential mechanism of ANG action in tumor growth and metastasis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA