Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Nat Commun ; 15(1): 6952, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39138204

RESUMEN

Biomolecular condensates play a significant role in chromatin activities, primarily by concentrating and compartmentalizing proteins and/or nucleic acids. However, their genomic landscapes and compositions remain largely unexplored due to a lack of dedicated computational tools for systematic identification in vivo. To address this, we develop CondSigDetector, a computational framework designed to detect condensate-like chromatin-associated protein co-occupancy signatures (CondSigs), to predict genomic loci and component proteins of distinct chromatin-associated biomolecular condensates. Applying this framework to mouse embryonic stem cells (mESC) and human K562 cells enable us to depict the high-resolution genomic landscape of chromatin-associated biomolecular condensates, and uncover both known and potentially unknown biomolecular condensates. Multi-omics analysis and experimental validation further verify the condensation properties of CondSigs. Additionally, our investigation sheds light on the impact of chromatin-associated biomolecular condensates on chromatin activities. Collectively, CondSigDetector provides an approach to decode the genomic landscape of chromatin-associated condensates, facilitating a deeper understanding of their biological functions and underlying mechanisms in cells.


Asunto(s)
Condensados Biomoleculares , Cromatina , Cromatina/metabolismo , Cromatina/genética , Humanos , Animales , Ratones , Células K562 , Condensados Biomoleculares/metabolismo , Genómica/métodos , Células Madre Embrionarias de Ratones/metabolismo , Biología Computacional/métodos , Genoma
2.
J Med Chem ; 67(2): 1079-1092, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38166388

RESUMEN

The DNA-encoded library (DEL) is a powerful hit generation tool for chemical biology and drug discovery; however, the optimization of DEL hits remained a daunting challenge for the medicinal chemistry community. In this study, hit compounds targeting the WIN binding domain of WDR5 were discovered by the initial three-cycle linear DEL selection, and their potency was further enhanced by a cascade DEL selection from the focused DEL designed based on the original first run DEL hits. As expected, these new compounds from the second run of focused DEL were more potent WDR5 inhibitors in the protein binding assay confirmed by the off-DNA synthesis. Interestingly, selected inhibitors exhibited good antiproliferative activity in two human acute leukemia cell lines. Taken together, this new cascade DEL selection strategy may have tremendous potential for finding high-affinity leads against WDR5 and provide opportunities to explore and optimize inhibitors for other targets.


Asunto(s)
ADN , Descubrimiento de Drogas , Humanos , Biblioteca de Genes , Unión Proteica , ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(42): e2219589120, 2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37812694

RESUMEN

NTRK (neurotrophic tyrosine receptor kinase) gene fusions that encode chimeric proteins exhibiting constitutive activity of tropomyosin receptor kinases (TRK), are oncogenic drivers in multiple cancer types. However, the underlying mechanisms in oncogenesis that involve various N-terminal fusion partners of NTRK fusions remain elusive. Here, we show that NTRK fusion proteins form liquid-like condensates driven by their N-terminal fusion partners. The kinase reactions are accelerated in these condensates where the complexes for downstream signaling activation are also concentrated. Our work demonstrates that the phase separation driven by NTRK fusions is not only critical for TRK activation, but the condensates formed through phase separation serve as organizational hubs for oncogenic signaling.


Asunto(s)
Neoplasias , Proteínas de Fusión Oncogénica , Humanos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/genética , Neoplasias/genética , Neoplasias/metabolismo , Fusión Génica , Receptor trkA/genética , Receptor trkA/metabolismo , Inhibidores de Proteínas Quinasas
4.
ACS Synth Biol ; 12(7): 2198-2202, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37348112

RESUMEN

Recently discovered CRISPR-associated transposases (CASTs) have been implemented as useful multiplexed genome editing tools, albeit only in a small group of bacteria. We demonstrated that the type I-F CAST from Vibrio cholerae could induce RNA-guided transposition in Bacillus subtilis and Corynebacterium glutamicum with efficiencies of 0.00018% and 0.027%, respectively. Lowering the culturing temperature to 16 °C in rich media increased the insertion efficiency to 3.64% in B. subtilis. By adding a positive selection against spectinomycin in the cargo DNA, up to 9 kb of the DNA fragment could be integrated at the target site with a 13.4% efficiency in C. glutamicum, which was difficult using the conventional approach. The successful implementation of CAST in these two academically important and industrial-relevant Firmicutes shows its great potential in a wide variety of bacteria and could be further optimized for multiplexed genome editing.


Asunto(s)
Sistemas CRISPR-Cas , Corynebacterium glutamicum , Sistemas CRISPR-Cas/genética , Bacillus subtilis/genética , Corynebacterium glutamicum/genética , ARN , Edición Génica
5.
JAMA Neurol ; 80(6): 578-587, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37126322

RESUMEN

Importance: Mesial temporal lobe epilepsy (MTLE) is the most common focal epilepsy subtype and is often refractory to antiseizure medications. While most patients with MTLE do not have pathogenic germline genetic variants, the contribution of postzygotic (ie, somatic) variants in the brain is unknown. Objective: To test the association between pathogenic somatic variants in the hippocampus and MTLE. Design, Setting, and Participants: This case-control genetic association study analyzed the DNA derived from hippocampal tissue of neurosurgically treated patients with MTLE and age-matched and sex-matched neurotypical controls. Participants treated at level 4 epilepsy centers were enrolled from 1988 through 2019, and clinical data were collected retrospectively. Whole-exome and gene-panel sequencing (each genomic region sequenced more than 500 times on average) were used to identify candidate pathogenic somatic variants. A subset of novel variants was functionally evaluated using cellular and molecular assays. Patients with nonlesional and lesional (mesial temporal sclerosis, focal cortical dysplasia, and low-grade epilepsy-associated tumors) drug-resistant MTLE who underwent anterior medial temporal lobectomy were eligible. All patients with available frozen tissue and appropriate consents were included. Control brain tissue was obtained from neurotypical donors at brain banks. Data were analyzed from June 2020 to August 2022. Exposures: Drug-resistant MTLE. Main Outcomes and Measures: Presence and abundance of pathogenic somatic variants in the hippocampus vs the unaffected temporal neocortex. Results: Of 105 included patients with MTLE, 53 (50.5%) were female, and the median (IQR) age was 32 (26-44) years; of 30 neurotypical controls, 11 (36.7%) were female, and the median (IQR) age was 37 (18-53) years. Eleven pathogenic somatic variants enriched in the hippocampus relative to the unaffected temporal neocortex (median [IQR] variant allele frequency, 1.92 [1.5-2.7] vs 0.3 [0-0.9]; P = .01) were detected in patients with MTLE but not in controls. Ten of these variants were in PTPN11, SOS1, KRAS, BRAF, and NF1, all predicted to constitutively activate Ras/Raf/mitogen-activated protein kinase (MAPK) signaling. Immunohistochemical studies of variant-positive hippocampal tissue demonstrated increased Erk1/2 phosphorylation, indicative of Ras/Raf/MAPK activation, predominantly in glial cells. Molecular assays showed abnormal liquid-liquid phase separation for the PTPN11 variants as a possible dominant gain-of-function mechanism. Conclusions and Relevance: Hippocampal somatic variants, particularly those activating Ras/Raf/MAPK signaling, may contribute to the pathogenesis of sporadic, drug-resistant MTLE. These findings may provide a novel genetic mechanism and highlight new therapeutic targets for this common indication for epilepsy surgery.


Asunto(s)
Epilepsia Refractaria , Epilepsia del Lóbulo Temporal , Epilepsia , Neocórtex , Humanos , Femenino , Adulto , Persona de Mediana Edad , Masculino , Epilepsia del Lóbulo Temporal/cirugía , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estudios Retrospectivos , Hipocampo/patología , Epilepsia/patología
6.
Nat Chem Biol ; 18(12): 1341-1350, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36229685

RESUMEN

Patients with castration-resistant prostate cancer inevitably acquire resistance to antiandrogen therapies in part because of androgen receptor (AR) mutations or splice variants enabling restored AR signaling. Here we show that ligand-activated AR can form transcriptionally active condensates. Both structured and unstructured regions of AR contribute to the effective phase separation of AR and disordered N-terminal domain plays a predominant role. AR liquid-liquid phase separation behaviors faithfully report transcriptional activity and antiandrogen efficacy. Antiandrogens can promote phase separation and transcriptional activity of AR-resistant mutants in a ligand-independent manner. We conducted a phase-separation-based phenotypic screen and identified ET516 that specifically disrupts AR condensates, effectively suppresses AR transcriptional activity and inhibits the proliferation and tumor growth of prostate cancer cells expressing AR-resistant mutants. Our results demonstrate liquid-liquid phase separation as an emerging mechanism underlying drug resistance and show that targeting phase separation may provide a feasible approach for drug discovery.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Neoplasias de la Próstata , Masculino , Humanos , Receptores Androgénicos/genética , Antagonistas de Andrógenos/farmacología , Antagonistas de Andrógenos/uso terapéutico , Ligandos , Resistencia a Antineoplásicos , Neoplasias de la Próstata/tratamiento farmacológico , Línea Celular Tumoral , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología
7.
Biochem Biophys Res Commun ; 600: 150-155, 2022 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-35219918

RESUMEN

SHP1 is a non-receptor protein tyrosine phosphatase that is widely expressed in hematopoietic cells such as white blood cells, neutrophils, and immune cells. SHP1 can regulate the occurrence and differentiation of immune cells and plays an important role as a tumor suppressor. Previous studies have suggested that SHP2, the homologous protein of phosphatase SHP1, can undergo liquid-liquid phase separation (LLPS). Therefore, in this study, we investigated if SHP1 is also capable of LLPS. To the best of our knowledge, our study is the first to reveal that SHP1 has the ability to undergo LLPS. In addition, we identified an important residue, SHP1-R360E, that can completely inhibit the LLPS ability of SHP1, but this mutation has no remarkable effect on SHP1's enzymatic activity. This allows us to explore the phosphatase activity and phase separation ability of SHP1 separately, providing a basis for future exploration of the phase separation mechanism of phosphatases.


Asunto(s)
Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Diferenciación Celular , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo
9.
J Biol Chem ; 296: 100538, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33722610

RESUMEN

The protein tyrosine phosphatase SHP2 mediates multiple signal transductions in various cellular pathways, controlled by a variety of upstream inputs. SHP2 dysregulation is causative of different types of cancers and developmental disorders, making it a promising drug target. However, how SHP2 is modulated by its different regulators remains largely unknown. Here, we use single-molecule fluorescence resonance energy transfer and molecular dynamics simulations to investigate this question. We identify a partially open, semiactive conformation of SHP2 that is intermediate between the known open and closed states. We further demonstrate a "multiple gear" regulatory mechanism, in which different activators (e.g., insulin receptor substrate-1 and CagA), oncogenic mutations (e.g., E76A), and allosteric inhibitors (e.g., SHP099) can shift the equilibrium of the three conformational states and regulate SHP2 activity to different levels. Our work reveals the essential role of the intermediate state in fine-tuning the activity of SHP2, which may provide new opportunities for drug development for relevant cancers.


Asunto(s)
Calgranulina A/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Piperidinas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/química , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Pirimidinas/metabolismo , Regulación Alostérica , Humanos , Simulación de Dinámica Molecular , Mutación , Unión Proteica , Conformación Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética
10.
RSC Adv ; 11(38): 23270-23279, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-35479790

RESUMEN

We report optimization by rational design of JMPDP-027, a potent cyclic peptide that interferes with the PD-1/PD-L1 protein-protein interaction. JMPDP-027 shows a potent restoring ability towards T-cells with an EC50 of 5.9 nM that is comparable to that of the anti-PD-1 monoclonal antibody pembrolizumab. In addition, JMPDP-027 shows not only high resistance to enzymatic hydrolysis in human serum but also no observable toxicity and potent in vivo anticancer activity comparable to that of the mouse PD-L1 antibody in a colon carcinoma (CT26) model. Cyclic peptide antagonists of this sort may provide novel drug candidates for cancer immunotherapy.

11.
J Chromatogr A ; 1636: 461790, 2021 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-33340746

RESUMEN

Epigenetic inheritance in mammals relies in part on propagation of DNA methylation patterns throughout development. UHRF1 (ubiquitin-like containing PHD and RING finger domains 1) is required for maintenance the methylation pattern. It was reported that UHRF1 is overexpressed in a number of cancer types, and its depletion has been established to inhibit growth and invasion of cancer cells. It has been considered as a new therapeutic target for cancer. In the present work, we described a method for screening inhibitors for blocking the formation of UHRF1-methylated DNA (mDNA) complex by using nonequilibrium capillary electrophoresis of the equilibrium mixture (NECEEM). A recombinant UHRF1 with the SRA domain (residues 408-643), a fluorescently labeled double strand mDNA (12 mer) and a known inhibitor mitoxantrone were employed for proof of concept. The method allows to measure the dissociation constant (Kd) of the UHRF1-mDNA complex as well as the rate kinetic constant for complex formation (kon) and dissociation (koff). A small chemical library composed of 60 natural compounds were used to validate the method. Sample pooling strategy was employed to improve the screening throughput. The merit of the method was confirmed by the discovery of two natural products proanthocyanidins and baicalein as the new inhibitors for blocking the formation of UHRF1-mDNA complex. Our work demonstrated that CE represents a straightforward and robust technique for studying UHRF1-mDNA interaction and screening of the inhibitors.


Asunto(s)
Antineoplásicos/análisis , Antineoplásicos/farmacología , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Metilación de ADN/genética , Ensayos de Selección de Medicamentos Antitumorales , Electroforesis Capilar/métodos , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Antineoplásicos/química , Metilación de ADN/efectos de los fármacos , Flavanonas/química , Flavanonas/farmacología , Humanos , Cinética , Mitoxantrona/química , Mitoxantrona/farmacología , Proantocianidinas/química , Proantocianidinas/farmacología
12.
Cell ; 183(2): 490-502.e18, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33002410

RESUMEN

The non-receptor protein tyrosine phosphatase (PTP) SHP2, encoded by PTPN11, plays an essential role in RAS-mitogen-activated protein kinase (MAPK) signaling during normal development. It has been perplexing as to why both enzymatically activating and inactivating mutations in PTPN11 result in human developmental disorders with overlapping clinical manifestations. Here, we uncover a common liquid-liquid phase separation (LLPS) behavior shared by these disease-associated SHP2 mutants. SHP2 LLPS is mediated by the conserved well-folded PTP domain through multivalent electrostatic interactions and regulated by an intrinsic autoinhibitory mechanism through conformational changes. SHP2 allosteric inhibitors can attenuate LLPS of SHP2 mutants, which boosts SHP2 PTP activity. Moreover, disease-associated SHP2 mutants can recruit and activate wild-type (WT) SHP2 in LLPS to promote MAPK activation. These results not only suggest that LLPS serves as a gain-of-function mechanism involved in the pathogenesis of SHP2-associated human diseases but also provide evidence that PTP may be regulated by LLPS that can be therapeutically targeted.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Células A549 , Animales , Niño , Preescolar , Femenino , Mutación con Ganancia de Función/genética , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Células Madre Embrionarias de Ratones , Mutación/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Transducción de Señal , Dominios Homologos src/genética
13.
J Hematol Oncol ; 13(1): 119, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32883331

RESUMEN

Tumor metastasis accounts for the majority of cancer-related deaths; it is therefore important to develop preclinical models that faithfully recapitulate disease progression. Here, we generated paired organoids derived from primary tumors and matched liver metastases in the same colorectal cancer (CRC) patients. Despite the fact that paired organoids exhibit comparable gene expression and cell morphology, organoids from metastatic lesions demonstrate more aggressive phenotypes, tumorigenesis, and metastatic capacity than those from primary lesions. Transcriptional analyses of the paired organoids reveal signature genes and pathways altered during the progression of CRC, including SOX2. Further study shows that inducible knockdown of SOX2 attenuated invasion, proliferation, and liver metastasis outgrowth. Taken together, we use patient-derived paired primary and metastatic cancer organoids to model CRC metastasis and illustrate that SOX2 is associated with CRC progression and may serve as a potential prognostic biomarker and therapeutic target of CRC.


Asunto(s)
Carcinoma/secundario , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/secundario , Modelos Biológicos , Invasividad Neoplásica/patología , Organoides , Animales , Biomarcadores de Tumor , Carcinoma/patología , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Neoplasias Hepáticas/patología , Ratones , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Organoides/efectos de los fármacos , Organoides/patología , Organoides/trasplante , Factores de Transcripción SOXB1/biosíntesis , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/fisiología
14.
Sci Rep ; 10(1): 5265, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32210319

RESUMEN

Anesthetics are used to produce hypnosis and analgesic effects during surgery, but anesthesia for a long time after the operation is not conducive to the recovery of animals or patients. Therefore, finding appropriate treatments to counter the effects of anesthetics could enhance postoperative recovery. In the current study, we discovered the novel role of a GluN2A-selective positive allosteric modulator (PAM) in ketamine-induced anesthesia and investigated the effects of the PAM combined with nalmefene and flumazenil (PNF) in reversing the actions of an anesthetic combination (ketamine-fentanyl-dexmedetomidine, KFD). PAM treatment dose-dependently decreased the duration of the ketamine-induced loss of righting reflex (LORR). Compared with those in the KFD group, the duration of LORR and the analgesic effect of the KFD + PNF group were obviously decreased. Meanwhile, successive administration of PNF and KFD had no adverse effects on the cardiovascular and respiratory systems. Both the KFD group and the KFD + PNF group showed no changes in hepatic and renal function or cognitive function in rats. Moreover, the recovery of motor coordination of the KFD + PNF group was faster than that of the KFD group. In summary, our results suggest the potential application of the PNF combination as an antagonistic treatment strategy for anesthesia.


Asunto(s)
Analgesia , Anestesia , Dexmedetomidina/antagonistas & inhibidores , Fentanilo/antagonistas & inhibidores , Flumazenil/farmacología , Antagonistas de Receptores de GABA-A/farmacología , Ketamina/antagonistas & inhibidores , Naltrexona/análogos & derivados , Antagonistas de Narcóticos/farmacología , Receptores de N-Metil-D-Aspartato/agonistas , Adyuvantes Anestésicos , Regulación Alostérica , Animales , Retraso en el Despertar Posanestésico/tratamiento farmacológico , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos , Femenino , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Naltrexona/farmacología , Nocicepción/efectos de los fármacos , Dimensión del Dolor , Ratas , Reflejo de Enderezamiento/efectos de los fármacos , Prueba de Desempeño de Rotación con Aceleración Constante
15.
Eur J Med Chem ; 183: 111694, 2019 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-31561044

RESUMEN

Isocitrate dehydrogenase 1 (IDH1), which catalyzes the conversion of isocitrate to α-ketoglutarate, is one of key enzymes in the tricarboxylic acid cycle (TCA). Hotspot mutation at Arg132 in IDH1 that alters the function of IDH1 by further converting the α-ketoglutarate(α-KG) to 2-hydroxyglutarate (2-HG) have been identified in a variety of cancers. Because the IDH1 mutations occur in a significant portion of gliomas and glioblastomas, it is important that IDH1 inhibitors have to be brain penetrant to treat IDH1-mutant brain tumors. Here we report the efforts to design and synthesize a novel serial of mutant IDH1 inhibitors with improved activity and the blood-brain barrier (BBB) penetration. We show that compound 5 exhibits good brain exposure and potent 2-HG inhibition in a HT1080-derived mouse xenograft model, which makes it a potential preclinical candidate to treat IDH1-mutant brain tumors.


Asunto(s)
Antineoplásicos/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Descubrimiento de Drogas , Glioma/tratamiento farmacológico , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Glioma/metabolismo , Glioma/patología , Humanos , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
16.
J Med Chem ; 62(6): 3107-3121, 2019 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-30835473

RESUMEN

Triapine, an iron chelator that inhibits ribonucleotide reductase, has been evaluated in clinical trials for cancer treatment. Triapine in combination with other chemotherapeutic agents shows promising efficacy in certain hematologic malignancies; however, it is less effective against many advanced solid tumors, probably due to the unsatisfactory potency and pharmacokinetic properties. In this report, we developed a triapine derivative IC25 (10) with potent antitumor activity. 10 Preferentially inhibited the proliferation of hematopoietic cancers by inducing mitochondria reactive oxygen species production and mitochondrial dysfunction. Unlike triapine, 10 executed cytotoxic action in a copper-dependent manner. 10-Induced up-expression of thioredoxin-interacting protein resulted in decreased thioredoxin activity to permit c-Jun N-terminal kinase and p38 activation and ultimately led to the execution of the cell death program. Remarkedly, 10 showed good bioavailability and inhibited tumor growth in mouse xenograft models. Taken together, our study identifies compound 10 as a copper-dependent antitumor agent, which may be applied to the treatment of hematopoietic cancers.


Asunto(s)
Antineoplásicos/farmacocinética , Muerte Celular/efectos de los fármacos , Cobre/metabolismo , Neoplasias Hematológicas/patología , Piridinas/farmacología , Tiosemicarbazonas/farmacología , Animales , Disponibilidad Biológica , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Activación Enzimática , Neoplasias Hematológicas/metabolismo , Humanos , MAP Quinasa Quinasa 4/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Piridinas/química , Piridinas/farmacocinética , Especies Reactivas de Oxígeno/metabolismo , Tiosemicarbazonas/química , Tiosemicarbazonas/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Cell Chem Biol ; 25(11): 1380-1388.e4, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30174312

RESUMEN

Selenoproteins, defined by the presence of selenocysteines (Sec), play important roles in a wide range of biological processes. All known selenoproteins are marked by the presence of Sec insertion sequence (SECIS) at their mRNA. The lack of an effective analytical method has hindered our ability to explore the selenoproteome and new selenoproteins beyond SECIS. Here, we develop a Sec-specific mass spectrometry-based technique, termed "SecMS," which allows the systematic profiling of selenoproteomes by selective alkylation of Sec. Using SecMS, we quantitatively characterized the age- and stress-regulated selenoproteomes for nine tissues from mice of different ages and mammalian cells, demonstrating tissue-specific selenoproteomes and an age-dependent decline in specific selenoproteins in brains and hearts. We established an integrated platform using SecMS and SECIS-independent selenoprotein (SIS) database and further identified five candidate selenoproteins. The application of this integrated platform provides an effective strategy to explore the selenoproteome independent of SECIS.


Asunto(s)
Espectrometría de Masas , Selenocisteína/análisis , Selenoproteínas/química , Envejecimiento , Alquilación , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Química Encefálica , Línea Celular , Femenino , Células HEK293 , Humanos , Masculino , Espectrometría de Masas/métodos , Ratones Endogámicos C57BL , Miocardio/química , Miocardio/metabolismo , Proteoma/química , Proteoma/metabolismo , Proteómica/métodos , Selenocisteína/metabolismo , Selenoproteínas/metabolismo , Estrés Fisiológico
18.
Proc Natl Acad Sci U S A ; 115(40): E9317-E9324, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30181285

RESUMEN

Protooncogene c-MYC, a master transcription factor, is a major driver of human tumorigenesis. Development of pharmacological agents for inhibiting c-MYC as an anticancer therapy has been a longstanding but elusive goal in the cancer field. E3 ubiquitin ligase cIAP1 has been shown to mediate the activation of c-MYC by destabilizing MAD1, a key antagonist of c-MYC. Here we developed a high-throughput assay for cIAP1 ubiquitination and identified D19, a small-molecule inhibitor of E3 ligase activity of cIAP1. We show that D19 binds to the RING domain of cIAP1 and inhibits the E3 ligase activity of cIAP1 by interfering with the dynamics of its interaction with E2. Blocking cIAP1 with D19 antagonizes c-MYC by stabilizing MAD1 protein in cells. Furthermore, we show that D19 and an improved analog (D19-14) promote c-MYC degradation and inhibit the oncogenic function of c-MYC in cells and xenograft animal models. In contrast, we show that activating E3 ubiquitin ligase activity of cIAP1 by Smac mimetics destabilizes MAD1, the antagonist of MYC, and increases the protein levels of c-MYC. Our study provides an interesting example using chemical biological approaches for determining distinct biological consequences from inhibiting vs. activating an E3 ubiquitin ligase and suggests a potential broad therapeutic strategy for targeting c-MYC in cancer treatment by pharmacologically modulating cIAP1 E3 ligase activity.


Asunto(s)
Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ubiquitinación/efectos de los fármacos , Animales , Antineoplásicos/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Med Chem ; 60(24): 10205-10219, 2017 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-29155585

RESUMEN

SHP2, a cytoplasmic protein-tyrosine phosphatase encoded by the PTPN11 gene, is involved in multiple cell signaling processes including Ras/MAPK and Hippo/YAP pathways. SHP2 has been shown to contribute to the progression of a number of cancer types including leukemia, gastric, and breast cancers. It also regulates T-cell activation by interacting with inhibitory immune checkpoint receptors such as the programmed cell death 1 (PD-1) and B- and T-lymphocyte attenuator (BTLA). Thus, SHP2 inhibitors have drawn great attention by both inhibiting tumor cell proliferation and activating T cell immune responses toward cancer cells. In this study, we report the identification of an allosteric SHP2 inhibitor 1-(4-(6-bromonaphthalen-2-yl)thiazol-2-yl)-4-methylpiperidin-4-amine (23) that locks SHP2 in a closed conformation by binding to the interface of the N-terminal SH2, C-terminal SH2, and phosphatase domains. Compound 23 suppresses MAPK signaling pathway and YAP transcriptional activity and shows antitumor activity in vivo. The results indicate that allosteric inhibition of SHP2 could be a feasible approach for cancer therapy.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Naftalenos/química , Piperidinas/química , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Tiazoles/química , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Administración Oral , Regulación Alostérica/efectos de los fármacos , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Sitios de Unión , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Femenino , Humanos , Concentración 50 Inhibidora , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación , Naftalenos/administración & dosificación , Naftalenos/farmacocinética , Fosfoproteínas/antagonistas & inhibidores , Piperidinas/administración & dosificación , Piperidinas/farmacocinética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/química , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Relación Estructura-Actividad , Tiazoles/administración & dosificación , Tiazoles/farmacocinética , Factores de Transcripción , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Señalizadoras YAP
20.
Eur J Med Chem ; 132: 157-172, 2017 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-28350999

RESUMEN

The inhibition of CYP17 to block androgen biosynthesis is a well validated strategy for the treatment of prostate cancer. Herein we reported the design, synthesis and structure-activity relationship (SAR) study for a series of novel 1,2,3,4- tetrahydrobenzo[4,5]thieno[2,3-c]pyridine derivatives. Some analogs demonstrated a potent inhibition to both rat and human CYP17 protein and reduced testosterone production in human H295R cell line. Some analogs also showed high selectivity against other CYP enzymes such as 3A4, 1A2, 2C9, 2C19 and 2D6, which may limit side effects due to drug-drug interactions. Among these analogs, the most potent compound 9c showed 1.5 fold more potent against rat and human CYP17 protein than that of abiraterone (IC50 = 16 nM and 20 nM vs. 25 nM and 36 nM respectively). In NCI-H295R cells, the inhibitory effect of compound 9c on testosterone production (52± 2%) was also more potent than that of abiraterone (74± 15%) at the concentration of 1 µM. Further, it was shown that 9c reduced plasma testosterone level in a dose-dependent manner in Sprague-Dawley rats. Thus, analog 9c maybe a potential agent used for the treatment of prostate cancer.


Asunto(s)
Piridinas/síntesis química , Piridinas/farmacología , Esteroide 17-alfa-Hidroxilasa/antagonistas & inhibidores , Testosterona/antagonistas & inhibidores , Androstenos/farmacología , Animales , Línea Celular , Descubrimiento de Drogas , Humanos , Concentración 50 Inhibidora , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Piridinas/química , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Testosterona/biosíntesis , Testosterona/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA