Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(17): e2320345121, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38630723

RESUMEN

The TWIK-related acid-sensitive K+ channel 3 (TASK3) belongs to the two-pore domain (K2P) potassium channel family, which regulates cell excitability by mediating a constitutive "leak" potassium efflux in the nervous system. Extracellular acidification inhibits TASK3 channel, but the molecular mechanism by which channel inactivation is coupled to pH decrease remains unclear. Here, we report the cryo-electron microscopy structures of human TASK3 at neutral and acidic pH. Structural comparison revealed selectivity filter (SF) rearrangements upon acidification, characteristic of C-type inactivation, but with a unique structural basis. The extracellular mouth of the SF was prominently dilated and simultaneously blocked by a hydrophobic gate. His98 protonation shifted the conformational equilibrium between the conductive and C-type inactivated SF toward the latter by engaging a cation-π interaction with Trp78, consistent with molecular dynamics simulations and electrophysiological experiments. Our work illustrated how TASK3 is gated in response to extracellular pH change and implies how physiological stimuli might directly modulate the C-type gating of K2P channels.


Asunto(s)
Canales de Potasio de Dominio Poro en Tándem , Protones , Humanos , Microscopía por Crioelectrón , Simulación de Dinámica Molecular , Canales de Potasio de Dominio Poro en Tándem/metabolismo
2.
Nat Chem Biol ; 20(7): 847-856, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38167918

RESUMEN

Pharmacological activation of voltage-gated ion channels by ligands serves as the basis for therapy and mainly involves a classic gating mechanism that augments the native voltage-dependent open probability. Through structure-based virtual screening, we identified a new scaffold compound, Ebio1, serving as a potent and subtype-selective activator for the voltage-gated potassium channel KCNQ2 and featuring a new activation mechanism. Single-channel patch-clamp, cryogenic-electron microscopy and molecular dynamic simulations, along with chemical derivatives, reveal that Ebio1 engages the KCNQ2 activation by generating an extended channel gate with a larger conductance at the saturating voltage (+50 mV). This mechanism is different from the previously observed activation mechanism of ligands on voltage-gated ion channels. Ebio1 caused S6 helices from residues S303 and F305 to perform a twist-to-open movement, which was sufficient to open the KCNQ2 gate. Overall, our findings provide mechanistic insights into the activation of KCNQ2 channel by Ebio1 and lend support for KCNQ-related drug development.


Asunto(s)
Activación del Canal Iónico , Canal de Potasio KCNQ2 , Simulación de Dinámica Molecular , Canal de Potasio KCNQ2/metabolismo , Canal de Potasio KCNQ2/química , Humanos , Activación del Canal Iónico/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/química , Animales , Técnicas de Placa-Clamp , Microscopía por Crioelectrón , Células HEK293 , Relación Estructura-Actividad
3.
Proc Natl Acad Sci U S A ; 117(52): 33426-33435, 2020 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-33318209

RESUMEN

Precise genetic engineering in specific cell types within an intact organism is intriguing yet challenging, especially in a spatiotemporal manner without the interference caused by chemical inducers. Here we engineered a photoactivatable Dre recombinase based on the identification of an optimal split site and demonstrated that it efficiently regulated transgene expression in mouse tissues spatiotemporally upon blue light illumination. Moreover, through a double-floxed inverted open reading frame strategy, we developed a Cre-activated light-inducible Dre (CALID) system. Taking advantage of well-defined cell-type-specific promoters or a well-established Cre transgenic mouse strain, we demonstrated that the CALID system was able to activate endogenous reporter expression for either bulk or sparse labeling of CaMKIIα-positive excitatory neurons and parvalbumin interneurons in the brain. This flexible and tunable system could be a powerful tool for the dissection and modulation of developmental and genetic complexity in a wide range of biological systems.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Ingeniería Genética , Genoma , Luz , Recombinasas/metabolismo , Animales , Encéfalo/metabolismo , Dependovirus/metabolismo , Expresión Génica , Genes Reporteros , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Integrasas/metabolismo , Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Factores de Tiempo
4.
Anal Chem ; 91(24): 15811-15817, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31625719

RESUMEN

Split aptamers (SPAs) are a pair of oligonucleotide fragments generated by cleaving a long parent aptamer. SPAs have many compelling advantages over the parent aptamer such as sandwich target binding, optimized concise structure, and low cost. However, only a limited number of SPAs have been developed so far because the traditional theory restricts the splitting to the functionally dispensable site that many parent aptamers do not possess. In this work, the traditional mechanism and hypothesis that SPAs can also be generated by splitting the parent aptamer at the functionally essential site while still preserving the biorecognition capability are challenged. To prove the hypothesis, three SPAs with Broken initial small-molecule binding Pockets (BPSPAs) are discovered and their binding capabilities are validated both in the wet lab and in silico. An allosteric binding mechanism of BPSPAs, in which a new binding pocket is formed upon the target binding, is revealed by all-atom microsecond-scale molecular dynamics simulations. Our work highlights the important role of MD simulations in predicting the ligand binding potency with functional nucleic acids at the molecular level. The findings will greatly promote discovery of new SPAs and their applications in molecular sensing in many fields.

5.
Exp Cell Res ; 363(2): 235-242, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29331389

RESUMEN

Increasing evidence has suggested that Capn4 is upregulated and functions as a potential tumor promoter in several human cancer types. However, the potential biological roles and regulatory mechanisms of Capn4 in colorectal cancer (CRC) remains unclear. Here, we found that Capn4 expression was elevated in CRC tissues than adjacent noncancerous tissues. Additionally, we also found that overexpression of Capn4 is significantly correlated with tumor progression and poor survival in CRC patients. Furthermore, our experimental data revealed that increased expression of Capn4 was observed in CRC cell lines and ectopic expression of Capn4 significantly enhanced in vitro cell proliferation, whereas knockdown of Capn4 suppressed CRC cells growth in vitro and in vivo. Moreover, our results indicate that Capn4 promotes cell proliferation by increasing MAPK7 expression, which has been reported to control the proliferation of many cancers. Mechanistically, Capn4 upregulates MAPK7 expression through activation of the Wnt/ß-Catenin pathway in CRC cells. Therefore, we identified a tumorigenic role of Capn4 in CRC and suggested a potential therapeutic target for CRC patients.


Asunto(s)
Calpaína/metabolismo , Proliferación Celular/fisiología , Neoplasias del Colon/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo
6.
Biochemistry ; 57(32): 4767-4775, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-29999306

RESUMEN

G protein-coupled receptors (GPCRs) make up the largest family of drug targets. The second extracellular loop (ECL2) and extracellular end of the third transmembrane helix (TM3) are basic structural elements of the GPCR ligand binding site. Currently, the disulfide bond between the two conserved cysteines in the ECL2 and TM3 is considered to be a basic GPCR structural feature. This disulfide bond has a significant effect on receptor dynamics and ligand binding. Here, molecular dynamics simulations and experimental results show that the two cysteines are distant from one another in the highest-population conformational state of ligand-free class A GPCRs and do not form a disulfide bond, indicating that the dynamics of the GPCR extracellular side are different from our conventional understanding. These surprising dynamics should have important effects on the drug binding process. On the basis of the two distinct ligand-free states, we suggest two kinetic processes for binding of ligands to GPCRs. These results challenge our commonly held beliefs regarding both GPCR structural features and ligand binding.


Asunto(s)
Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Disulfuros/química , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Estructura Secundaria de Proteína
7.
Eur Biophys J ; 45(6): 511-21, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27038819

RESUMEN

Caveolin-1 is the principle membrane protein of caveolae and plays an important role in various cellular processes. The protein contains two helices (H1 and H2) connected by a three-residue break. Although caveolin-1 is assumed to adopt a U-shaped conformation in the transmembrane domain, with both the N-terminus and C-terminus exposed to the cytoplasm, the structure and dynamics of caveolin-1 in membranes are still unclear. Here, we performed six molecular dynamics simulations to characterize the structure and dynamics of caveolin-1 (residues D82-S136; Cav182-136) in a caveolae-mimicking asymmetric lipid bilayer. The simulations reveal that the structure of the caveolin scaffolding domain of caveolin-1 is dynamic, as it could be either fully helical or partly unstructured. Cav182-136 inserts into the inner leaflet of the asymmetric lipid bilayer with a stable U-shaped conformation and orients almost vertical to the bilayer surface. The simulations also provide new insights into the effects of caveolin-1 on the morphology of caveolae and the possible interacting site of cholesterol on caveolin-1.


Asunto(s)
Caveolina 1/química , Caveolina 1/metabolismo , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Simulación de Dinámica Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Estructura Secundaria de Proteína
8.
Proc Natl Acad Sci U S A ; 110(50): 20093-8, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24277843

RESUMEN

The S4 segment and the S4-S5 linker of voltage-gated potassium (Kv) channels are crucial for voltage sensing. Previous studies on the Shaker and Kv1.2 channels have shown that phosphatidylinositol-4,5-bisphosphate (PIP2) exerts opposing effects on Kv channels, up-regulating the current amplitude, while decreasing the voltage sensitivity. Interactions between PIP2 and the S4 segment or the S4-S5 linker in the closed state have been highlighted to explain the effects of PIP2 on voltage sensitivity. Here, we show that PIP2 preferentially interacts with the S4-S5 linker in the open-state KCNQ2 (Kv7.2) channel, whereas it contacts the S2-S3 loop in the closed state. These interactions are different from the PIP2-Shaker and PIP2-Kv1.2 interactions. Consistently, PIP2 exerts different effects on KCNQ2 relative to the Shaker and Kv1.2 channels; PIP2 up-regulates both the current amplitude and voltage sensitivity of the KCNQ2 channel. Disruption of the interaction of PIP2 with the S4-S5 linker by a single mutation decreases the voltage sensitivity and current amplitude, whereas disruption of the interaction with the S2-S3 loop does not alter voltage sensitivity. These results provide insight into the mechanism of PIP2 action on KCNQ channels. In the closed state, PIP2 is anchored at the S2-S3 loop; upon channel activation, PIP2 interacts with the S4-S5 linker and is involved in channel gating.


Asunto(s)
Activación del Canal Iónico/fisiología , Canal de Potasio KCNQ2/química , Canal de Potasio KCNQ2/metabolismo , Modelos Moleculares , Fosfatidilinositol 4,5-Difosfato/metabolismo , Conformación Proteica , Animales , Células CHO , Cricetinae , Cricetulus , ADN Complementario/genética , Activación del Canal Iónico/genética , Canal de Potasio KCNQ2/genética , Simulación de Dinámica Molecular , Mutagénesis , Técnicas de Placa-Clamp , Xenopus laevis
9.
Biochem Biophys Res Commun ; 468(1-2): 125-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26529547

RESUMEN

Previous molecular dynamics (MD) simulations of ion-lipid interactions have focused on pure phospholipid bilayers. Many functional microdomains in membranes have a complex composition of cholesterol and phospholipids. Here, we reveal the distinctiveness of the interactions and the effects of the ions on a cholesterol-rich bilayer by performing MD simulations of a cholesterol-rich bilayer with a Na(+)/K(+) mixture or a Na(+)/K(+)/Ca(2+)/Mg(2+) mixture. The simulations reveal that Ca(2+) maintains its dominant role in the interaction with the cholesterol-rich bilayer, but the binding affinity of Mg(2+) to the cholesterol-rich bilayer is even weaker than the affinities of Na(+) and K(+), whereas its interaction with pure phospholipid bilayers is strong and is only slightly weaker than that of Ca(2+). Additionally, it was found that the presence of additional divalent cations induces the headgroups of phospholipids to be more perpendicular to the membrane surface, reducing the lateral movement of lipids and slightly altering the ordering and packing of the cholesterol-rich bilayer, different from divalent cations, which strongly influence that ordering and packing of pure phospholipid bilayers. Therefore, this study indicates that cholesterol in the membrane could affect the interactions between membrane and cations. The findings could be helpful in understanding the biological processes relevant to regulation of cations in cholesterol-rich regions.


