Abstract INTRODUCTION AND HYPOTHESIS Mesenchymal cells known as myo-/fibroblasts (MFs) are critical immunosuppressors under gut mucosal homeostasis. Expression of immune checkpoint PD-L1 by MFs plays a key role in the control of T cell inflammatory responses. In Crohn’s disease (CD), MFs switch their activity from immunosuppressive to pro-inflammatory, where they are also known as Inflammatory Fibroblasts. However, the mechanisms responsible for these pathological changes in MF activity are unknown. Map-kinase-activated protein kinase 2 (MK2) is a major regulator of inflammation in the gut. MK2 is downstream of p38 signaling, it evokes a sub-pathway that directly regulates the production of key inflammatory cytokines implicated in CD (such as TNF-α, IL-1, and IL-6). Thus, we hypothesized that activation of MK2 signaling is critical to the pathological changes in MFs during the immunopathogenesis of CD. METHODS Human normal and CD tissues and derived MFs, as well as animal models relevant to CD, were used in this study. MF signaling/activity was analyzed using RNAseq, qRT-PCR, western blot (WB), cytokine/chemokine multiplex arrays, and confocal microscopy. RESULTS In situ analysis demonstrated an increase in MK2 activity within the inflamed compared to the non-inflamed CD and healthy control intestinal tissues, which was confirmed by WB and multiplex signaling array analysis. In situ increase in MK2 activity in CD intestinal mucosa was greatly associated with mesenchymal stromal cells that bear a “myofibroblast” phenotype (positive for α-SMA expression). An increase in MK2 activity was also observed in primary MF cultures isolated from CD (CD-MFs) when compared to normal (N-) MFs. MK2 activity within CD-MFs was also associated with a significant decrease in the expression of the immunosuppressive checkpoint PD-L1 and an increase in the expression of inflammatory CCL2 and IL-6. Inhibition of MK2 activity within CD-MFs through using the MK2-specific inhibitor PF-3644022 (10 μM) reversed the inflammatory activity of MFs. Remarkably, we observed a differential role of p38 and MK2 in the regulation of PD-L1 expression in MFs: while p38 was required for basal expression of PD-L1, activation of MK2 downregulates PD-L1 expression. These data indicate a unique role of MK2 activation in pathological reprogramming of MFs in CD. Use of MK2 inhibitor in a therapeutic modality in chronic DSS and IL-10 KO murine models of CD also significantly reduce MF-linked inflammatory responses in vivo. CONCLUSION Our data suggest that an increase in MK2 activity in CD is critical to the reprogramming of the MF from immunosuppressive toward pathological Inflammatory fibroblasts. Targeting MK2 activity within MFs could be a desirable strategy for improving the efficacy of current IBD therapeutic approaches.
An entry from the Cambridge Structural Database, the world’s repository for small molecule crystal structures. The entry contains experimental data from a crystal diffraction study. The deposited dataset for this entry is freely available from the CCDC and typically includes 3D coordinates, cell parameters, space group, experimental conditions and quality measures.
Abstract Background: Pre-B cell Acute lymphoblastic leukemia (B ALL) high risk (HR) subgroups continue to result in significant mortality and morbidity of pediatric oncology patients. T cell ALL is higher risk and therapy has made less improvement than B ALL with a higher rate of significant poor outcomes. Novel treatment strategies are required to overcome chemotherapy resistance and improve mortality/morbidity for HR B ALL. Leonurine is a bioactive alkaloid that is naturally occurring only in Herbra Leonuri which has been used in traditional herbal medicine. Traditionally, it has been used for menstrual disorders. Research has further described its ability to scavenge oxygen free radicals, anticoagulation properties, and other anti-inflammatory properties[1]. There are investigations in its role for myocardial infarction, stroke, chronic kidney disease, and other inflammatory disorders[2]. Our research data that suggests that leonurine and its derivatives have antileukemic effects.Methods/Results: We analyzed multiple derivatives of leonurine and selected a potent candidate based on cell viability assays use for further testing designated as investigational leonurine derivative (ILD). WST1 proliferation studies comparing ILD to vehicle were performed in cell lines Nalm6, 697, Molt4, CEM, and JM1 at multiple time points. The half maximal inhibitory concentration (IC50) values was variable depending on the treatment time and cell line although all values were consistently between 1.2-4.4 uM. Apoptosis activity was determined by flow cytometry Anexin/7AAD assays showed increased apoptosis in cell lines treated with ILD for Nalm6, Molt4, and 697 cell lines. Caspase 3/7 activity was increased in cells treated with ILD when compared to vehicle treatment. DNA damage assays were performed which revealed only an increased frequency in single strand breaks and not double strand breaks. Western blot was performed to determine levels of PI3K, p-AKT, BCL2/BCL-XL and caspases. The blots suggest that apoptosis may be a result of increased activation of PI3K/AKT signaling. We performed RNAseq on cells treated with ILD at the 24-hour time point and present gene ontology data for this analysis. Nalm6, 697, and Molt4 cell lines expressing GFP and Luciferase were injected into NRG mice as means for in vivo pharmacologic testing. NRG mice injected with these cell lines were treated with ILD five days a week for a total of 3 weeks. Nalm 6 leukemia cells showed minimal differences between treatment and control groups.Conclusion: In summary, leonurine derivatives have a promising impact on apoptosis and cell survival. Further investigation into mechanisms and pharmacokinetics/dynamics will be more revealing. Study of leonurine derivatives will result in further translational and therapeutic applications. Citation Format: Joseph W. Schramm, Melanie Ehudin, Bing He, Chingakham Singh, Daniel Bogush, Jeremy Hengst, Diwakar Bastihalli Tukaramrao, Arati Sharma, Dhimant Desai, Sinisa Dovat. Leonurine derivatives as a potential novel therapeutic approach to acute lymphoblastic leukemias (ALL). [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3833.
Supplementing chemotherapy and radiotherapy with selenium has been shown to have benefits against various cancers. This approach has also been shown to alleviate the side effects associated with standard cancer therapies and improve the quality of life in patients. In addition, selenium levels in patients have been correlated with various cancers and have served as a diagnostic marker to track the efficiency of treatments or to determine whether these selenium levels cause or are a result of the disease. This concise review presents a survey of the selenium-based literature, with a focus on hematological malignancies, to demonstrate the significant impact of selenium in different cancers. The anti-cancer mechanisms and signaling pathways regulated by selenium, which impart its efficacious properties, are discussed. An outlook into the relationship between selenium and cancer is highlighted to guide future cancer therapy development.
High-valent Fe IV =O species are common intermediates in biological and artificial catalysts. Heme and nonheme S=1 Fe IV =O sites have been synthesized and studied for decades but little quantitative experimental comparison of their electronic structures has been available, due to the lack of direct methods focused on the iron. This study allows a rigorous determination of the electronic structure of a nonheme Fe IV =O center and its comparison to an Fe IV =O heme site using 1s2p resonant inelastic X-ray scattering (RIXS) and Fe L-edge X-ray absorption spectroscopy (XAS). Further, variable temperature magnetic circular dichroism (VT-MCD) of the ligand field transitions, combined with nuclear resonance vibrational spectroscopy of the two S=1 Fe IV =O systems show that the equatorial ligand field decreases from a nonheme to a heme Fe IV =O site. Alternatively, RIXS and Fe L-edge XAS combined with MCD show that the Fe d π orbitals are unperturbed in the Fe IV =O heme relative to the nonheme site because the strong axial Fe-O bond uncouples the Fe d π orbitals from the porphyrin π -system. As a consequence, the thermodynamics and kinetics of the H-atom abstraction reactions are actually very similar for heme compound II and nonheme Fe IV =O active sites.
A superoxide-bridged dicopper(II) complex, [CuII2(XYLO)(O2•-)]2+ (1) (XYLO = binucleating m-xylyl derivative with a bridging phenolate ligand donor and two bis(2-{2-pyridyl}ethyl)amine arms), was generated from chemical oxidation of the peroxide-bridged dicopper(II) complex [CuII2(XYLO)(O22-)]+ (2), using ferrocenium (Fc+) derivatives, in 2-methyltetrahydrofuran (MeTHF) at -125 °C. Using Me10Fc+, a 1 ⇆ 2 equilibrium was established, allowing for calculation of the reduction potential of 1 as -0.525 ± 0.01 V vs Fc+/0. Addition of 1 equiv of strong acid to 2 afforded the hydroperoxide-bridged dicopper(II) species [CuII2(XYLO)(OOH)]2+ (3). An acid-base equilibrium between 3 and 2 was achieved through spectral titrations using a derivatized phosphazene base. The pKa of 3 was thus determined to be 24 ± 0.6 in MeTHF at -125 °C. Using a thermodynamic square scheme and the Bordwell relationship, the hydroperoxo complex (3) O-H bond dissociation free energy (BDFE) was calculated as 81.8 ± 1.5 (BDE = 86.8) kcal/mol. The observed oxidizing capability of [CuII2(XYLO)(O2•-)]2+ (1), as demonstrated in H atom abstraction reactions with certain phenolic ArO-H and hydrocarbon C-H substrates, provides direct support for this experimentally determined O-H BDFE. A kinetic study reveals a very fast reaction of TEMPO-H with 1 in MeTHF, with k (-100 °C) = 5.6 M-1 s-1. Density functional theory (DFT) calculations reveal how the structure of 1 may minimize stabilization of the superoxide moiety, resulting in its enhanced reactivity. The thermodynamic insights obtained herein highlight the importance of the interplay between ligand design and the generation and properties of copper (or other metal ion) bound O2-derived reduced species, such as pKa, reduction potential, and BDFE; these may be relevant to the capabilities (i.e., oxidizing power) of reactive oxygen intermediates in metalloenzyme chemical system mediated oxidative processes.
Secondary coordination sphere interactions are critical in facilitating the formation, stabilization, and enhanced reactivity of high-valent oxidants required for essential biochemical processes. Herein, we compare the C–H bond oxidizing capabilities of spectroscopically characterized synthetic heme iron(IV) oxo complexes, F8Cmpd-II (F8 = tetrakis(2,6-difluorophenyl)porphyrinate), and a 2,6-lutidinium triflate (LutH+) Lewis acid adduct involving ferryl O-atom hydrogen-bonding, F8Cmpd-II(LutH+). Second-order rate constants utilizing C–H and C–D substrates were obtained by UV–vis spectroscopic monitoring, while products were characterized and quantified by EPR spectroscopy and gas chromatography (GC). With xanthene, F8Cmpd-II(LutH+) reacts 40 times faster (k2 = 14.2 M–1 s–1; −90 °C) than does F8Cmpd-II, giving bixanthene plus xanthone and the heme product [F8FeIIIOH2]+. For substrates with greater C–H bond dissociation energies (BDEs) F8Cmpd-II(LutH+) reacts with the second order rate constants k2(9,10-dihydroanthracene; DHA) = 0.485 M–1 s–1 and k2(fluorene) = 0.102 M–1 s–1 (–90 °C); by contrast, F8Cmpd-II is unreactive toward these substrates. For xanthene vs xanthene-(d2), large, nonclassical deuterium kinetic isotope effects are roughly estimated for both F8Cmpd-II and F8Cmpd-II(LutH+). The deuterated H-bonded analog, F8Cmpd-II(LutD+), was also prepared; for the reaction with DHA, an inverse KIE (compared to F8Cmpd-II(LutH+)) was observed. This work originates/inaugurates experimental investigation of the reactivity of authentic H-bonded heme-based FeIV═O compounds, critically establishing the importance of oxo H-bonding (or protonation) in heme complexes and enzyme active sites.