Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Transl Med ; 22(1): 193, 2024 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-38388430

RESUMO

Aberrant upregulation of the ubiquitin-specific protease 14 (USP14) has been found in some malignant tumors, including oral squamous cell carcinoma (OSCC). In this study, we further demonstrated that aberrantly overexpressed USP14 was also closely related to adverse clinicopathological features and poor prognosis in patients with OSCC, so we hypothesized that USP14 might act as a tumor-promoting factor during the progression of OSCC. Notably, we originally proved that USP14 is a deubiquitinating enzyme for phosphofructokinase-1 liver type (PFKL), a key rate-limiting enzyme involved in the glycolytic pathway. USP14 interacts with PFKL and enhances its stability through deubiquitination in OSCC cells, which in turn enhances PFKL-mediated glycolytic metabolism and ultimately promote cellular proliferation, migration, and tumorigenesis. In this work, we have also demonstrated for the first time that USP14 is a critical regulator of glycolysis in OSCC and verified a novel mechanism whereby it is involved in tumor metastasis and growth. Collectively, our findings provide novel insights into the tumor-promoting role of USP14 and establish mechanistic foundations for USP14-targeting therapies.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço , Neoplasias Bucais/genética , Fosfofrutoquinase-1 , Fígado , Glicólise , Proliferação de Células , Proteases Específicas de Ubiquitina , Linhagem Celular Tumoral , Ubiquitina Tiolesterase
2.
Acta Biochim Biophys Sin (Shanghai) ; 54(8): 1133-1139, 2022 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-35866602

RESUMO

The coronavirus papain-like protease (PLpro) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for viral polypeptide cleavage and the deISGylation of interferon-stimulated gene 15 (ISG15), which enable it to participate in virus replication and host innate immune pathways. Therefore, PLpro is considered an attractive antiviral drug target. Here, we show that parthenolide, a germacrane sesquiterpene lactone, has SARS-CoV-2 PLpro inhibitory activity. Parthenolide covalently binds to Cys-191 or Cys-194 of the PLpro protein, but not the Cys-111 at the PLpro catalytic site. Mutation of Cys-191 or Cys-194 reduces the activity of PLpro. Molecular docking studies show that parthenolide may also form hydrogen bonds with Lys-192, Thr-193, and Gln-231. Furthermore, parthenolide inhibits the deISGylation but not the deubiquitinating activity of PLpro in vitro. These results reveal that parthenolide inhibits PLpro activity by allosteric regulation.


Assuntos
Tratamento Farmacológico da COVID-19 , Proteases Semelhantes à Papaína de Coronavírus , Antivirais/farmacologia , Humanos , Interferons , Lactonas , Simulação de Acoplamento Molecular , Papaína/química , Papaína/metabolismo , Peptídeo Hidrolases/metabolismo , SARS-CoV-2 , Sesquiterpenos , Sesquiterpenos de Germacrano , Ubiquitina/metabolismo
3.
Immunity ; 37(1): 74-84, 2012 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-22727489

RESUMO

Epithelial keratinocyte proliferation is an essential element of wound repair, and abnormal epithelial proliferation is an intrinsic element in the skin disorder psoriasis. The factors that trigger epithelial proliferation in these inflammatory processes are incompletely understood. Here we have shown that regenerating islet-derived protein 3-alpha (REG3A) is highly expressed in keratinocytes during psoriasis and wound repair and in imiquimod-induced psoriatic skin lesions. The expression of REG3A by keratinocytes is induced by interleukin-17 (IL-17) via activation of keratinocyte-encoded IL-17 receptor A (IL-17RA) and feeds back on keratinocytes to inhibit terminal differentiation and increase cell proliferation by binding to exostosin-like 3 (EXTL3) followed by activation of phosphatidylinositol 3 kinase (PI3K) and the kinase AKT. These findings reveal that REG3A, a secreted intestinal antimicrobial protein, can promote skin keratinocyte proliferation and can be induced by IL-17. This observation suggests that REG3A may mediate the epidermal hyperproliferation observed in normal wound repair and in psoriasis.