Asunto(s)
Calcio/metabolismo , Colesterol/metabolismo , Membrana Dobles de Lípidos/metabolismo , Magnesio/metabolismo , Fosfolípidos/metabolismo , Calcio/química , Cationes Bivalentes/química , Cationes Bivalentes/metabolismo , Colesterol/química , Membrana Dobles de Lípidos/química , Magnesio/química , Simulación de Dinámica Molecular , Fosfolípidos/química
10.
J Pharmacol Exp Ther ; 351(3): 596-604, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25232191

RESUMEN

Human ether-a-go-go-related gene (hERG) and KCNQ channels are two classes of voltage-gated potassium channels. Specific mutations have been identified that are causal for type II long QT (LQT2) syndrome, neonatal epilepsy, and benign familial neonatal convulsions. Increasing evidence from clinical studies suggests that LQT2 and epilepsy coexist in some patients. Therefore, an integral approach to investigating and treating the two diseases is likely more effective. In the current study, we found that NS1643 [1,3-bis-(2-hydroxy-5-trifluoromethyl-phenyl)-urea], a previously reported hERG activator, is also an activator of KCNQ channels. It potentiates the neuronal KCNQ2, KCNQ4, and KCNQ2/Q3 channels, but not the cardiac KCNQ1. The effects of NS1643 on the KCNQ2 channel include left shifting of voltage for reaching 50% of the maximum conductance and slowing of deactivation. Analysis of the dose-response curve of NS1643 revealed an EC50 value of 2.44 ± 0.25 µM. A hydrophobic phenylalanine (F137) located at the middle region of the voltage-sensing domain was identified as critical for NS1643 activity on KCNQ2. When testing NS1643 effects in rescuing LQT2 hERG mutants and the KCNQ2 BFNC mutants, we found it is particularly efficacious in some cases. Considering the substantial relationship between LQT2 and epilepsy, these findings reveal that NS1643 is a useful compound to elucidate the causal connection of LQT2 and epilepsy. More generally, this may provide a strategy in the development of therapeutics for LQT2 and epilepsy.


Asunto(s)
Cresoles/metabolismo , Cresoles/farmacología , Epilepsia/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Canal de Potasio KCNQ2/metabolismo , Compuestos de Fenilurea/metabolismo , Compuestos de Fenilurea/farmacología , Animales , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Canales de Potasio Éter-A-Go-Go/agonistas , Humanos , Canal de Potasio KCNQ2/agonistas , Estructura Secundaria de Proteína
11.
Sci China Life Sci ; 67(5): 986-995, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38319473

RESUMEN

The adenosine subfamily G protein-coupled receptors A2AR and A2BR have been identified as promising cancer immunotherapy candidates. One of the A2AR/A2BR dual antagonists, AB928, has progressed to a phase II clinical trial to treat rectal cancer. However, the precise mechanism underlying its dual-antagonistic properties remains elusive. Herein, we report crystal structures of the A2AR complexed with AB928 and a selective A2AR antagonist 2-118. The structures revealed a common binding mode on A2AR, wherein the ligands established extensive interactions with residues from the orthosteric and secondary pockets. In contrast, the cAMP assay and A2AR and A2BR molecular dynamics simulations indicated that the ligands adopted distinct binding modes on A2BR. Detailed analysis of their chemical structures suggested that AB928 readily adapted to the A2BR pocket, while 2-118 did not due to intrinsic differences. This disparity potentially accounted for the difference in inhibitory efficacy between A2BR and A2AR. This study serves as a valuable structural template for the future development of selective or dual inhibitors targeting A2AR/A2BR for cancer therapy.


Asunto(s)
Antagonistas del Receptor de Adenosina A2 , Simulación de Dinámica Molecular , Receptor de Adenosina A2A , Humanos , Antagonistas del Receptor de Adenosina A2/química , Receptor de Adenosina A2A/química , Receptor de Adenosina A2A/metabolismo , Sitios de Unión , Ligandos , Cristalografía por Rayos X , Unión Proteica , Receptor de Adenosina A2B/metabolismo , Receptor de Adenosina A2B/química
12.
ACS Cent Sci ; 10(3): 579-594, 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38559310

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) is an attractive cancer therapeutic target. Unfortunately, targeting STAT3 with small molecules has proven to be very challenging, and for full activation of STAT3, the cooperative phosphorylation of both tyrosine 705 (Tyr705) and serine 727 (Ser727) is needed. Further, a selective inhibitor of STAT3 dual phosphorylation has not been developed. Here, we identified a low nanomolar potency and highly selective small-molecule STAT3 inhibitor that simultaneously inhibits both STAT3 Tyr705 and Ser727 phosphorylation. YY002 potently inhibited STAT3-dependent tumor cell growth in vitro and achieved potent suppression of tumor growth and metastasis in vivo. More importantly, YY002 exhibited favorable pharmacokinetics, an acceptable safety profile, and superior antitumor efficacy compared to BBI608 (STAT3 inhibitor that has advanced into phase III trials). For the mechanism, YY002 is selectively bound to the STAT3 Src Homology 2 (SH2) domain over other STAT members, which strongly suppressed STAT3 nuclear and mitochondrial functions in STAT3-dependent cells. Collectively, this study suggests the potential of small-molecule STAT3 inhibitors as possible anticancer therapeutic agents.

13.
Environ Sci Technol ; 47(20): 11802-9, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24044724

RESUMEN

Hydroxylated and methoxylated polybrominated diphenyl ethers (HO-/MeO-PBDEs) have received increasing attention for their potential endocrine disrupting activities and widely environmental distribution. However, little information is available for the anti-androgenic activities, and the molecular mechanism of interactions with androgen receptor (AR) is not fully understood. In the present study, cell line assay and computational simulation were integrated to systematically explore the molecular mechanism of interactions between chemicals and AR. The metabolites with similar molecular structures exhibited different anti-androgenic activity while none of them showed androgenic activity. According to the multisystem molecular dynamics simulation, minute differences in the structure of ligands induced dramatic different conformational transition of AR-ligand binding domain (LBD). The Helix12 (H12) component of active ligands occupied AR-LBD could become stable, but this component continued to fluctuate in inactive ligands occupied AR-LBD. Settling time and reposition of H12 obtained in dynamics process are important factors governing anti-androgenic activities. The related settling times were characteristic of anti-androgenic potencies of the tested chemicals. Overall, in our study, the stable reposition of H12 is characterized as a computational mark for identifying AR antagonists from PBDE metabolites, or even other various environmental pollutants.


Asunto(s)
Éteres Difenilos Halogenados/farmacología , Simulación de Dinámica Molecular , Receptores Androgénicos/química , Receptores Androgénicos/metabolismo , Antagonistas de Receptores Androgénicos/farmacología , Línea Celular , Flutamida/farmacología , Genes Reporteros , Humanos , Hidroxilación , Ligandos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Factores de Tiempo
14.
J Mol Biol ; 435(6): 167979, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36716818

RESUMEN

Adhesion G protein-coupled receptors (aGPCRs) play a pivotal role in human immune responses, cellular communication, organ development, and other processes. GPR110 belongs to the aGPCR subfamily VI and was initially identified as an oncogene involved in lung and prostate cancers. GPR110 contains tandem adhesion domains at the extracellular region that mediate inter-cellular signaling. However, the structural organization and signaling mechanism for these tandem domains remain unclear. Here, we report the crystal structure of a GPR110 fragment composing the SEA, HormR, and GAIN domains at 2.9 Å resolution. The structure together with MD simulations reveal rigid connections between these domains that are stabilized by complementary interfaces. Strikingly, we found N-linked carbohydrates attached to N389 of the GAIN domain form extensive contacts with the preceding HormR domain. These interactions appear to be critical for folding, as removal of the glycosylation site greatly decreases expression of the GPR110 extracellular fragment. We further demonstrate that the ligand synaptamide fits well within the hydrophobic pocket occupied by the Stachel peptide in the rest state. This suggests that the agonist may function by removing the Stachel peptide which in turn redocks to the orthosteric pocket for receptor activation. Taken together, our structural findings and analyses provide novel insights into the activation mechanism for aGPCRs.


Asunto(s)
Proteínas Oncogénicas , Receptores Acoplados a Proteínas G , Humanos , Masculino , Ligandos , Péptidos/química , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Transducción de Señal , Proteínas Oncogénicas/agonistas , Proteínas Oncogénicas/química , Etanolaminas/química , Dominios Proteicos , Cristalografía por Rayos X , Simulación de Dinámica Molecular
15.
Cell Rep ; 42(8): 112926, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37543949

RESUMEN

Volume-regulated anion channels (VRACs) are hexamers of LRRC8 proteins that are crucial for cell volume regulation. N termini (NTs) of the obligatory LRRC8A subunit modulate VRACs activation and ion selectivity, but the underlying mechanisms remain poorly understood. Here, we report a 2.8-Å cryo-electron microscopy structure of human LRRC8A that displays well-resolved NTs. Amino-terminal halves of NTs fold back into the pore and constrict the permeation path, thereby determining ion selectivity together with an extracellular selectivity filter with which it works in series. They also interact with pore-surrounding helices and support their compact arrangement. The C-terminal halves of NTs interact with intracellular loops that are crucial for channel activation. Molecular dynamics simulations indicate that low ionic strength increases NT mobility and expands the radial distance between pore-surrounding helices. Our work suggests an unusual pore architecture with two selectivity filters in series and a mechanism for VRAC activation by cell swelling.


Asunto(s)
Proteínas de la Membrana , Humanos , Microscopía por Crioelectrón , Proteínas de la Membrana/metabolismo , Aniones/metabolismo , Tamaño de la Célula , Concentración Osmolar
16.
J Med Chem ; 66(9): 6218-6238, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-36880691

RESUMEN

Nowadays, small-molecule drugs have become an indispensable part of tumor immunotherapy. Accumulating evidence has indicated that specifically blocking PGE2/EP4 signaling to induce robust antitumor immune response represents an attractive immunotherapy strategy. Herein, a 2H-indazole-3-carboxamide containing compound 1 was identified as a EP4 antagonist hit by screening our in-house small-molecule library. Systematic structure-activity relationship exploration leads to the discovery of compound 14, which displayed single-nanomolar EP4 antagonistic activity in a panel of cell functional assays, high subtype selectivity, and favorable drug-like profiles. Moreover, compound 14 profoundly inhibited the up-regulation of multiple immunosuppression-related genes in macrophages. Oral administration of compound 14, either as monotherapy or in combination with an anti-PD-1 antibody, significantly impaired tumor growth via enhancing cytotoxic CD8+ T cell-mediated antitumor immunity in a syngeneic colon cancer model. Thus, these results demonstrate the potential of compound 14 as a candidate for developing novel EP4 antagonists for tumor immunotherapy.


Asunto(s)
Neoplasias del Colon , Indazoles , Subtipo EP4 de Receptores de Prostaglandina E , Humanos , Neoplasias del Colon/patología , Inmunoterapia , Prostaglandinas , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Transducción de Señal , Indazoles/química , Indazoles/farmacología
17.
Clin Cancer Res ; 29(4): 815-830, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36374556

RESUMEN

PURPOSE: Pancreatic cancer is the worst prognosis among all human cancers, and novel effective treatments are urgently needed. Signal transducer and activator of transcription 3 (STAT3) has been demonstrated as a promising target for pancreatic cancer. Meanwhile, selectively targeted STAT3 with small molecule remains been challenging. EXPERIMENTAL DESIGN: To specifically identify STAT3 inhibitors, more than 1.3 million compounds were screened by structure-based virtual screening and confirmed with the direct binding assay. The amino acid residues that WB436B bound to were verified by induced-fit molecular docking simulation, RosettaLigand computations, and site-directed mutagenesis. On-target effects of WB436B were examined by microscale thermophoresis, surface plasmon resonance, in vitro kinase assay, RNA sequencing, and selective cell growth inhibition assessment. In vivo studies were performed in four animal models to evaluate effects of WB436B on tumor growth and metastasis. Kaplan-Meier analyses were used to assess survival. RESULTS: WB436B selectively bound to STAT3 over other STAT families protein, and in vitro antitumor activities were improved by 10 to 1,000 fold than the representative STAT3 inhibitors. WB436B selectively inhibits STAT3-Tyr705 phosphorylation, STAT3 target gene expression, and the viability of STAT3-dependent pancreatic cancer cells. WB436B significantly suppresses tumor growth and metastasis in vivo and prolongs survival of tumor-bearing mice. Mechanistic studies showed that WB436B have unique binding sites located in STAT3 Src homology 2 domain. CONCLUSIONS: Our work presents the first-in-class selective STAT3 inhibitor WB436B as a potential therapeutic candidate for the treatment of pancreatic cancer.


Asunto(s)
Neoplasias Pancreáticas , Factor de Transcripción STAT3 , Humanos , Animales , Ratones , Factor de Transcripción STAT3/metabolismo , Simulación del Acoplamiento Molecular , Línea Celular Tumoral , Dominios Homologos src , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fosforilación , Proliferación Celular , Apoptosis , Neoplasias Pancreáticas
18.
J Mol Cell Biol ; 14(1)2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35022758

RESUMEN

Two-pore domain potassium (K2P) channels gate primarily within the selectivity filter, termed 'C-type' gating. Due to the lack of structural insights into the nonconductive (closed) state, 'C-type' gating mechanisms remain elusive. Here, molecular dynamics (MD) simulations on TREK-1, a K2P channel, revealed that M4 helix movements induce filter closing in a novel 'deeper-down' structure that represents a 'C-type' closed state. The 'down' structure does not represent the closed state as previously proposed and instead acts as an intermediate state in gating. The study identified the allosteric 'seesaw' mechanism of M4 helix movements in modulating filter closing. Finally, guided by this recognition of K2P gating mechanisms, MD simulations revealed that gain-of-function mutations and small-molecule activators activate TREK-1 by perturbing state transitions from open to closed states. Together, we reveal a 'C-type' closed state and provide mechanical insights into gating procedures and allosteric regulations for K2P channels.


Asunto(s)
Activación del Canal Iónico , Canales de Potasio de Dominio Poro en Tándem , Simulación de Dinámica Molecular , Canales de Potasio de Dominio Poro en Tándem/química , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo
19.
J Med Chem ; 65(15): 10285-10299, 2022 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-35878013

RESUMEN

Under the known pharmacological activation mechanisms, activators allosterically or directly open potassium channel gates. However, herein, molecular dynamics simulations on TREK-1, a member of the channel class gated at the filter, suggested that negatively charged activators act with a gate-independent mechanism where compounds increase currents by promoting ions passing through the central cavity. Then, based on studies of KCNQ2, we uncovered that this noncanonical activation mechanism is shared by the other channel class gated at the helix-bundle crossing. Rational drug design found a novel KCNQ2 agonist, CLE030, which stably binds to the central cavity. Functional analysis, molecular dynamics simulations, and calculations of the potential of mean force revealed that the carbonyl oxygen of CLE030 influences permeant ions in the central cavity to contribute to its activation effects. Together, this study discovered a ligand-to-ion activation mechanism for channels that bypasses their gates and thus is conserved across subfamilies with different gates.


Asunto(s)
Activación del Canal Iónico , Simulación de Dinámica Molecular , Iones/farmacología
20.
J Med Chem ; 65(11): 7896-7917, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35640059

RESUMEN

Cancer cells can effectively suppress the natural immune response in humans, and prostaglandin E2 (PGE2) is a key mediator in the development of tumor cell resistance to immunotherapy. As a major contributor to PGE2-elicited immunosuppressive activity, the EP4 receptor promotes tumor development and progression in the tumor microenvironment, and the development of selective and potent EP4 receptor antagonists should have promising potential for tumor immunotherapy. Aiming at improving the drug-like properties, a series of 4,7-dihydro-5H-thieno[2,3-c]pyran derivatives were designed and synthesized through a scaffold hopping strategy. The most promising compound 47 exhibited good EP4 antagonistic activity and excellent subtype selectivity, as well as favorable drug-like properties. It effectively suppressed the expression of multiple immunosuppression-related genes in macrophages. Meanwhile, oral administration of compound 47, alone or in combination with anti-PD-1 antibody, significantly enhanced the antitumor immune response and inhibited tumor growth in the mouse CT26 colon carcinoma model.


Asunto(s)
Neoplasias del Colon , Subtipo EP4 de Receptores de Prostaglandina E , Animales , Neoplasias del Colon/patología , Dinoprostona , Inmunoterapia , Ratones , Subtipo EP2 de Receptores de Prostaglandina E , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA