Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(21): e2403685121, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38743625

RESUMO

The tumor suppressor LKB1 is a serine/threonine protein kinase that is frequently mutated in human lung adenocarcinoma (LUAD). LKB1 regulates a complex signaling network that is known to control cell polarity and metabolism; however, the pathways that mediate the tumor-suppressive activity of LKB1 are incompletely defined. To identify mechanisms of LKB1-mediated growth suppression, we developed a spheroid-based cell culture assay to study LKB1-dependent growth. We then performed genome-wide CRISPR screens in spheroidal culture and found that LKB1 suppresses growth, in part, by activating the PIKFYVE lipid kinase. Finally, we used chemical inhibitors and a pH-sensitive reporter to determine that LKB1 impairs growth by promoting the internalization of wild-type EGFR in a PIKFYVE-dependent manner.


Assuntos
Quinases Proteína-Quinases Ativadas por AMP , Fosfatidilinositol 3-Quinases , Proteínas Serina-Treonina Quinases , Esferoides Celulares , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Quinases Proteína-Quinases Ativadas por AMP/metabolismo , Quinases Proteína-Quinases Ativadas por AMP/genética , Esferoides Celulares/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/genética , Proliferação de Células , Linhagem Celular Tumoral , Sistemas CRISPR-Cas , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética
2.
Appl Microbiol Biotechnol ; 104(18): 7853-7865, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32725322

RESUMO

Daptomycin is a last resort antibiotic for the treatment of infections caused by many Gram-positive bacterial strains, including vancomycin-resistant Enterococcus (VRE) and methicillin- and vancomycin-resistant Staphylococcus aureus (MRSA and VRSA). However, the emergence of daptomycin-resistant strains of S. aureus and Enterococcus in recent years has renewed interest in synthesizing daptomycin analogs to overcome resistance mechanisms. Within this context, three aromatic prenyltransferases have been shown to accept daptomycin as a substrate, and the resulting prenylated analog was shown to be more potent against Gram-positive strains than the parent compound. Consequently, utilizing prenyltransferases to derivatize daptomycin offered an attractive alternative to traditional synthetic approaches, especially given the molecule's structural complexity. Herein, we report exploiting the ability of prenyltransferase CdpNPT to synthesize alkyl-diversified daptomycin analogs in combination with a library of synthetic non-native alkyl-pyrophosphates. The results revealed that CdpNPT can transfer a variety of alkyl groups onto daptomycin's tryptophan residue using the corresponding alkyl-pyrophosphates, while subsequent scaled-up reactions suggested that the enzyme can alkylate the N1, C2, C5, and C6 positions of the indole ring. In vitro antibacterial activity assays using 16 daptomycin analogs revealed that some of the analogs displayed 2-80-fold improvements in potency against MRSA, VRE, and daptomycin-resistant strains of S. aureus and Enterococcus faecalis. Thus, along with the new potent analogs, these findings have established that the regio-chemistry of alkyl substitution on the tryptophan residue can modulate daptomycin's potency. With additional protein engineering to improve the regio-selectivity, the described method has the potential to become a powerful tool for diversifying complex indole-containing molecules. KEY POINTS: • CdpNPT displays impressive donor promiscuity with daptomycin as the acceptor. • CdpNPT catalyzes N1-, C2-, C5-, and C6-alkylation on daptomycin's tryptophan residue. • Differential alkylation of daptomycin's tryptophan residue modulates its activity.


Assuntos
Daptomicina , Staphylococcus aureus Resistente à Meticilina , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Daptomicina/farmacologia , Testes de Sensibilidade Microbiana , Staphylococcus aureus , Vancomicina
3.
Appl Microbiol Biotechnol ; 104(10): 4383-4395, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32189045

RESUMO

Aromatic prenyltransferases are known for their extensive promiscuity toward aromatic acceptor substrates and their ability to form various carbon-carbon and carbon-heteroatom bonds. Of particular interest among the prenyltransferases is NphB, whose ability to geranylate cannabinoid precursors has been utilized in several in vivo and in vitro systems. It has therefore been established that prenyltransferases can be utilized as biocatalysts for the generation of useful compounds. However, recent observations of non-native alkyl-donor promiscuity among prenyltransferases indicate the role of NphB in biocatalysis could be expanded beyond geranylation reactions. Therefore, the goal of this study was to elucidate the donor promiscuity of NphB using different acceptor substrates. Herein, we report distinct donor profiles between NphB-catalyzed reactions involving the known substrate 1,6-dihydroxynaphthalene and an FDA-approved drug molecule sulfabenzamide. Furthermore, we report the first instance of regiospecific, NphB-catalyzed N-alkylation of sulfabenzamide using a library of non-native alkyl-donors, indicating the biocatalytic potential of NphB as a late-stage diversification tool. KEY POINTS: • NphB can utilize the antibacterial drug sulfabenzamide as an acceptor. • The donor profile of NphB changes dramatically with the choice of acceptor. • NphB performs a previously unknown regiospecific N-alkylation on sulfabenzamide. • Prenyltransferases like NphB can be utilized as drug-alkylating biocatalysts.


Assuntos
Dimetilaliltranstransferase/metabolismo , Streptomyces/enzimologia , Alquilação , Biocatálise , Dimetilaliltranstransferase/química , Cinética , Espectroscopia de Ressonância Magnética , Naftóis/metabolismo , Prenilação , Sensibilidade e Especificidade , Streptomyces/genética , Especificidade por Substrato , Sulfonamidas/metabolismo
4.
Arthroscopy ; 31(5): 901-10, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25661860

RESUMO

PURPOSE: This study analyzed the interaction of the anteromedial and posterolateral portions of the anterior cruciate ligament (ACL) in resisting medial and lateral tibiofemoral compartment subluxations under multiple loading conditions. METHODS: By use of a 6-df robotic simulator, 10 human cadaveric knees were tested in 3 states: intact ACL, partial ACL (loss of either the anteromedial bundle [AMB] or posterolateral bundle [PLB]), and deficient ACL. The testing profile involved anterior-posterior translation and internal-external rotation, as well as 3 pivot-shift loading conditions with varying internal rotation torque (1- or 5-Nm) and coupled anterior force (35- or 100-N). Digitization of anatomic landmarks provided tibiofemoral compartment translations and centers of tibial rotation. RESULTS: During pivot-shift testing (100-N anterior force, 1-Nm internal rotation torque, and 7-Nm valgus), the lateral and medial compartment anterior translation increased by a mean of 2.5 ± 0.8 mm (P = .016) and 3.4 ± 2.0 mm (P = .001), respectively, on AMB sectioning and 1.3 ± 0.9 mm (P = .329) and 0.6 ± 0.7 mm (P = .544), respectively, on PLB sectioning. Higher internal rotation torque (5 Nm v 1 Nm) on pivot-shift testing reduced central and medial anterior translation after ACL sectioning. There was no change in internal rotation on AMB or PLB sectioning. During the Lachman test (100-N), AMB and PLB sectioning increased central translation by 3.6 ± 1.6 mm (P = .001) and 0.7 ± 0.6 mm (P = .498), respectively. CONCLUSIONS: Both ACL bundles function synergistically in resisting medial and lateral compartment subluxations on the Lachman and pivot-shift tests. The AMB provided more restraint to anterior tibial translation during both tests as compared with the PLB. PLB sectioning produced no statistically significant change in anterior translation on the Lachman or pivot-shift test. Neither bundle contributed to resisting internal rotation. CLINICAL RELEVANCE: An ACL graft designed to duplicate the AMB would theoretically resist medial and lateral compartment anterior subluxations under multiple loading conditions. The PLB provides a secondary restraint at low flexion angles. Neither ACL bundle resists internal tibial rotation or allows a positive pivot-shift subluxation.


Assuntos
Ligamento Cruzado Anterior/fisiopatologia , Luxação do Joelho/prevenção & controle , Luxação do Joelho/fisiopatologia , Adulto , Lesões do Ligamento Cruzado Anterior , Cadáver , Feminino , Humanos , Articulação do Joelho/fisiopatologia , Masculino , Pessoa de Meia-Idade , Rotação , Torque
5.
Arthroscopy ; 31(10): 1981-90, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26033460

RESUMO

PURPOSE: First, we aimed to investigate the ability of a single bone-patellar tendon-bone graft placed in the anatomic center of the femoral and tibial attachment sites to restore normal tibiofemoral compartment translations and tibial rotation. Second, we aimed to investigate what combination of anterior load and internal rotation torque applied during a pivot-shift test produces maximal anterior tibiofemoral subluxations. METHODS: We used a 6-df robotic simulator to test 10 fresh-frozen cadaveric specimens under anterior cruciate ligament (ACL)-intact, ACL-sectioned, and ACL-reconstructed conditions measuring anterior translations of the medial, central, and lateral tibiofemoral compartments and degrees of tibial rotation. Specimens were loaded under Lachman, anterior limit, and internal rotation conditions, as well as 3 different pivot-shift conditions. RESULTS: On ACL sectioning, compartment translations in the Lachman and 3 pivot-shift tests increased significantly and were restored to ACL-intact values after single-graft ACL reconstruction. In the pivot-shift tests, the single graft restored lateral and medial compartment translations (e.g., group 3, within 1.3 ± 0.6 mm and 0.8 ± 0.6 mm, respectively, of the ACL-intact state and internal rotation within 0.7° ± 1.2°). Anterior subluxation of the medial compartment during pivot-shift loading was reduced when internal rotation torque was increased from 1 to 5 Nm (P < .0001). CONCLUSIONS: A single-graft ACL reconstruction performed at the central femoral and tibial ACL attachment sites restored anterior-posterior translation and tibial rotation motion limits. In addition, rotational knee stability as defined by tibiofemoral compartment translations was restored under all simulated pivot-shift testing conditions. CLINICAL RELEVANCE: This study provides in vitro evidence to support the clinical use of single-graft ACL reconstructions in restoring tibiofemoral compartment translations. It also shows the advantage of describing ACL insufficiency in terms of medial and lateral compartment subluxations as compared with the common approach of describing changes in central tibial translations and rotations.


Assuntos
Reconstrução do Ligamento Cruzado Anterior/métodos , Enxertos Osso-Tendão Patelar-Osso/fisiologia , Amplitude de Movimento Articular , Ligamento Cruzado Anterior/cirurgia , Lesões do Ligamento Cruzado Anterior , Fenômenos Biomecânicos , Cadáver , Fêmur/lesões , Fêmur/cirurgia , Humanos , Luxações Articulares/etiologia , Articulação do Joelho/cirurgia , Robótica , Rotação , Tíbia/lesões , Tíbia/cirurgia , Torque
6.
Biochemistry ; 53(46): 7160-9, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25268658

RESUMO

Rhodopsin is a model system for understanding membrane protein folding. Recently, conditions that allow maximally denaturing rhodopsin without causing aggregation have been determined, opening the door to the first structural characterization of denatured states of rhodopsin by nuclear magnetic resonance (NMR) and electron paramagnetic resonance (EPR) spectroscopy. One-dimensional 1H NMR spectra confirm a progressive increase in flexibility of resonances in rhodopsin with increasing denaturant concentrations. Two-dimensional 1H-15N HSQC spectra of [15N]-α-lysine-labeled rhodopsin in which signals arise primarily from residues in the cytoplasmic (CP) domain and of [15N]-α,ε-tryptophan-labeled rhodopsin in which signals arise only from transmembrane (TM) and extracellular (EC) residues indicate qualitatively that EC and CP domains may be differentially affected by denaturation. To obtain residue-specific information, particular residues in EC and CP domains were investigated by site-directed spin labeling. EPR spectra of the spin-labeled samples indicate that the EC residues retain more rigidity in the denatured states than the CP residues. These results support the notion of residual structure in denatured states of rhodopsin.


Assuntos
Desnaturação Proteica , Rodopsina/química , Sequência de Aminoácidos , Animais , Células COS , Membrana Celular/química , Chlorocebus aethiops , Espectroscopia de Ressonância de Spin Eletrônica , Células HEK293 , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Dobramento de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
7.
Science ; 383(6683): eadj1415, 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38330136

RESUMO

Lung adenocarcinoma (LUAD) and small cell lung cancer (SCLC) are thought to originate from different epithelial cell types in the lung. Intriguingly, LUAD can histologically transform into SCLC after treatment with targeted therapies. In this study, we designed models to follow the conversion of LUAD to SCLC and found that the barrier to histological transformation converges on tolerance to Myc, which we implicate as a lineage-specific driver of the pulmonary neuroendocrine cell. Histological transformations are frequently accompanied by activation of the Akt pathway. Manipulating this pathway permitted tolerance to Myc as an oncogenic driver, producing rare, stem-like cells that transcriptionally resemble the pulmonary basal lineage. These findings suggest that histological transformation may require the plasticity inherent to the basal stem cell, enabling tolerance to previously incompatible oncogenic driver programs.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-myc , Carcinoma de Pequenas Células do Pulmão , Humanos , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/patologia , Adenocarcinoma de Pulmão/terapia , Células Epiteliais/patologia , Pulmão/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Carcinoma de Pequenas Células do Pulmão/genética , Carcinoma de Pequenas Células do Pulmão/patologia , Carcinoma de Pequenas Células do Pulmão/terapia , Oncogenes , Linhagem da Célula , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-akt/genética , Terapia de Alvo Molecular
8.
Nat Biotechnol ; 42(3): 437-447, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37563300

RESUMO

Although single-nucleotide variants (SNVs) make up the majority of cancer-associated genetic changes and have been comprehensively catalogued, little is known about their impact on tumor initiation and progression. To enable the functional interrogation of cancer-associated SNVs, we developed a mouse system for temporal and regulatable in vivo base editing. The inducible base editing (iBE) mouse carries a single expression-optimized cytosine base editor transgene under the control of a tetracycline response element and enables robust, doxycycline-dependent expression across a broad range of tissues in vivo. Combined with plasmid-based or synthetic guide RNAs, iBE drives efficient engineering of individual or multiple SNVs in intestinal, lung and pancreatic organoids. Temporal regulation of base editor activity allows controlled sequential genome editing ex vivo and in vivo, and delivery of sgRNAs directly to target tissues facilitates generation of in situ preclinical cancer models.


Assuntos
Edição de Genes , Neoplasias , Camundongos , Animais , Sistemas CRISPR-Cas/genética , RNA Guia de Sistemas CRISPR-Cas , Neoplasias/genética , Neoplasias/terapia , Pulmão
9.
Mol Cancer ; 12: 16, 2013 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-23452820

RESUMO

BACKGROUND: New therapies are urgently needed for patients with small cell lung cancer (SCLC). Chemotherapy and targeted therapies, including the Bcl-2 inhibitor ABT-737, may induce tumor cell autophagy. Autophagy can promote survival of cancer cells under stress and comprise a pathway of escape from cytotoxic therapies. METHODS: We explored the combination of ABT-737 and chloroquine, an inhibitor of autophagy, in preclinical models of SCLC. These included cell culture analyses of viability and of autophagic and apoptotic pathway induction, as well as in vivo analyses of efficacy in multiple xenograft models. RESULTS: Combination treatment of SCLC lines with ABT-737 and chloroquine decreased viability and increased caspase-3 activation over treatment with either single agent. ABT-737 induced several hallmarks of autophagy. However, knockdown of beclin-1, a key regulator of entry into autophagy, diminished the efficacy of ABT-737, suggesting either that the effects of chloroquine were nonspecific or that induction but not completion of autophagy is necessary for the combined effect of ABT-737 and chloroquine. ABT-737 and chloroquine in SCLC cell lines downregulated Mcl-1 and upregulated NOXA, both of which may promote apoptosis. Treatment of tumor-bearing mice demonstrated that chloroquine could enhance ABT-737-mediated tumor growth inhibition against NCI-H209 xenografts, but did not alter ABT-737 response in three primary patient-derived xenograft models. CONCLUSION: These data suggest that although ABT-737 can induce autophagy in SCLC, autophagic inhibition by choroquine does not markedly alter in vivo response to ABT-737 in relevant preclinical models, arguing against this as a treatment strategy for SCLC.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia , Proteína Beclina-1 , Compostos de Bifenilo/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Cloroquina/administração & dosagem , Resistencia a Medicamentos Antineoplásicos , Feminino , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Nitrofenóis/administração & dosagem , Piperazinas/administração & dosagem , RNA Interferente Pequeno/genética , Carcinoma de Pequenas Células do Pulmão/metabolismo , Carcinoma de Pequenas Células do Pulmão/patologia , Sulfonamidas/administração & dosagem , Carga Tumoral/efeitos dos fármacos , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
bioRxiv ; 2023 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-37873210

RESUMO

We recently described our initial efforts to develop a model for small cell lung cancer (SCLC) derived from human embryonic stem cells (hESCs) that were differentiated to form pulmonary neuroendocrine cells (PNECs), a putative cell of origin for neuroendocrine-positive SCLC. Although reduced expression of the tumor suppressor genes TP53 and RB1 allowed the induced PNECs to form subcutaneous growths in immune-deficient mice, the tumors did not display the aggressive characteristics of SCLC seen in human patients. Here we report that the additional, doxycycline-regulated expression of a transgene encoding wild-type or mutant cMYC protein promotes rapid growth, invasion, and metastasis of these hESC-derived cells after injection into the renal capsule. Similar to others, we find that the addition of cMYC encourages the formation of the SCLC-N subtype, marked by high levels of NEUROD1 RNA. Using paired primary and metastatic samples for RNA sequencing, we observe that the subtype of SCLC does not change upon metastatic spread and that production of NEUROD1 is maintained. We also describe histological features of these malignant, SCLC-like tumors derived from hESCs and discuss potential uses of this model in efforts to control and better understand this recalcitrant neoplasm.

11.
ChemCatChem ; 15(19)2023 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-37954549

RESUMO

Aza-substitution, the replacement of aromatic CH groups with nitrogen atoms, is an established medicinal chemistry strategy for increasing solubility, but current methods of accessing functionalized azaindoles are limited. In this work, indole-alkylating aromatic prenyltransferases (PTs) were explored as a strategy to directly functionalize azaindole-substituted analogs of natural products. For this, a series of aza-l-tryptophans (Aza-Trp) featuring N-substitution of every aromatic CH position of the indole ring and their corresponding cyclic Aza-l-Trp-l-proline dipeptides (Aza-CyWP), were synthesized as substrate mimetics for the indole-alkylating PTs FgaPT2, CdpNPT, and FtmPT1. We then demonstrated most of these substrate analogs were accepted by a PT, and the regioselectivity of each prenylation was heavily influenced by the position of the N-substitution. Remarkably, FgaPT2 was found to produce cationic N-prenylpyridinium products, representing not only a new substrate class for indole PTs but also a previously unobserved prenylation mode. The discovery that nitrogenous indole bioisosteres can be accepted by PTs thus provides access to previously unavailable chemical space in the search for bioactive indolediketopiperazine analogs.

12.
bioRxiv ; 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37904985

RESUMO

The tumor suppressor LKB1 is a serine/threonine protein kinase that is frequently mutated in human lung adenocarcinoma (LUAD). LKB1 regulates a complex signaling network that is known to control cell polarity and metabolism; however, the pathways that mediate the tumor suppressive activity of LKB1 are incompletely defined. To identify mechanisms of LKB1- mediated growth suppression we developed a spheroid-based cell culture assay to study LKB1- dependent growth. Using this assay, along with genome-wide CRISPR screens and validation with orthogonal methods, we discovered that LKB1 suppresses growth, in part, by activating the PIKFYVE lipid kinase, which promotes the internalization of wild-type EGFR. Our findings reveal a new mechanism of regulation of EGFR, which may have implications for the treatment of LKB1 -mutant LUAD.

13.
Cancer Discov ; 13(4): 1002-1025, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-36715544

RESUMO

KRAS is the most frequently mutated oncogene in human lung adenocarcinomas (hLUAD), and activating mutations frequently co-occur with loss-of-function mutations in TP53 or STK11/LKB1. However, mutation of all three genes is rarely observed in hLUAD, even though engineered comutation is highly aggressive in mouse lung adenocarcinoma (mLUAD). Here, we provide a mechanistic explanation for this difference by uncovering an evolutionary divergence in the regulation of triosephosphate isomerase (TPI1). In hLUAD, TPI1 activity is regulated via phosphorylation at Ser21 by the salt inducible kinases (SIK) in an LKB1-dependent manner, modulating flux between the completion of glycolysis and production of glycerol lipids. In mice, Ser21 of TPI1 is a Cys residue that can be oxidized to alter TPI1 activity without a need for SIKs or LKB1. Our findings suggest this metabolic flexibility is critical in rapidly growing cells with KRAS and TP53 mutations, explaining why the loss of LKB1 creates a liability in these tumors. SIGNIFICANCE: Utilizing phosphoproteomics and metabolomics in genetically engineered human cell lines and genetically engineered mouse models (GEMM), we uncover an evolutionary divergence in metabolic regulation within a clinically relevant genotype of human LUAD with therapeutic implications. Our data provide a cautionary example of the limits of GEMMs as tools to study human diseases such as cancers. This article is highlighted in the In This Issue feature, p. 799.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Triose-Fosfato Isomerase , Animais , Humanos , Camundongos , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Mutação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Triose-Fosfato Isomerase/genética , Triose-Fosfato Isomerase/metabolismo
14.
Cancer Res ; 82(22): 4261-4273, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36112789

RESUMO

Mutationally activated BRAF is detected in approximately 7% of human lung adenocarcinomas, with BRAFT1799A serving as a predictive biomarker for treatment of patients with FDA-approved inhibitors of BRAFV600E oncoprotein signaling. In genetically engineered mouse (GEM) models, expression of BRAFV600E in the lung epithelium initiates growth of benign lung tumors that, without additional genetic alterations, rarely progress to malignant lung adenocarcinoma. To identify genes that cooperate with BRAFV600E for malignant progression, we used Sleeping Beauty-mediated transposon mutagenesis, which dramatically accelerated the emergence of lethal lung cancers. Among the genes identified was Rbms3, which encodes an RNA-binding protein previously implicated as a putative tumor suppressor. Silencing of RBMS3 via CRISPR/Cas9 gene editing promoted growth of BRAFV600E lung organoids and promoted development of malignant lung cancers with a distinct micropapillary architecture in BRAFV600E and EGFRL858R GEM models. BRAFV600E/RBMS3Null lung tumors displayed elevated expression of Ctnnb1, Ccnd1, Axin2, Lgr5, and c-Myc mRNAs, suggesting that RBMS3 silencing elevates signaling through the WNT/ß-catenin signaling axis. Although RBMS3 silencing rendered BRAFV600E-driven lung tumors resistant to the effects of dabrafenib plus trametinib, the tumors were sensitive to inhibition of porcupine, an acyltransferase of WNT ligands necessary for their secretion. Analysis of The Cancer Genome Atlas patient samples revealed that chromosome 3p24, which encompasses RBMS3, is frequently lost in non-small cell lung cancer and correlates with poor prognosis. Collectively, these data reveal the role of RBMS3 as a lung cancer suppressor and suggest that RBMS3 silencing may contribute to malignant NSCLC progression. SIGNIFICANCE: Loss of RBMS3 cooperates with BRAFV600E to induce lung tumorigenesis, providing a deeper understanding of the molecular mechanisms underlying mutant BRAF-driven lung cancer and potential strategies to more effectively target this disease.


Assuntos
Adenocarcinoma de Pulmão , Carcinogênese , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Proteínas Proto-Oncogênicas B-raf , Proteínas de Ligação a RNA , Transativadores , Animais , Humanos , Camundongos , Adenocarcinoma de Pulmão/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Proliferação de Células , Pulmão/patologia , Neoplasias Pulmonares/genética , Mutagênese , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas de Ligação a RNA/genética , Transativadores/metabolismo , Via de Sinalização Wnt , Carcinogênese/genética
15.
Catalysts ; 10(11)2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33643664

RESUMO

Tryprostatin A and B are prenylated, tryptophan-containing, diketopiperazine natural products, displaying cytotoxic activity through different mechanisms of action. The presence of the 6-methoxy substituent on the indole moiety of tryprostatin A was shown to be essential for the dual inhibition of topoisomerase II and tubulin polymerization. However, the inability to perform late-stage modification of the indole ring has limited the structure-activity relationship studies of this class of natural products. Herein, we describe an efficient chemoenzymatic approach for the late-stage modification of tryprostatin B using a cyclic dipeptide N-prenyltransferase (CdpNPT) from Aspergillus fumigatus, which generates novel analogs functionalized with allylic, benzylic, heterocyclic, and diene moieties. Notably, this biocatalytic functionalizational study revealed high selectivity for the indole C6 position. Seven of the 11 structurally characterized analogs were exclusively C6-alkylated, and the remaining four contained predominant C6-regioisomers. Of the 24 accepted substrates, 10 provided >50% conversion and eight provided 20-50% conversion, with the remaining six giving <20% conversion under standard conditions. This study demonstrates that prenyltransferase-based late-stage diversification enables direct access to previously inaccessible natural product analogs.

16.
J Thorac Oncol ; 15(9): 1522-1534, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32599072

RESUMO

OBJECTIVES: SCLC represents 15% of all lung cancer diagnoses in the United States and has a particularly poor prognosis. We hypothesized that kinases regulating SCLC survival pathways represent therapeutically targetable vulnerabilities whose inhibition may improve SCLC outcome. METHODS: A short-hairpin RNA (shRNA) library targeting all human kinases was introduced in seven chemonaive patient-derived xenografts (PDX) and the cells were cultured in vitro and in vivo. On harvest, lost or depleted shRNAs were considered as regulating-cell survival pathways and deemed essential kinases. RESULTS: Unsupervised hierarchical cluster analysis of recovered shRNAs separated the PDXs into two clusters, suggesting kinase-based heterogeneity among the SCLC PDXs. A total of 23 kinases were identified as essential in two or more PDXs, with mechanistic Target of Rapamycin (mTOR) a candidate essential kinase in four. mTOR phosphorylation status correlated with PDX sensitivity to mTOR kinase inhibition, and mTOR inhibition sensitized the PDX to cisplatin and etoposide. In the PDX in which mTOR was defined as essential, mTOR inhibition caused a 43% decrease in tumor volume at 21 days (p < 0.01). Combining mTOR inhibition with cisplatin and etoposide decreased PDX tumor volume 96% compared with cisplatin and etoposide alone at 70 days (p < 0.002). Chemoresistance did not develop with the combination of mTOR inhibition and cisplatin and etoposide in mTOR-essential PDX over 105 days. The prevalence of phospho-mTOR-Ser-2448 in a tissue microarray of chemonaive SCLC was 27%, thus, identifying an important SCLC subtype that might benefit from the addition of mTOR inhibition to standard chemotherapy. CONCLUSIONS: These studies reveal that kinases can define SCLC subgroups, can identify therapeutic vulnerabilities, and can potentially be used to optimize therapeutic approaches. Significance We used functional genomics to identify kinases regulating SCLC survival. mTOR was identified as essential in a subset of PDXs. mTOR inhibition decreased PDX growth, sensitized PDX to cisplatin and etoposide, and prevented chemoresistance.


Assuntos
Neoplasias Pulmonares , Carcinoma de Pequenas Células do Pulmão , Cisplatino/farmacologia , Etoposídeo/farmacologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Sirolimo , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Carcinoma de Pequenas Células do Pulmão/genética , Serina-Treonina Quinases TOR , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Photochem Photobiol ; 95(3): 787-802, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30582615

RESUMO

The chlorophyll-derivative chlorin e6 (Ce6) identified in the retinas of deep-sea ocean fish is proposed to play a functional role in red bioluminescence detection. Fluorescence and 1 H NMR spectroscopy studies with the bovine dim-light photoreceptor, rhodopsin, indicate that Ce6 weakly binds to it with µm affinity. Absorbance spectra prove that red light sensitivity enhancement is not brought about by a shift in the absorbance maximum of rhodopsin. 19 F NMR experiments with samples where 19 F labels are either placed at the cytoplasmic binding site or incorporated as fluorinated retinal indicate that the cytoplasmic domain is highly perturbed by binding, while little to no changes are detected near the retinal. Binding of Ce6 also inhibits G-protein activation. Chemical shift changes in 1 H-15 N NMR spectroscopy of 15 N-Trp labeled bovine rhodopsin reveal that Ce6 binding perturbs the entire structure. These results provide experimental evidence that Ce6 is an allosteric modulator of rhodopsin.


Assuntos
Porfirinas/metabolismo , Rodopsina/metabolismo , Regulação Alostérica , Sequência de Aminoácidos , Animais , Bovinos , Clorofilídeos , Luz , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Conformação Proteica , Retina/metabolismo , Rodopsina/química , Espectrometria de Fluorescência
18.
Medchemcomm ; 10(8): 1465-1475, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31534661

RESUMO

Aromatic prenyltransferases from natural product biosynthetic pathways display relaxed specificity for their aromatic substrates. While a growing body of evidence suggests aromatic prenyltransferases to be more tolerant towards their alkyl-donor substrates, most studies aimed at probing their donor-substrate specificity are limited to only a small set of alkyl pyrophosphate donors, restricting their broader utility as biocatalysts for synthetic applications. Here, we assess the donor substrate specificity of an l-tryptophan C4-prenyltransferase, also known as C4-dimethylallyltryptophan synthase, FgaPT2 from Aspergillus fumigatus, using an array of 34 synthetic unnatural alkyl-pyrophosphate analogues, and demonstrate FgaPT2 can catalyze the transfer of 25 of the 34 non-native alkyl groups from their corresponding synthetic alkyl-pyrophosphate analogues at N1, C3, C4 and C5 position of tryptophan in a normal and reverse manner. The kinetic studies and regio-chemical analysis of the alkyl-l-tryptophan products suggest that the alkyl-donor transfer by FgaPT2 is a function of the stability of the carbocation and the steric factors in the active site of the enzyme. Further, to demonstrate the biocatalytic utility of FgaPT2, this study also highlights the FgaPT2-catalyzed synthesis of a small set of alkyl-diversified indolocarbazole analogues. These results reveal FgaPT2 to be more tolerant to diverse non-native alkyl-donor substrates beyond their known acceptor substrate promiscuity and set the stage for its development as a novel biocatalytic tool for the differential alkylation of natural products for drug discovery and other synthetic applications.

19.
Neurosci Lett ; 447(1): 68-72, 2008 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-18845226

RESUMO

Human cannabinoid receptors 1 (hCB(1)R) and 2 (hCB(2)R) are expressed in the CNS and couple to G(i)/G(o)-proteins. The aim of this study was to compare coupling of hCB(1)R and hCB(2)R to G(alpha)(i2)beta(1)gamma(2) in Sf9 insect cells. High-affinity agonist binding at hCB(1)R, but not at hCB(2)R, was resistant to guanine nucleotides. hCB(1)R activated G(alpha)(i2)beta(1)gamma(2) much more rapidly than hCB(2)R in the [(35)S]guanosine 5'-[gamma-thio]triphosphate ([(35)S]GTPgammaS) binding assay. Moreover, hCB(1)R exhibited a higher constitutive activity than hCB(2)R as assessed by the relative inhibitory effects of inverse agonists on [(35)S]GTPgammaS binding and steady-state high-affinity GTPase activity compared to the stimulatory effects of the hCB(1/2)R agonist CP 55,940 [(-)-cis-3-[2-hydroxy-4-(1,1-dimethylheptyl)phenyl]-trans-4-(3-hydroxypropyl)cyclohexanol]. G(alpha)(i2)beta(1)gamma(2) coupled to hCB(2)R exhibited higher GDP- and GTPgammaS-affinities than G(alpha)(i2)beta(1)gamma(2) coupled to hCB(1)R. NaCl effectively reduced constitutive activity of hCB(1)R but not of hCB(2)R. Collectively, hCB(1)R and hCB(2)R couple differentially to G(alpha)(i2)beta(1)gamma(2). Moreover, hCB(1)R exhibits higher constitutive activity than hCB(2)R. These differences point to distinct functions of hCB(1)R and hCB(2)R in the CNS.


Assuntos
Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Animais , Linhagem Celular Transformada , Cicloexanóis/farmacologia , Relação Dose-Resposta a Droga , GTP Fosfo-Hidrolases/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Humanos , Insetos , Ligação Proteica/efeitos dos fármacos , Receptor CB1 de Canabinoide/agonistas , Fatores de Tempo
20.
J Clin Invest ; 127(8): 2957-2967, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28650343

RESUMO

Seneca Valley virus (SVV) is an oncolytic picornavirus with selective tropism for neuroendocrine cancers. It has shown promise as a cancer therapeutic in preclinical studies and early-phase clinical trials. Here, we have identified anthrax toxin receptor 1 (ANTXR1) as the receptor for SVV using genome-wide loss-of-function screens. ANTXR1 is necessary for permissivity in vitro and in vivo. However, robust SVV replication requires an additional innate immune defect. We found that SVV interacts directly and specifically with ANTXR1, that this interaction is required for SVV binding to permissive cells, and that ANTXR1 expression is necessary and sufficient for infection in cell lines with decreased expression of antiviral IFN genes at baseline. Finally, we identified the region of the SVV capsid that is responsible for receptor recognition using cryoelectron microscopy of the SVV-ANTXR1-Fc complex. These studies identify ANTXR1, a class of receptor that is shared by a mammalian virus and a bacterial toxin, as the cellular receptor for SVV.


Assuntos
Proteínas do Capsídeo/química , Proteínas de Neoplasias/química , Picornaviridae , Receptores de Superfície Celular/química , Receptores Virais/química , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Microscopia Crioeletrônica , Feminino , Perfilação da Expressão Gênica , Genoma , Proteínas de Fluorescência Verde/química , Humanos , Camundongos , Camundongos Nus , Proteínas dos Microfilamentos , Terapia Viral Oncolítica , Vírus Oncolíticos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA