Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
1.
Cancer Res Commun ; 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38984877

ABSTRACT

Endothelial Notch signaling is critical for tumor angiogenesis. Notch1 blockade can interfere with tumor vessel function but causes tissue hypoxia and gastrointestinal toxicity. Notch4 is primarily expressed in endothelial cells, where it may promote angiogenesis; however, effective therapeutic targeting of Notch4 has not been successful. We developed highly specific Notch4-blocking antibodies, 6-3-A6 and humanized E7011, allowing therapeutic targeting of Notch4 to be assessed in tumor models. Notch4 was expressed on tumor endothelial cells in multiple cancer models, and endothelial expression was associated with response to E7011/6-3-A6. Anti-Notch4 treatment significantly delayed tumor growth in mouse models of breast, skin, and lung cancer. Enhanced tumor inhibition occurred when anti-Notch4 treatment was used in combination with chemotherapeutics. Endothelial transcriptomic analysis of murine breast tumors treated with 6-3-A6 identified significant changes in pathways of vascular function but caused only modest change in canonical Notch signaling. Analysis of early and late treatment timepoints revealed significant differences in vessel area and perfusion in response to anti-Notch4 treatment. We conclude that targeting Notch4 improves tumor growth control through endothelial intrinsic mechanisms.

2.
Front Mol Neurosci ; 17: 1379089, 2024.
Article in English | MEDLINE | ID: mdl-38628370

ABSTRACT

Protein phosphorylation, a key regulator of cellular processes, plays a central role in brain function and is implicated in neurological disorders. Information on protein phosphorylation is expected to be a clue for understanding various neuropsychiatric disorders and developing therapeutic strategies. Nonetheless, existing databases lack a specific focus on phosphorylation events in the brain, which are crucial for investigating the downstream pathway regulated by neurotransmitters. To overcome the gap, we have developed a web-based database named "Kinase-Associated Neural PHOspho-Signaling (KANPHOS)." This paper presents the design concept, detailed features, and a series of improvements for KANPHOS. KANPHOS is designed to support data-driven research by fulfilling three key objectives: (1) enabling the search for protein kinases and their substrates related to extracellular signals or diseases; (2) facilitating a consolidated search for information encompassing phosphorylated substrate genes, proteins, mutant mice, diseases, and more; and (3) offering integrated functionalities to support pathway and network analysis. KANPHOS is also equipped with API functionality to interact with external databases and analysis tools, enhancing its utility in data-driven investigations. Those key features represent a critical step toward unraveling the complex landscape of protein phosphorylation in the brain, with implications for elucidating the molecular mechanisms underlying neurological disorders. KANPHOS is freely accessible to all researchers at https://kanphos.jp.

3.
CPT Pharmacometrics Syst Pharmacol ; 13(6): 954-969, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38528813

ABSTRACT

Lenvatinib is a receptor tyrosine kinase (RTK) inhibitor targeting vascular endothelial growth factor (VEGF) receptors 1-3, fibroblast growth factor (FGF) receptors 1-4, platelet-derived growth factor receptor-α (PDGFRα), KIT, and RET that have been implicated in pathogenic angiogenesis, tumor growth, and cancer. The primary objective of this work was to evaluate, by establishing quantitative relationships, whether lenvatinib exposure and longitudinal serum biomarker data (VEGF, Ang-2, Tie-2, and FGF-23) are predictors for change in longitudinal tumor size which was assessed based on data from 558 patients with radioiodine-refractory differentiated thyroid cancer (RR-DTC) receiving either lenvatinib or placebo treatment. Lenvatinib PK was best described by a 3-compartment model with simultaneous first- and zero-order absorption and linear elimination from the central compartment with significant covariates (body weight, albumin <30 g/dL, ALP>ULN, RR-DTC, RCC, HCC subjects, and concomitant CYP3A inhibitors). Except for body weight, none of the covariates have any clinically meaningful effect on exposure to lenvatinib. Longitudinal biomarker measurements over time were reasonably well defined by a PK/PD model with common EC50, Emax, and a slope for disease progression for all biomarkers. Longitudinal tumor measurements over time were reasonably well defined by a tumor growth inhibition Emax model, which in addition to lenvatinib exposure, included model-predicted relative changes from baseline over time for Tie-2 and Ang-2 as having significant association with tumor response. The developed PK/PD models pave the way for dose optimization and potential prediction of clinical response.


Subject(s)
Iodine Radioisotopes , Phenylurea Compounds , Quinolines , Thyroid Neoplasms , Humans , Quinolines/pharmacokinetics , Quinolines/administration & dosage , Quinolines/therapeutic use , Quinolines/blood , Quinolines/pharmacology , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/therapeutic use , Phenylurea Compounds/blood , Phenylurea Compounds/pharmacology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/blood , Thyroid Neoplasms/pathology , Male , Female , Middle Aged , Adult , Iodine Radioisotopes/pharmacokinetics , Iodine Radioisotopes/therapeutic use , Aged , Biomarkers, Tumor/blood , Models, Biological , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/blood , Vascular Endothelial Growth Factor A/blood , Receptor, TIE-2/blood , Young Adult , Angiopoietin-2/blood
4.
Chemistry ; 30(19): e202303955, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38268122

ABSTRACT

A Fe3+ complex with N3S3-type tripod ligand, 1, reacts with O2 in CH3OH to generate formaldehyde, which has been studied structurally, spectroscopically, and electrochemically. Complex 1 crystallizes as an octahedral structure with crystallographic C3 symmetry around the metal, with Fe-N=2.2917(17) Å and Fe-S=2.3574(6) Å. UV-vis spectrum of 1 in CH3OH under Ar shows an intense band at 572 nm (ϵ 4,100 M-1cm-1), which shifts to 590 nm (ϵ 2,860 M-1cm-1) by the addition of O2, and a new peak appeared at 781 nm (ϵ 790 M-1cm-1). Such a spectral change is not observed in CH2Cl2. Cyclic voltammogram (CV) of 1 in CH2Cl2 under Ar gives reversible redox waves assigned to Fe2+/Fe3+ and Fe3+/Fe4+ couples at -1.60 V (ΔE=69 mV) and -0.53 V (ΔE=71 mV) vs Fc/Fc+, respectively. In contrast, in CH3OH, the reversible redox waves, albeit accompanied by a positive shift of the Fe2+/Fe3+ couple, are observed at -1.20 V (ΔE=85 mV) and -0.53 V (ΔE=64 mV) vs Fc/Fc+ under Ar. Interestingly, a catalytic current was observed for the CV of 1 in CH3OH in the presence of CH3ONa under Ar, when the sweep rate was slowed down.

5.
Mol Cancer Ther ; 23(2): 235-247, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37816248

ABSTRACT

E7130 is a novel anticancer agent created from total synthetic study of the natural compound norhalichondrin B. In addition to inhibiting microtubule dynamics, E7130 also ameliorates tumor-promoting aspects of the tumor microenvironment (TME) by suppressing cancer-associated fibroblasts (CAF) and promoting remodeling of tumor vasculature. Here, we demonstrate TME amelioration by E7130 using multi-imaging modalities, including multiplexed mass cytometry [cytometry by time-of-flight (CyTOF)] analysis, multiplex IHC analysis, and MRI. Experimental solid tumors characterized by large numbers of CAFs in TME were treated with E7130. E7130 suppressed LAP-TGFß1 production, a precursor of TGFß1, in CAFs but not in cancer cells; an effect that was accompanied by a reduction of circulating TGFß1 in plasma. To our best knowledge, this is the first report to show a reduction of TGFß1 production in TME. Furthermore, multiplex IHC analysis revealed reduced cellularity and increased TUNEL-positive apoptotic cells in E7130-treated xenografts. Increased microvessel density (MVD) and collagen IV (Col IV), an extracellular matrix (ECM) component associated with endothelial cells, were also observed in the TME, and plasma Col IV levels were also increased by E7130 treatment. MRI revealed increased accumulation of a contrast agent in xenografts. Moreover, diffusion-weighted MRI after E7130 treatment indicated reduction of tumor cellularity and interstitial fluid pressure. Overall, our findings strongly support the mechanism of action that E7130 alters the TME in therapeutically beneficial ways. Importantly, from a translational perspective, our data demonstrated MRI as a noninvasive biomarker to detect TME amelioration by E7130, supported by consistent changes in plasma biomarkers.


Subject(s)
Antimitotic Agents , Cancer-Associated Fibroblasts , Neoplasms, Experimental , Neoplasms , Animals , Humans , Cancer-Associated Fibroblasts/pathology , Vascular Remodeling , Tumor Microenvironment , Endothelial Cells/pathology , Neoplasms/drug therapy , Antimitotic Agents/pharmacology
6.
Expert Rev Proteomics ; 20(11): 291-298, 2023.
Article in English | MEDLINE | ID: mdl-37787112

ABSTRACT

INTRODUCTION: Since the emergence of the cholinergic hypothesis of Alzheimer's disease (AD), acetylcholine has been viewed as a mediator of learning and memory. Donepezil improves AD-associated learning deficits and memory loss by recovering brain acetylcholine levels. However, it is associated with side effects due to global activation of acetylcholine receptors. Muscarinic acetylcholine receptor M1 (M1R), a key mediator of learning and memory, has been an alternative target. The importance of targeting a specific pathway downstream of M1R has recently been recognized. Elucidating signaling pathways beyond M1R that lead to learning and memory holds important clues for AD therapeutic strategies. AREAS COVERED: This review first summarizes the role of acetylcholine in aversive learning, one of the outputs used for preliminary AD drug screening. It then describes the phosphoproteomic approach focused on identifying acetylcholine intracellular signaling pathways leading to aversive learning. Finally, the intracellular mechanism of donepezil and its effect on learning and memory is discussed. EXPERT OPINION: The elucidation of signaling pathways beyond M1R by phosphoproteomic approach offers a platform for understanding the intracellular mechanism of AD drugs and for developing AD therapeutic strategies. Clarifying the molecular mechanism that links the identified acetylcholine signaling to AD pathophysiology will advance the development of AD therapeutic strategies.


Subject(s)
Acetylcholine , Alzheimer Disease , Humans , Acetylcholine/pharmacology , Acetylcholine/therapeutic use , Receptor, Muscarinic M1/metabolism , Donepezil/pharmacology , Donepezil/therapeutic use , Signal Transduction , Alzheimer Disease/drug therapy
7.
Mol Cancer Ther ; 22(4): 499-510, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36696578

ABSTRACT

Eribulin is a microtubule dynamics inhibitor with tumor microenvironment modulation activity such as vascular remodeling activity. Here, we investigated antitumor and immunomodulatory activities of eribulin and its liposomal formulation (eribulin-LF) as monotherapies or in combination with anti-programmed death 1 (PD-1) Ab. The antitumor activity of eribulin or eribulin-LF as monotherapy or in combination with anti-PD-1 Ab was examined in a P-glycoprotein-knockout 4T1 model. Eribulin and eribulin-LF showed stronger antitumor activity in immunocompetent mice compared with immunodeficient mice, indicating that they have immunomodulatory activity that underlies its antitumor activity. Combination therapy of eribulin and eribulin-LF with anti-PD-1 Ab showed antitumor activity, and the combination activity of eribulin-LF with anti-PD-1 Ab was observed at a lower dose and longer interval of administration compared with that using eribulin. To examine the immunomodulatory activity of eribulin and eribulin-LF and its underlying mechanisms, we performed flow cytometry, IHC, and gene expression profiling. IHC and flow cytometry revealed that eribulin-LF increased microvessel density and intratumoral populations of cytotoxic T cells and natural killer cells rather than eribulin. Gene expression profiling demonstrated that eribulin-LF induces IFNγ signaling. Furthermore, IHC also showed that eribulin-LF increased infiltration of CD8-positive cells together with increased CD31-positive cells. Eribulin-LF also increased ICAM-1 expression, which is essential for lymphocyte adhesion to vascular endothelial cells. In conclusion, eribulin showed combination antitumor activity with anti-PD-1 Ab via immunomodulation due to its vascular remodeling activity, and the liposomal formulation showed improved antitumor activity over the standard formulation.


Subject(s)
Liposomes , Vascular Remodeling , Animals , Mice , Endothelial Cells , Cell Line, Tumor
8.
Molecules ; 29(1)2023 Dec 29.
Article in English | MEDLINE | ID: mdl-38202788

ABSTRACT

The iron(II) complex with cis,cis-1,3,5-tris(benzylamino)cyclohexane (Bn3CY) (1) has been synthesized and characterized, which reacted with dioxygen to form the peroxo complex 2 in acetone at -60 °C. On the basis of spectroscopic measurements for 2, it was confirmed that the peroxo complex 2 has a trans-µ-1,2 fashion. Additionally, the peroxo complex 2 was reacted with benzoate anion as a bridging agent to give a peroxo complex 3. The results of resonance Raman and 1H-NMR studies supported that the peroxo complex 3 is a cis-µ-1,2-peroxodiiron(III) complex. These spectral features were interpreted by using DFT calculations.

9.
Int J Mol Sci ; 23(19)2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36232945

ABSTRACT

Dopamine regulates emotional behaviors, including rewarding and aversive behaviors, through the mesolimbic dopaminergic pathway, which projects dopamine neurons from the ventral tegmental area to the nucleus accumbens (NAc). Protein phosphorylation is critical for intracellular signaling pathways and physiological functions, which are regulated by neurotransmitters in the brain. Previous studies have demonstrated that dopamine stimulated the phosphorylation of intracellular substrates, such as receptors, ion channels, and transcription factors, to regulate neuronal excitability and synaptic plasticity through dopamine receptors. We also established a novel database called KANPHOS that provides information on phosphorylation signals downstream of monoamines identified by our kinase substrate screening methods, including dopamine, in addition to those reported in the literature. Recent advances in proteomics techniques have enabled us to clarify the mechanisms through which dopamine controls rewarding and aversive behaviors through signal pathways in the NAc. In this review, we discuss the intracellular phosphorylation signals regulated by dopamine in these two emotional behaviors.


Subject(s)
Dopamine , Ventral Tegmental Area , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Neurotransmitter Agents/metabolism , Nucleus Accumbens/metabolism , Phosphorylation , Receptors, Dopamine/metabolism , Transcription Factors/metabolism , Ventral Tegmental Area/metabolism
10.
Cell Rep ; 40(10): 111309, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36070693

ABSTRACT

Dysfunctional dopamine signaling is implicated in various neuropsychological disorders. Previously, we reported that dopamine increases D1 receptor (D1R)-expressing medium spiny neuron (MSN) excitability and firing rates in the nucleus accumbens (NAc) via the PKA/Rap1/ERK pathway to promote reward behavior. Here, the results show that the D1R agonist, SKF81297, inhibits KCNQ-mediated currents and increases D1R-MSN firing rates in murine NAc slices, which is abolished by ERK inhibition. In vitro ERK phosphorylates KCNQ2 at Ser414 and Ser476; in vivo, KCNQ2 is phosphorylated downstream of dopamine signaling in NAc slices. Conditional deletion of Kcnq2 in D1R-MSNs reduces the inhibitory effect of SKF81297 on KCNQ channel activity, while enhancing neuronal excitability and cocaine-induced reward behavior. These effects are restored by wild-type, but not phospho-deficient KCNQ2. Hence, D1R-ERK signaling controls MSN excitability via KCNQ2 phosphorylation to regulate reward behavior, making KCNQ2 a potential therapeutical target for psychiatric diseases with a dysfunctional reward circuit.


Subject(s)
Dopamine , KCNQ2 Potassium Channel , Mental Disorders , Nerve Tissue Proteins , Animals , Dopamine/metabolism , KCNQ2 Potassium Channel/metabolism , Mental Disorders/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Phosphorylation , Receptors, Dopamine D1/metabolism , Reward
11.
Inorg Chem ; 61(35): 13956-13967, 2022 Sep 05.
Article in English | MEDLINE | ID: mdl-36000984

ABSTRACT

Geometric isomers of mononuclear ruthenium(II) complexes, distal-/proximal-[Ru(tpy)(dpda)Cl]+ (d-/p-RuCl, tpy = 2,2':6',2″-terpyridine, dpda = 2,7-bis(2-pyridyl)-1,8-diazaanthracene), were newly synthesized to comprehensively investigate the geometric and electronic structures and distinctive aspects in various reactions between isomers. The ultraviolet (UV)-visible absorption spectra of d-/p-RuCl isomers show intense bands for metal-to-ligand charge transfer (MLCT) at close wavelengths of 576 and 573 nm, respectively. However, time-dependent density functional theory (TD-DFT) calculations suggest that the MLCT transition of d-RuCl involves mainly single transitions to the π* orbital of the dpda ligand in contrast to mixing of the π* orbitals of the dpda and tpy ligands for p-RuCl. The aquation reaction (1.5 × 10-3 s-1) of p-RuCl to yield proximal-[Ru(tpy)(dpda)(OH2)]2+ (p-RuH2O) is faster than that (5.3 × 10-6 s-1) of d-RuCl in D2O/CD3OD (4:1 v/v) by three orders of magnitude, which resulted from the longer Ru-Cl bond by 0.017 Å and the distorted angle (100.2(3)°) of Cl-Ru-N (a nitrogen of dpda, being on a tpy plane) due to the steric repulsion between Cl and dpda for p-RuCl. Electrochemical measurements showed that d-RuH2O undergoes a 2-step oxidation reaction of 1H+-coupled 1e- processes of RuII-OH2/RuIII-OH and RuIII-OH/RuIV═O at pH 1-9, whereas p-RuH2O undergoes a 1-step oxidation reaction of a 2H+-coupled 2e- process of RuII-OH2/RuIV═O in the pH range of pH 1-10. The irreversible photoisomerization from d-RuH2O to p-RuH2O was observed in aqueous solution with an internal quantum yield (Φ) of 5.4 × 10-3% at 520 nm, which is lower compared with Φ = 1.1-2.1% of mononuclear Ru(II) aquo complexes with similar bidentate ligands instead of dpda by three orders of magnitude. This is possibly ascribed to the faster nonradiative decay rate from the excited 3MLCT state to the ground state for d-RuH2O due to the lower π* level of dpda ligands according to the energy-gap law: the rate decreases exponentially with the increasing energy gap.


Subject(s)
Ruthenium , Ligands , Light , Oxidation-Reduction , Protons , Ruthenium/chemistry
12.
Neurochem Res ; 47(9): 2757-2772, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35624196

ABSTRACT

The structural plasticity of dendritic spines plays a critical role in NMDA-induced long-term potentiation (LTP) in the brain. The small GTPases RhoA and Ras are considered key regulators of spine morphology and enlargement. However, the regulatory interaction between RhoA and Ras underlying NMDA-induced spine enlargement is largely unknown. In this study, we found that Rho-kinase/ROCK, an effector of RhoA, phosphorylated SynGAP1 (a synaptic Ras-GTPase activating protein) at Ser842 and increased its interaction with 14-3-3ζ, thereby activating Ras-ERK signaling in a reconstitution system in HeLa cells. We also found that the stimulation of NMDA receptor by glycine treatment for LTP induction stimulated SynGAP1 phosphorylation, Ras-ERK activation, spine enlargement and SynGAP1 delocalization from the spines in striatal neurons, and these effects were prevented by Rho-kinase inhibition. Rho-kinase-mediated phosphorylation of SynGAP1 appeared to increase its dissociation from PSD95, a postsynaptic scaffolding protein located at postsynaptic density, by forming a complex with 14-3-3ζ. These results suggest that Rho-kinase phosphorylates SynGAP1 at Ser842, thereby activating the Ras-ERK pathway for NMDA-induced morphological changes in dendritic spines.


Subject(s)
Dendritic Spines , Long-Term Potentiation , ras GTPase-Activating Proteins , 14-3-3 Proteins/metabolism , Animals , Dendritic Spines/metabolism , HeLa Cells , Hippocampus/metabolism , Humans , Long-Term Potentiation/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , N-Methylaspartate/metabolism , ras GTPase-Activating Proteins/metabolism , rhoA GTP-Binding Protein
13.
J Magn Reson Imaging ; 56(6): 1820-1831, 2022 12.
Article in English | MEDLINE | ID: mdl-35524730

ABSTRACT

BACKGROUND: Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) can provide insight into tumor perfusion. However, a method that can quantitatively measure the intra-tumor distribution of tumor voxel clusters with a distinct range of Ktrans and ve values remains insufficiently explored. HYPOTHESIS: Two-dimensional cluster analysis may quantify the distribution of a tumor voxel subregion with a distinct range of Ktrans and ve values in human breast cancer xenografts. STUDY TYPE: Prospective longitudinal study. ANIMAL MODEL: Twenty-two female athymic nude mice with MCF-7 xenograft, treated with E7130, a tumor-microenvironmental ameliorator, or saline. FIELD STRENGTH/SEQUENCE: 9.4 Tesla, turbo rapid acquisition with relaxation enhancement, and spoiled gradient-echo sequences. ASSESSMENT: We performed two-dimensional k-means clustering to identify tumor voxel clusters with a distinct range of Ktrans and ve values on Days 0, 2, and 5 after treatment, calculated the ratio of the number of tumor voxels in each cluster to the total number of tumor voxels, and measured the normalized distances defined as the ratio of the distance between each tumor voxel and the nearest tumor margin to a tumor radius. STATISTICAL TESTS: Unpaired t-tests, Dunnett's multiple comparison tests, and Chi-squared test were used. RESULTS: The largest and second largest clusters constituted 44.4% and 27.5% of all tumor voxels with cluster centroid values of Ktrans at 0.040 min-1 and 0.116 min-1 , and ve at 0.131 and 0.201, respectively. At baseline (Day 0), the average normalized distances for the largest and second largest clusters were 0.33 and 0.24, respectively. E7130-treated group showed the normalized distance of the initial largest cluster decreasing to 0.25, while that of the second largest cluster increasing to 0.31. Saline-treated group showed no change. DATA CONCLUSION: A two-dimensional cluster analysis might quantify the spatial distribution of a tumor subregion with a distinct range of Ktrans and ve values. LEVEL OF EVIDENCE: 1 TECHNICAL EFFICACY: Stage 1.


Subject(s)
Breast Neoplasms , Contrast Media , Humans , Mice , Female , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Prospective Studies , Mice, Nude , Longitudinal Studies , Magnetic Resonance Imaging/methods , Cluster Analysis
14.
Int J Mol Sci ; 24(1)2022 Dec 26.
Article in English | MEDLINE | ID: mdl-36613848

ABSTRACT

The N-methyl-D-aspartate receptor (NMDAR)-mediated structural plasticity of dendritic spines plays an important role in synaptic transmission in the brain during learning and memory formation. The Rho family of small GTPase RhoA and its downstream effector Rho-kinase/ROCK are considered as one of the major regulators of synaptic plasticity and dendritic spine formation, including long-term potentiation (LTP). However, the mechanism by which Rho-kinase regulates synaptic plasticity is not yet fully understood. Here, we found that Rho-kinase directly phosphorylated discs large MAGUK scaffold protein 2 (DLG2/PSD-93), a major postsynaptic scaffold protein that connects postsynaptic proteins with NMDARs; an ionotropic glutamate receptor, which plays a critical role in synaptic plasticity. Stimulation of striatal slices with an NMDAR agonist induced Rho-kinase-mediated phosphorylation of PSD-93 at Thr612. We also identified PSD-93-interacting proteins, including DLG4 (PSD-95), NMDARs, synaptic Ras GTPase-activating protein 1 (SynGAP1), ADAM metallopeptidase domain 22 (ADAM22), and leucine-rich glioma-inactivated 1 (LGI1), by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Among them, Rho-kinase increased the binding of PSD-93 to PSD-95 and NMDARs. Furthermore, we found that chemical-LTP induced by glycine, which activates NMDARs, increased PSD-93 phosphorylation at Thr612, spine size, and PSD-93 colocalization with PSD-95, while these events were blocked by pretreatment with a Rho-kinase inhibitor. These results indicate that Rho-kinase phosphorylates PSD-93 downstream of NMDARs, and suggest that Rho-kinase mediated phosphorylation of PSD-93 increases the association with PSD-95 and NMDARs to regulate structural synaptic plasticity.


Subject(s)
Receptors, N-Methyl-D-Aspartate , rho-Associated Kinases , rho-Associated Kinases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Chromatography, Liquid , Tandem Mass Spectrometry , Neuronal Plasticity/physiology , Synaptic Transmission/physiology , Disks Large Homolog 4 Protein/metabolism , Synapses/metabolism , Hippocampus/metabolism
15.
Cancer Res ; 82(2): 292-306, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34753772

ABSTRACT

Combination therapies consisting of immune checkpoint inhibitors plus anti-VEGF therapy show enhanced antitumor activity and are approved treatments for patients with renal cell carcinoma (RCC). The immunosuppressive roles of VEGF in the tumor microenvironment are well studied, but those of FGF/FGFR signaling remain largely unknown. Lenvatinib is a receptor tyrosine kinase inhibitor that targets both VEGFR and FGFR. Here, we examine the antitumor activity of anti-PD-1 mAb combined with either lenvatinib or axitinib, a VEGFR-selective inhibitor, in RCC. Both combination treatments showed greater antitumor activity and longer survival in mouse models versus either single agent treatment, whereas anti-PD-1 mAb plus lenvatinib had enhanced antitumor activity compared with anti-PD-1 mAb plus axitinib. Flow cytometry analysis showed that lenvatinib decreased the population of tumor-associated macrophages and increased that of IFNγ-positive CD8+ T cells. Activation of FGFR signaling inhibited the IFNγ-stimulated JAK/STAT signaling pathway and decreased expression of its target genes, including B2M, CXCL10, and PD-L1. Furthermore, inhibition of FGFR signaling by lenvatinib restored the tumor response to IFNγ stimulation in mouse and human RCC cell lines. These preclinical results reveal novel roles of tumor FGFR signaling in the regulation of cancer immunity through inhibition of the IFNγ pathway, and the inhibitory activity of lenvatinib against FGFRs likely contributes to the enhanced antitumor activity of combination treatment comprising lenvatinib plus anti-PD-1 mAb. SIGNIFICANCE: FGFR pathway activation inhibits IFNγ signaling in tumor cells, and FGFR inhibition with lenvatinib enhances antitumor immunity and the activity of anti-PD-1 antibodies.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Interferon-gamma/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Phenylurea Compounds/administration & dosage , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Quinolines/administration & dosage , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Antibodies, Monoclonal/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Female , Humans , Kidney Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Programmed Cell Death 1 Receptor/immunology , Treatment Outcome
16.
J Neurochem ; 160(3): 325-341, 2022 02.
Article in English | MEDLINE | ID: mdl-34878647

ABSTRACT

The nucleus accumbens (NAc) plays critical roles in emotional behaviors, including aversive learning. Aversive stimuli such as an electric foot shock increase acetylcholine (ACh) in the NAc, and muscarinic signaling appears to increase neuronal excitability and aversive learning. Muscarinic signaling inhibits the voltage-dependent potassium KCNQ current which regulates neuronal excitability, but the regulatory mechanism has not been fully elucidated. Phosphorylation of KCNQ2 at threonine 217 (T217) and its inhibitory effect on channel activity were predicted. However, whether and how muscarinic signaling phosphorylates KCNQ2 in vivo remains unclear. Here, we found that PKC directly phosphorylated KCNQ2 at T217 in vitro. Carbachol and a muscarinic M1 receptor (M1R) agonist facilitated KCNQ2 phosphorylation at T217 in NAc/striatum slices in a PKC-dependent manner. Systemic administration of the cholinesterase inhibitor donepezil, which is commonly used to treat dementia, and electric foot shock to mice induced the phosphorylation of KCNQ2 at T217 in the NAc, whereas phosphorylation was suppressed by an M1R antagonist. Conditional deletion of Kcnq2 in the NAc enhanced electric foot shock induced aversive learning. Our findings indicate that muscarinic signaling induces the phosphorylation of KCNQ2 at T217 via PKC activation for aversive learning.


Subject(s)
Avoidance Learning/physiology , KCNQ2 Potassium Channel/metabolism , Nerve Tissue Proteins/metabolism , Nucleus Accumbens/metabolism , Parasympathetic Nervous System/physiology , Protein Kinase C/metabolism , Receptors, Muscarinic/physiology , Animals , Carbachol/pharmacology , Cholinesterase Inhibitors/pharmacology , Donepezil/pharmacology , KCNQ2 Potassium Channel/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Nerve Tissue Proteins/genetics , Phosphorylation , Receptor, Muscarinic M2/drug effects
17.
Clin Cancer Res ; 27(17): 4848-4858, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34108184

ABSTRACT

PURPOSE: In REFLECT, lenvatinib demonstrated an effect on overall survival (OS) by confirmation of noninferiority to sorafenib in unresectable hepatocellular carcinoma. This analysis assessed correlations between serum or tissue biomarkers and efficacy outcomes from REFLECT. EXPERIMENTAL DESIGN: Serum biomarkers (VEGF, ANG2, FGF19, FGF21, and FGF23) were measured by ELISA. Gene expression in tumor tissues was measured by the nCounter PanCancer Pathways Panel. Pharmacodynamic changes in serum biomarker levels from baseline, and associations of clinical outcomes with baseline biomarker levels, were evaluated. RESULTS: Four hundred and seven patients were included in the serum analysis set (lenvatinib n = 279, sorafenib n = 128); 58 patients were included in the gene-expression analysis set (lenvatinib n = 34, sorafenib n = 24). Both treatments were associated with increases in VEGF; only lenvatinib was associated with increases in FGF19 and FGF23 at all time points. Lenvatinib-treated responders had greater increases in FGF19 and FGF23 versus nonresponders at cycle 4, day 1 (FGF19: 55.2% vs. 18.3%, P = 0.014; FGF23: 48.4% vs. 16.4%, P = 0.0022, respectively). Higher baseline VEGF, ANG2, and FGF21 correlated with shorter OS in both treatment groups. OS was longer for lenvatinib than sorafenib [median, 10.9 vs. 6.8 months, respectively; HR, 0.53; 95% confidence interval (CI), 0.33-0.85; P-interaction = 0.0397] with higher baseline FGF21. In tumor tissue biomarker analysis, VEGF/FGF-enriched groups showed improved OS with lenvatinib versus the intermediate VEGF/FGF group (HR, 0.39; 95% CI, 0.16-0.91; P = 0.0253). CONCLUSIONS: Higher baseline levels of VEGF, FGF21, and ANG2 may be prognostic for shorter OS. Higher baseline FGF21 may be predictive for longer OS with lenvatinib compared with sorafenib, but this needs confirmation.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/analysis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/mortality , Liver Neoplasms/drug therapy , Liver Neoplasms/mortality , Phenylurea Compounds/therapeutic use , Quinolines/therapeutic use , Sorafenib/therapeutic use , Biomarkers, Tumor/pharmacokinetics , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/chemistry , Humans , Liver Neoplasms/blood , Liver Neoplasms/chemistry , Predictive Value of Tests , Survival Rate
18.
Mol Cancer Res ; 19(9): 1510-1521, 2021 09.
Article in English | MEDLINE | ID: mdl-34039647

ABSTRACT

Immune checkpoint blockade (ICB) therapy has substantially improved the outcomes of patients with many types of cancers, including renal cell carcinoma (RCC). Initially studied as monotherapy, immunotherapy-based combination regimens have improved the clinical benefit achieved by ICB monotherapy and have revolutionized RCC treatment. While biomarkers like PD-L1 and tumor mutational burden (TMB) are FDA approved as biomarkers for ICB monotherapy, there are no known biomarkers for combination immunotherapies. Here, we describe the clinical outcomes and genomic determinants of response from a phase Ib/II clinical trial on patients with advanced RCC evaluating the efficacy of lenvatinib, a multi-kinase inhibitor mainly targeting VEGFR and FGFR plus pembrolizumab, an anti-PD1 immunotherapy. Concurrent treatment with lenvatinib and pembrolizumab resulted in an objective response rate of 79% (19/24) and tumor shrinkage in 96% (23/24) of patients. While tumor mutational burden (TMB) did not predict for clinical benefit, germline HLA-I diversity strongly impacted treatment efficacy. Specifically, HLA-I evolutionary divergence (HED), which measures the breadth of a patient's immunopeptidome, was associated with both improved clinical benefit and durability of response. Our results identify lenvatinib plus pembrolizumab as a highly active treatment strategy in RCC and reveal HLA-I diversity as a critical determinant of efficacy for this combination. HED also predicted better survival in a separate cohort of patients with RCC following therapy with anti-PD-1-based combination therapy. IMPLICATIONS: These findings have substantial implications for RCC therapy and for understanding immunogenetic mechanisms of efficacy and warrants further investigation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/pathology , Genetic Variation , HLA Antigens/genetics , Kidney Neoplasms/pathology , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Female , Follow-Up Studies , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Male , Middle Aged , Phenylurea Compounds/administration & dosage , Prognosis , Quinolines/administration & dosage , Survival Rate
19.
Dalton Trans ; 50(24): 8292-8296, 2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34013949

ABSTRACT

The crystal structure of an unprecedented silver complex of O-acetylsinigrin has shown chelating bonds of the sulfated thiohydroximate and an η2-bond of the ethylene moiety with Ag(i). Mechanistic studies on the formation and decomposition of the complex by the 1H NMR measurements and theoretical calculations with the DFT method indicated relevance to the glucosinolate degradation in biological systems.


Subject(s)
Coordination Complexes/chemistry , Glucosinolates/chemistry , Silver/chemistry , Biomimetic Materials/chemistry , Chelating Agents/chemistry , Glycoside Hydrolases/chemistry , Models, Molecular
20.
JCI Insight ; 6(10)2021 05 24.
Article in English | MEDLINE | ID: mdl-33857021

ABSTRACT

Antipsychotics often cause tardive dyskinesia, an adverse symptom of involuntary hyperkinetic movements. Analysis of the US Food and Drug Administration Adverse Event Reporting System and JMDC insurance claims revealed that acetaminophen prevented the dyskinesia induced by dopamine D2 receptor antagonists. In vivo experiments further showed that a 21-day treatment with haloperidol increased the number of vacuous chewing movements (VCMs) in rats, an effect that was inhibited by oral acetaminophen treatment or intracerebroventricular injection of N-(4-hydroxyphenyl)-arachidonylamide (AM404), an acetaminophen metabolite that acts as an activator of the transient receptor potential vanilloid 1 (TRPV1). In mice, haloperidol-induced VCMs were also mitigated by treatment with AM404 applied to the dorsal striatum, an effect not seen in TRPV1-deficient mice. Acetaminophen prevented the haloperidol-induced decrease in the number of c-Fos+preproenkephalin+ striatal neurons in wild-type mice but not in TRPV1-deficient mice. Finally, chemogenetic stimulation of indirect pathway medium spiny neurons in the dorsal striatum decreased haloperidol-induced VCMs. These results suggest that acetaminophen activates the indirect pathway neurons by activating TRPV1 channels via AM404.


Subject(s)
Acetaminophen , Dopamine D2 Receptor Antagonists/adverse effects , Dyskinesia, Drug-Induced , TRPV Cation Channels , Acetaminophen/pharmacology , Acetaminophen/therapeutic use , Animals , Disease Models, Animal , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/etiology , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , TRPV Cation Channels/drug effects , TRPV Cation Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...