Assuntos
Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/metabolismo , Diferenciação Celular , Queratinócitos/citologia , Queratinócitos/metabolismo , Lectinas Tipo C/metabolismo , Pele/lesões , Pele/metabolismo , Animais , Antígenos de Neoplasias/genética , Biomarcadores Tumorais/genética , Diferenciação Celular/genética , Proliferação de Células , Epiderme/efeitos dos fármacos , Epiderme/lesões , Epiderme/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-17/farmacologia , Queratinócitos/efeitos dos fármacos , Lectinas Tipo C/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , N-Acetilglucosaminiltransferases/metabolismo , Proteínas Associadas a Pancreatite , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Psoríase/imunologia , Psoríase/metabolismo , Psoríase/patologia , Transdução de Sinais , Pele/efeitos dos fármacos , Cicatrização/genética
4.
Acta Pharmacol Sin ; 42(4): 604-612, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32694757

RESUMO

The kinase FLT3 internal tandem duplication (FLT3-ITD) is related to poor clinical outcomes of acute myeloid leukemia (AML). FLT3 inhibitors have provided novel strategies for the treatment of FLT3-ITD-positive AML. But they are limited by rapid development of acquired resistance and refractory in monotherapy. Recent evidence shows that inducing the degradation of FLT3-mutated protein is an attractive strategy for the treatment of FLT3-ITD-positive AML, especially those with FLT3 inhibitor resistance. In this study we identified Wu-5 as a novel USP10 inhibitor inducing the degradation of FLT3-mutated protein. We showed that Wu-5 selectively inhibited the viability of FLT3 inhibitor-sensitive (MV4-11, Molm13) and -resistant (MV4-11R) FLT3-ITD-positive AML cells with IC50 of 3.794, 5.056, and 8.386 µM, respectively. Wu-5 (1-10 µM) dose-dependently induced apoptosis of MV4-11, Molm13, and MV4-11R cells through the proteasome-mediated degradation of FLT3-ITD. We further demonstrated that Wu-5 directly interacted with and inactivated USP10, the deubiquitinase for FLT3-ITD in vitro (IC50 value = 8.3 µM) and in FLT3-ITD-positive AML cells. Overexpression of USP10 abrogated Wu-5-induced FLT3-ITD degradation and cell death. Also, the combined treatment of Wu-5 and crenolanib produced synergistic cell death in FLT3-ITD-positive cells via the reduction of both FLT3 and AMPKα proteins. In support of this, AMPKα inhibitor compound C synergistically enhanced the anti-leukemia effect of crenolanib, while AMPKα activator metformin inhibited the anti-leukemia effect of crenolanib. In summary, we demonstrate that Wu-5, a novel USP10 inhibitor, can overcome FLT3 inhibitor resistance and synergistically enhance the anti-AML effect of crenolanib through targeting FLT3 and AMPKα pathway.


Assuntos
Apoptose/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tiofenos/farmacologia , Ubiquitina Tiolesterase/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/metabolismo , Antineoplásicos/farmacologia , Benzimidazóis/farmacologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Piperidinas/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise/efeitos dos fármacos , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Tirosina Quinase 3 Semelhante a fms/metabolismo
5.
Cancer Invest ; 37(6): 242-252, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31296070

RESUMO

Drug resistance to TKIs and the existance of CML leukemia stem cells is an urgent problem. In this study, we demonstrate that quinacrine (QC) induces apoptosis in BCR-ABL positive CML and acute lymphoblastic leukemia (ALL) cells. Interestingly, QC inhibits the colony formation of primary CD34+ progenitor/stem leukemia cells from CML patients. QC targets RNA polymerase I, which produces ribosomal (r)RNA, involving in protein translation process. Also, QC treatment prolongs CML-like mice survival and inhibits K562 tumor growth in vivo. In conclusion, we demonstrate that QC depletes BCR-ABL protein and suppresses Ph-positive leukemia cells in vitro and in vivo.


Assuntos
Proteínas de Fusão bcr-abl/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Quinacrina/uso terapêutico , Animais , Antígenos CD34/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Inibidores de Proteínas Quinases/uso terapêutico
6.
Cancer Cell Int ; 19: 56, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30911287

RESUMO

BACKGROUND: T-cell acute lymphoblastic leukemia (T-ALL) is a lymphoid malignancy caused by the oncogenic transformation of immature T-cell progenitors with poor outcomes. WP1130 has shown potent activity against a variety of cancer but whether WP1130 has anti-T-ALL activity is not clear. USP24, one target of WP1130, is one of the largest deubiquitinases and its detailed mechanism is poorly understood. The aim of this study was to explore whether WP1130 could suppress T-ALL and the role of USP24 in T-ALL. METHODS: Molecular docking and cellular thermal shift assay were performed to determine whether and how WP1130 directly interact with USP24. Mitochondrial transmembrane potential assay was measured via Rhodamine 123 staining. USP24 was reactivated using the deactivated CRISPR-associated protein 9 (dCas9)-synergistic activation mediator (SAM) system. The in vivo results were examined by tumor xenografts in NOD-SCID mice. All statistical analyses were performed with the SPSS software package. RESULTS: WP1130 treatment decreased the viability and induces apoptosis of T-ALL cells both in vitro and in vivo. Furthermore, we demonstrated that knockdown of USP24 but not USP9X could significantly induce growth inhibition and apoptosis of T-ALL cells. Oncomine database showed that USP24 expression was upregulated in T-ALL samples and Kaplan-Meier results indicated that the USP24 was negatively but USP9X was positively associated with survival in T-ALL patients. Additionally, we proposed that WP1130 directly interacts with the activity site pocket of USP24 in T-ALL cells, which leads to the decrease of its substrates Mcl-1. Mechanistically, WP1130 induces apoptosis by accelerating the collapse of mitochondrial transmembrane potential via USP24-Mcl-1 axis. CONCLUSIONS: Altogether, using WP1130 as a chemical probe, we demonstrate that USP24 but not USP9X is a novel target in T-ALL cells. Moreover, we uncovered that WP1130 induces apoptosis by accelerating the collapse of mitochondrial transmembrane potential via USP24-Mcl-1 axis. These results provide that USP24-Mcl-1 axis may represent a novel strategy in the treatment of T-ALL and WP1130 is a promising lead compound for developing anti-T-ALL drugs.

7.
Chemphyschem ; 19(5): 596-611, 2018 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-29205732

RESUMO

The notoriously non-luminescent uncycled azophenine (Q) was harnessed with Bodipy and zinc(II)porphyrin antennas to probe its fluorescence properties, its ability to act as a singlet excited state energy acceptor and to mediate the transfer. Two near-IR emissions are depicted from time-resolved fluorescence spectroscopy, which are most likely due to the presence of tautomers of very similar calculated total energies (350 cm-1 ; DFT; B3LYP). The rates for energy transfer, kET (S1 ), for 1 Bodipy*→Q are in the order of 1010 -1011  s-1 and are surprisingly fast when considering the low absorptivity properties of the lowest energy charge transfer excited state of azophenine. The rational is provided by the calculated frontier molecular orbitals (MOs) which show atomic contributions in the C6 H4 C≡CC6 H4 arms, thus favoring the double electron exchange mechanism. In the mixed-antenna Bodipy-porphyrin star molecule, the rate for 1 Bodipy*→porphyrin has also been evaluated (≈16×1010  s-1 ) and is among the fastest rates reported for Bodipy-zinc(II)porphyrin pairs. This astonishing result is again explained from the atomic contributions of the C6 H4 C≡CC6 H4 and C≡CC6 H4 arms thus favouring the Dexter process. Here, for the first time, this process is found to be sensitively temperature-dependent. The azophenine turns out to be excellent for electronic communication.

8.
Acta Pharmacol Sin ; 39(3): 492-498, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29168472

RESUMO

Deubiquitinating protease USP7 is a promising therapeutic target for cancer treatment, and interest in developing USP7 inhibitors has greatly increased. In the present study, we reported a series of natural pentacyclic triterpenes with USP7 inhibitory activity in vitro. Among them, both the ursane triterpenes and oleanane triterpenes were more active than the lupine triterpenes, whereas ursolic acid was the most potent with IC50 of 7.0±1.5 µmol/L. Molecular docking studies showed that ursolic acid might occupy the ubiquitin binding pocket of USP7, with the 17-carboxyl group and 3-hydroxyl group playing a vital role in the USP7-ursolic acid interaction. Using the cellular thermal shift assay, we demonstrated that ursolic acid interacted with USP7 in RPMI8226 human myeloma cells. Ursolic acid dose-dependently inhibited the proliferation of the myeloma cells with IC50 of 6.56 µmol/L, accompanied by reductions in USP7 substrates such as MDM2, UHRF1 and DNMT1. Overexpression of USP7 partially, but significantly attenuated ursolic acid-induced cell death as well as downregulation of MDM2, UHRF1 and DNMT1. In conclusion, we demonstrate for the first time that pentacyclic triterpenes represent a novel scaffold for developing USP7 inhibitors and that USP7 inhibition contributes to the anti-cancer effect of ursolic acid.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Triterpenos Pentacíclicos/farmacologia , Peptidase 7 Específica de Ubiquitina/antagonistas & inibidores , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Relação Dose-Resposta a Droga , Humanos , Simulação de Acoplamento Molecular , Neoplasias/patologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Relação Estrutura-Atividade , Triterpenos/antagonistas & inibidores , Triterpenos/farmacologia , Ubiquitina-Proteína Ligases , Peptidase 7 Específica de Ubiquitina/biossíntese , Ácido Ursólico
9.
Cancer Cell Int ; 17: 101, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29142505

RESUMO

Cancer stem cells (CSCs) are rare but accounted for tumor initiation, progression, metastasis, relapse and therapeutic resistance. Ubiquitination and deubiquitination of stemness-related proteins are essential for CSC maintenance and differentiation, even leading to execute various stem cell fate choices. Deubiquitinating enzymes (DUBs), specifically disassembling ubiquitin chains, are important to maintain the balance between ubiquitination and deubiquitination. In this review, we have focused on the DUBs regulation of stem cell fate determination. For example, we discuss deubiquitinase inhibition may lead stem cell transcription factors and CSCs-related protein degradation. Also, CSCs microenvironment is regulated by DUBs activity. Our review provides a new insight into DUBs activity by emphasizing their cellular role in regulating stem cell fate and illustrates the opportunities for the application of DUBs inhibitors in the CSC-targeted therapy.

10.
Inorg Chem ; 56(21): 13140-13151, 2017 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-29022705

RESUMO

Azophenine, (α-C6H5NH)2(C6H5-N═C6H2═N-C6H5), well known to be non-emissive, was rigidified by replacing two amine protons by two difluoroboranes (BF2+) and further functionalized at the para-positions of the phenyl groups by luminescent trans-ArC≡C-Pt(PR3)2-C≡C ([Pt]) arms [Ar = C6H4 (R = Et), hexa(n-hexyl)truxene) (Tru; R = Bu)]. Two effects are reported. First, the linking of these [Pt] arms with the central azophenine (C6H4-N═C6H2(NH)2═N-C6H4; Q) generates very low energy charge-transfer (CT) singlet and triplet excited states (3,1([Pt]-to-Q)*) with absorption bands extending all the way to 800 nm. Second, the rigidification of azophenine by the incorporation of BF2+ units renders the low-lying CT singlet state clearly emissive at 298 and 77 K in the near-IR region. DFT computations place the triplet emission in the 1200-1400 nm range, but no phosphorescence was detected. The photophysical properties are investigated, and circumstantial evidence for slow triplet energy transfers, 3Tru* → Q, is provided.

11.
Phys Chem Chem Phys ; 19(32): 21532-21539, 2017 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-28762412

RESUMO

An azophenine derivative was synthesized by coupling truxene and azophenine via the copper-free Sonagashira reaction using Pd2(dba)3 and As(PPh)3 as catalysts. The crystal structure of this heavy azophenine model (∼4000) was made and the identity of the structure was confirmed. By introducing truxene groups into this cross-conjugated structure, the deactivating rotations around the NH-C6H4 bonds were slowed down, which rendered this derivative near-IR emissive at 298 K. This species provided then the appropriate spectral and kinetic signatures for knowing where and what to look for in emeraldine, which was called non-emissive. Besides, two other compounds were also synthesized as models for this azophenine derivative for comparison and interpretation purposes.

12.
J Proteome Res ; 13(8): 3571-82, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25027693

RESUMO

Sentrin/SUMO (small ubiquitin-like modifier)-specific proteases (SENPs) have been implicated in the development of prostate cancer. However, due to the low abundance of SUMO-modified proteins and high activity of SENPs, the SUMO substrates affected by SENPs in prostate cancer cells are largely unknown. Here, we identified SI2, a novel cell-permeable SENP-specific inhibitor, by high-throughput screening. Using SI2 as a way of inhibiting the activity of SENPs and the SUMO stably transfected PC3 cells as a prostate cancer model, in combination with the stable isotope labeling with amino acids (SILAC) quantitative proteomic technique, we identified more than 900 putative target proteins of SUMO, in which 231 proteins were further subjected to bioinformatic analysis. In the highly enriched spliceosome pathway, we validated that USP39, HSPA1A, and HSPA2 were novel target proteins of SUMO. Furthermore, we demonstrated that K6, K16, K29, K51, and K73 were the SUMOylation sites of USP39. Mutation of these SUMO modification sites of USP39 further promoted the proliferation-enhancing effect of USP39 on prostate cancer cells. This study provides the SUMOproteome of PC3 cells and reveals that SUMOylation of spliceosome factors may be implicated in the pathogenesis of prostate cancer. Optimization of SI2 for isotype-specific SENP inhibitors warrants further investigation.


Assuntos
Benzoatos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Endopeptidases/metabolismo , Neoplasias da Próstata/metabolismo , Inibidores de Proteases/farmacologia , Spliceossomos/metabolismo , Sumoilação/fisiologia , Benzoatos/química , Benzoatos/isolamento & purificação , Sítios de Ligação/genética , Western Blotting , Compostos Bicíclicos Heterocíclicos com Pontes/química , Compostos Bicíclicos Heterocíclicos com Pontes/isolamento & purificação , Catepsina B/metabolismo , Catepsina D/metabolismo , Cromatografia Líquida , Biologia Computacional , Cisteína Endopeptidases , Células HEK293 , Proteínas de Choque Térmico HSP70/metabolismo , Ensaios de Triagem em Larga Escala , Humanos , Imunoprecipitação , Concentração Inibidora 50 , Marcação por Isótopo , Masculino , Plasmídeos/genética , Inibidores de Proteases/química , Inibidores de Proteases/isolamento & purificação , Proteômica , Espectrometria de Massas em Tandem , Proteases Específicas de Ubiquitina/genética , Proteases Específicas de Ubiquitina/metabolismo
13.
Med Eng Phys ; 119: 104031, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37634913

RESUMO

For robot-assisted pelvic fracture reduction, at least two bone needles need to be inserted into the ilium of the affected pelvis, and the robot clamping device is connected with the bone needles. The biomechanical properties of the pelvic musculoskeletal tissues are different with the different Spatial Position and Orientation (SPO) of the bone needles. In order to determine the optimal SPO of bone needle pairs, the constraints between the bone needles and the pelvis are analyzed, and the SPO vectors of 150 groups bone needles are obtained by the KNN-hierarchical clustering method; a batch modeling method of bone needles with different SPO is proposed. 150 finite element models of damaged pelvic musculoskeletal tissue with different SPO of bone needles are established and simulated. The stress and strain distribution homogenization of musculoskeletal tissue with bone needles as evaluation index, the simulation results of 150 models are evaluated. Results show that, the anterior superior iliac spine and the anterior inferior iliac spine are suitable regions to place bone needles in the pelvis, and the optimal distribution of the needle combination is found in this region. The overall stress and strain distribution of the damaged pelvic musculoskeletal tissue under the large reduction force is the best.


Assuntos
Fraturas Ósseas , Tração , Humanos , Agulhas , Pelve , Fixação de Fratura
14.
Cell Signal ; 110: 110837, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37544636

RESUMO

BACKGROUND: Despite some progress having been made regarding the treatment of T-cell acute lymphoblastic leukemia (T-ALL), the prognosis of T-ALL, particularly adult T-ALL, is still poor. Identifying novel, effective anti-T-ALL drugs is of great significance. Anlotinib, an oral tyrosine kinase inhibitor currently utilized in the treatment of lung cancer, exhibited a promising anti-T-ALL effect. A comprehensive study should therefore be conducted to explore both the in vitro as well as in vivo mechanisms of the anti-T-ALL effects of anlotinib. METHODS: CCK8 assays and flow cytometry were employed to investigate the viability, cell cycle distribution, and apoptosis of T-ALL cell lines when treated with anlotinib. T-ALL xenograft mouse models were established to examine the in vivo antileukemic effects of anlotinib. Cellular and molecular analysis of T-ALL were conducted to define the underlying mechanisms. RESULTS: In vitro, anlotinib significantly inhibited the viability, induced G2/M phase arrest and apoptosis in T-ALL cell lines in a concentration-dependent pattern. In vivo, anlotinib also demonstrated a strong anti-tumor effect at doses that are well-tolerated. Interestingly, anlotinib could decrease the protein levels of the intracellular domains of NOTCH1 (ICN1) and c-Myc, two important targets for T-ALL. Mechanistically, anlotinib-induced c-Myc reduction was associated with proteasome-mediated degradation, while the ICN1 reduction was not due to protein degradation or transcriptional repression. CONCLUSIONS: The present study showed that anlotinib may be a promising anti-T-ALL candidate drug, and simultaneous reduction of the protein levels of both ICN1 and c-Myc may contribute to the anti-T-ALL efficacy of anlotinib.


Assuntos
Leucemia-Linfoma Linfoblástico de Células Precursoras , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Quinolinas , Humanos , Camundongos , Animais , Linhagem Celular Tumoral , Transdução de Sinais , Indóis/farmacologia , Indóis/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Proliferação de Células , Apoptose
15.
Exp Hematol Oncol ; 11(1): 42, 2022 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-35836282

RESUMO

Pyruvate kinase M2 (PKM2) plays an important role in the metabolism and proliferation of leukemia cells. Here, we show that deubiquitinase JOSD2, a novel tumor suppressor, blocks PKM2 nuclear localization by reducing its K433 acetylation in acute myeloid leukemia (AML). Firstly, we show that JOSD2 is significantly down-regulated in primary AML cells. Reconstitute of JOSD2 in AML cells significantly inhibit cell viability and induce cell apoptosis. Next, PKM2 is identified as a novel interaction protein of JOSD2 by mass spectrometry, co- immunoprecipitation and co-immunofluorescence in HL60 cells. However, JOSD2 does not affect PKM2 protein stability. We then found out that JOSD2 inhibits nuclear localization of PKM2 by reducing its K433 acetylation modification, accompanied by decreased downstream gene expression through non-glycolytic functions. Finally, JOSD2 decreases AML progression in vivo. Taken together, we propose that JOSD2 blocks PKM2 nuclear localization and reduces AML progression.

16.
Sci China Life Sci ; 65(8): 1624-1635, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35235149

RESUMO

Deubiquitinates (DUBs) alter the stabilities, localizations or activities of substrates by removing their ubiquitin conjugates, which are closely related to the development of inflammatory response. Here, we show that ubiquitin-specific protease 47 (USP47) prevents inflammation development in inflammatory bowel disease (IBD). Compared with wild-type mice, Usp47 knockout mice are more susceptible to dextran sodium sulfate (DSS)-induced acute and chronic colitis with higher inflammatory cytokines expression and severe intestinal tissue damage. Chimeric mouse experiments suggest that non-hematopoietic cells mainly contribute to the phenotype. And, DSS-induced colitis of the Usp47 knockout mice depends on commensal bacteria. Mechanistically, down-regulation of USP47 aggravates the activation of NF-κB signaling pathway by increasing the K63-linked poly-ubiquitination of tumor necrosis factor receptor-associated factor 6 (TRAF6) in intestinal epithelial cells. Furthermore, the expression of USP47, negatively correlated with the degree of inflammation, is lower at colonic inflammatory lesions than that non-inflammatory sites from the intestine from ulcerative colitis (UC) and Crohn's disease (CD) patients. These data, taken together, indicate that USP47 regulates intestinal inflammation through de-ubiquitination of K63-linked poly-ubiquitination TRAF6 in intestinal epithelial cells.


Assuntos
Colite , Fator 6 Associado a Receptor de TNF , Proteases Específicas de Ubiquitina , Animais , Colite/induzido quimicamente , Colite/patologia , Células Epiteliais/metabolismo , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/metabolismo , Proteases Específicas de Ubiquitina/genética , Proteases Específicas de Ubiquitina/metabolismo
17.
Front Nutr ; 9: 915776, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35983487

RESUMO

Laoxianghuang, fermented from Citrus medica L. var. Sarcodactylis Swingle of the Rutaceae family, is a medicinal food. The volatiles of Laoxianghuang fermented in different years were obtained by solid-phase microextraction combined with gas chromatography-mass spectrometry (SPME-GC-MS). Meanwhile, the evolution of its component-flavor function during the fermentation process was explored in depth by combining chemometrics and performance analyses. To extract the volatile compounds from Laoxianghuang, the fiber coating, extraction time, and desorption temperature were optimized in terms of the number and area of peaks. A polydimethylsiloxane/divinylbenzene (PDMS/DVB) with a thickness of 65 µm fiber, extraction time of 30 min, and desorption temperature of 200 °C were shown to be the optimal conditions. There were 42, 44, 52, 53, 53, and 52 volatiles identified in the 3rd, 5th, 8th, 10th, 15th, and 20th years of fermentation of Laoxianghuang, respectively. The relative contents were 97.87%, 98.50%, 98.77%, 98.85%, 99.08%, and 98.36%, respectively. Terpenes (mainly limonene, γ-terpinene and cymene) displayed the highest relative content and were positively correlated with the year of fermentation, followed by alcohols (mainly α-terpineol, ß-terpinenol, and γ-terpineol), ketones (mainly cyclohexanone, D(+)-carvone and ß-ionone), aldehydes (2-furaldehyde, 5-methylfurfural, and 1-nonanal), phenols (thymol, chlorothymol, and eugenol), esters (bornyl formate, citronellyl acetate, and neryl acetate), and ethers (n-octyl ether and anethole). Principal component analysis (PCA) and hierarchical cluster analysis (HCA) showed a closer relationship between the composition of Laoxianghuang with similar fermentation years of the same gradient (3rd-5th, 8th-10th, and 15th-20th). Partial least squares discriminant analysis (PLS-DA) VIP scores and PCA-biplot showed that α-terpineol, γ-terpinene, cymene, and limonene were the differential candidate biomarkers. Flavor analysis revealed that Laoxianghuang exhibited wood odor from the 3rd to the 10th year of fermentation, while herb odor appeared in the 15th and the 20th year. This study analyzed the changing pattern of the flavor and function of Laoxianghuang through the evolution of the composition, which provided a theoretical basis for further research on subsequent fermentation.

18.
Chem Biol Interact ; 351: 109770, 2022 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-34861246

RESUMO

INTRODUCTION: Despite the rapid progress in the diagnosis and treatment, the prognosis of some types of non-Hodgkin's lymphoma (NHL), especially those with double-hit or double-expressor genotypes, remains poor. Novel targets and compounds are needed to improve the prognosis of NHL. METHODS: We investigated the effect of ZCL-082, a novel boron-containing compound with anti-proliferating activity against ovarian cancer cells, on NHL cells and human peripheral blood mononuclear cells by CCK-8 assay, Annexin V/PI double staining assay, RH123/PI double staining, Western blot, and immunohistochemistry. NF-κB pathway activity was analyzed using luciferase reporter gene assay and RT-PCR. The location of p65 was detected by immunofluorescence and nuclear/cytoplasmic fractionation assay. Immunoprecipitation and chromatin immunoprecipitation assays were used to detect the binding between p65 and p300. CETSA and molecular docking assay were carried out to test the interaction between ZCL-082 and p90 ribosomal S6 kinase 1 (RSK1). Kinase reaction was conducted to examine the inhibition of RSK1 kinase activity by ZCL-082. RESULTS: We found that ZCL-082 can induce the apoptosis of various NHL cell lines in vitro and in vivo. ZCL-082 significantly inhibits TNFα- or LPS-induced NF-κB activation without disturbing TNFα-induced IκBα degradation or the nuclear translocation and DNA-binding ability of p65. However, ZCL-082 markedly suppresses the phosphorylation of p65 on Ser536 and the interaction between p65 and p300. The overexpression of the phosphomimetic mutant of p65 at Ser536 partially abrogates ZCL-082-induced cell death. We further found that ZCL-082 directly binds to and inhibits the activity of RSK1. RSK1 can phosphorylate RelA/p65 on Ser536 and its overexpression is associated with the poor prognosis of lymphoma. The overexpression of RSK1 partially rescues ZCL-082-induced cell death. Molecular docking studies show that ZCL-082 fits well with the N-terminal kinase domain of RSK1. Furthermore, the combination of ZCL-082 and BCL-2 inhibitor ABT-199 has a synergistic apoptosis-inducing effect against double-hit lymphoma cell line OCI-Ly10. DISCUSSION: We found that ZCL-082 is a highly promising anti-lymphoma compound that targets RSK1 and interferes with the RSK1/NF-κB signaling pathway. The combination of ZCL-082 with BCL-2 inhibitor may represent a novel strategy to improve the outcome of double-hit or double-expressor lymphoma.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Compostos de Boro/uso terapêutico , Linfoma não Hodgkin/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos/farmacologia , Compostos de Boro/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Fator de Transcrição RelA/química , Fator de Transcrição RelA/metabolismo
19.
Int J Biol Sci ; 18(6): 2515-2526, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35414773

RESUMO

Rationale: In multiple myeloma (MM), the activities of non-homologous end joining (NHEJ) and homologous recombination repair (HR) are increased compared with healthy controls. Whether and how IKZF1 as an enhancer of MM participates in the DNA repair pathway of tumor cells remains elusive. Methods: We used an endonuclease AsiSI-based system and quantitative chromatin immunoprecipitation assay (qChIP) analysis to test whether IKZF1 is involved in DNA repair. Immunopurification and mass spectrometric (MS) analysis were performed in MM1.S cells to elucidate the molecular mechanism that IKZF1 promotes DNA damage repair. The combination effect of lenalidomide or USP7 inhibitor with PARP inhibitor on cell proliferation was evaluated using MM cells in vitro and in vivo. Results: We demonstrate that IKZF1 specifically promotes homologous recombination DNA damage repair in MM cells, which is regulated by its interaction with CtIP and USP7. In this process, USP7 could regulate the stability of IKZF1 through its deubiquitinating activity. The N-terminal zinc finger domains of IKZF1 and the ubiquitin-like domain of USP7 are necessary for their interaction. Furthermore, targeted inhibition IKZF1 or USP7 could sensitize MM cells to PARP inhibitor treatment in vitro and in vivo. Conclusions: Our findings identify USP7 as a deubiquitinating enzyme for IKZF1 and uncover a new function of IKZF1 in DNA damage repair. In translational perspective, the combination inhibition of IKZF1 or USP7 with PARP inhibitor deserves further evaluation in clinical trials for the treatment of MM.


Assuntos
Antineoplásicos , Mieloma Múltiplo , Antineoplásicos/farmacologia , Reparo do DNA/genética , Endodesoxirribonucleases , Humanos , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Reparo de DNA por Recombinação , Peptidase 7 Específica de Ubiquitina/genética , Peptidase 7 Específica de Ubiquitina/metabolismo
20.
Biomark Res ; 9(1): 66, 2021 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-34454635

RESUMO

Deubiquitinases (DUBs) are enzymes that control the stability, interactions or localization of most cellular proteins by removing their ubiquitin modification. In recent years, some DUBs, such as USP7, USP9X and USP10, have been identified as promising therapeutic targets in hematological malignancies. Importantly, some potent inhibitors targeting the oncogenic DUBs have been developed, showing promising inhibitory efficacy in preclinical models, and some have even undergone clinical trials. Different DUBs perform distinct function in diverse hematological malignancies, such as oncogenic, tumor suppressor or context-dependent effects. Therefore, exploring the biological roles of DUBs and their downstream effectors will provide new insights and therapeutic targets for the occurrence and development of hematological malignancies. We summarize the DUBs involved in different categories of hematological malignancies including leukemia, multiple myeloma and lymphoma. We also present the recent development of DUB inhibitors and their applications in hematological malignancies. Together, we demonstrate DUBs as potential therapeutic drug targets in hematological malignancies.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA