7 research outputs found

    Soluble epoxide hydrolase inhibitors: design, synthesis, in vitro profiling and in vivo evaluation in murine models of pain

    Get PDF
    Trabajo presentado en el ASPET Annual Meeting at Experimental Biology 2022, celebrado en Philadelphia, PA (Estados Unidos), del 2 al 5 de abril de 2022This research by the Grant PID2020-118127RB-I00 funded by MCIN/AEI/10.13039/501100011033 and by “ERDF A way of making Europe” to S.V. Financial support from Fundació Bosch i Gimpera, Universitat de Barcelona (F2I grant), to S.V., and from the Xunta de Galicia (ED431G 2019/02 and ED431C 2018/21) to M.I.L. are acknowledged. Partial support was provided by NIH-NIEHS River Award R35 ES03443, NIH-NIEHS Superfund Program P42 ES004699, NINDS R01 DK107767, and NIDDK R01 DK103616 to B.D.H. S.C. acknowledges a PhD fellowship from the Universitat de Barcelona (APIF grant)

    Life-long exposome and its effects on neuroinflammation related to Alzheimer’s disease

    No full text

    Pharmacological inhibition of soluble epoxide hydrolase during brain development induces long-term benefices in 5XFAD mice

    No full text
    Trabajo presentado en el XII Simposi de Neurobiologia, Societat Catalana de Biologia, celebrado en Barcelona (España), los días 7 y 8 de junio de 2022Modulation of the risk of Alzheimer¿s disease (AD) may begin early in life. During embryo development and maturation, the brain receives maternal physiological influences and establishes new epigenetic patterns that build its level of resilience to late diseases. We treated wild-type pregnant mice with the soluble epoxide hydrolase (sEH) inhibitor TPPU until their pups were weaned. The male progenitors were AD transgenic mice from the strain 5XFAD. At two months of age, male and female mice were blindly analyzed for learning and memory performance and their brain tissue was preserved for further analysis. Once genotyped, we found that 5XFAD mice worn from vehicle-treated mothers showed a poor response in cognitive tests of object recognition and spatial location. Notably, those 5XFAD mice from TPPU-treated mothers showed similar performance to their wild-type siblings. At the molecular level, tau pathology shown by increased hippocampal p-tau levels in 5XFAD mice was totally prevented in those whose mothers were treated with TPPU. Furthermore, TPPU treatment also modified the expression of epigenetic markers. Overall, we confirmed the potential of the enzyme sEH as a new target to fight AD and demonstrated that its inhibition in the developing brain produces long-term preventive effects against neurodegeneration.MCIN/AEI (PID2019-106285RB); AGAUR (2017-SGR-106); CSIC (2019AEP038)

    2-(Piperidin-4-yl)acetamides as Potent Inhibitors of Soluble Epoxide Hydrolase with Anti-Inflammatory Activity

    No full text
    The pharmacological inhibition of soluble epoxide hydrolase (sEH) has been suggested as a potential therapy for the treatment of pain and inflammatory diseases through the stabilization of endogenous epoxyeicosatrienoic acids. Numerous potent sEH inhibitors (sEHI) have been developed, however many contain highly lipophilic substituents limiting their availability. Recently, a new series of benzohomoadamantane-based ureas endowed with potent inhibitory activity for the human and murine sEH was reported. However, their very low microsomal stability prevented further development. Herein, a new series of benzohomoadamantane-based amides were synthetized, fully characterized, and evaluated as sEHI. Most of these amides were endowed with excellent inhibitory potencies. A selected compound displayed anti-inflammatory effects with higher effectiveness than the reference sEHI, TPPU.This research was funded by the Spanish Ministerio de Economía, Industria y Competitividad (Grants SAF2017-82771-R to S.V., PID2019-106285RB-C22 to C.S.), Grant PID2020-118127RB-I00 funded by MCIN/AEI/10.13039/501100011033 and by “ERDF A way of making Europe” to S.V., the Xunta de Galicia (ED431G 2019/02 and ED431C 2018/21), the Generalitat de Catalunya (2017 SGR 106, 2017 SGR 124 and 2017 SGR 1707). S.C. acknowledges a PhD fellowship from the Universitat de Barcelona (APIF grant). Partial support was provided by NIH-NIEHS River Award R35 ES03443, NIH-NIEHS Superfund Program P42 ES004699, NINDS R01 DK107767, and NIDDK R01 DK103616 to B.D.H. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health

    A new family of subnanomolar inhibitors of soluble epoxide hydrolase

    No full text
    Trabajo presentado en el ASPET Annual Meeting at Experimental Biology 2022, celebrado en Philadelphia, PA (Estados Unidos), del 2 al 5 de abril de 2022Epoxyeicosatrienoic acids (EETs) are endogenous chemical mediators derived from arachidonic acid that show anti-inflammatory, antihypertensive, analgesic, angiogenic, and antiatherosclerotic effects. Soluble epoxide hydrolase (sEH) converts EETs to the corresponding dihydroxyeicosatrienoic acids (DHETs), whereby the biological effects of EETs are diminished, eliminated, or altered.1 Therefore, inhibition of sEH has been suggested as a novel pharmacological approach for the treatment of pain-related disorders and various inflammatory diseases, and a few sEH inhibitors (sEHI) have reached clinical trialsThis research was funded by Grants PID2020-118127RB-I00 and PID2019-107991RB-I00 funded by MCIN/AEI/ 10.13039/501100011033 and by “ERDF A way of making Europe” to S.V. and C.E. Financial support from Generalitat de Catalunya (2017 SGR106) and Fundació Bosch i Gimpera, Universitat de Barcelona (F2I grant), to S.V., and from the Xunta de Galicia (ED431G 2019/02 and ED431C 2018/21) to M.I.L. are acknowledged. Partial support was provided by NIH-NIEHS River Award R35 ES03443, NIH-NIEHS Superfund Program P42 ES004699, NINDS R01 DK107767, and NIDDK R01DK103616 to B.D.H

    Neuroprotective Epigenetic Changes Induced by Maternal Treatment with an Inhibitor of Soluble Epoxide Hydrolase Prevents Early Alzheimer′s Disease Neurodegeneration

    No full text
    Modulation of Alzheimer′s disease (AD) risk begins early in life. During embryo development and postnatal maturation, the brain receives maternal physiological influences and establishes epigenetic patterns that build its level of resilience to late-life diseases. The soluble epoxide hydrolase inhibitor N-[1-(1-oxopropyl)-4-piperidinyl]-N′-[4-(trifluoromethoxy)phenyl] urea (TPPU), reported as ant-inflammatory and neuroprotective against AD pathology in the adult 5XFAD mouse model of AD, was administered to wild-type (WT) female mice mated to heterozygous 5XFAD males during gestation and lactation. Two-month-old 5XFAD male and female offspring of vehicle-treated dams showed memory loss as expected. Remarkably, maternal treatment with TPPU fully prevented memory loss in 5XFAD. TPPU-induced brain epigenetic changes in both WT and 5XFAD mice, modulating global DNA methylation (5-mC) and hydroxymethylation (5-hmC) and reducing the gene expression of some histone deacetylase enzymes (Hdac1 and Hdac2), might be on the basis of the long-term neuroprotection against cognitive impairment and neurodegeneration. In the neuropathological analysis, both WT and 5XFAD offspring of TPPU-treated dams showed lower levels of AD biomarkers of tau hyperphosphorylation and microglia activation (Trem2) than the offspring of vehicle-treated dams. Regarding sex differences, males and females were similarly protected by maternal TPPU, but females showed higher levels of AD risk markers of gliosis and neurodegeneration. Taken together, our results reveal that maternal treatment with TPPU impacts in preventing or delaying memory loss and AD pathology by inducing long-term modifications in the epigenetic machinery and its marks.This research was funded by the Spanish Ministerio de Ciencia e Innovación and the Agencia Estatal de Investigación (MCIN/AEI/10.13039/501100011033, grants PID2019-106285RB and PDC2021-121096) and by the European Union “NextGenerationEU”/PRTR; CSIC (2019AEP038); CERCA program/Generalitat de Catalunya; and 2017SGR106 from AGAUR

    Soluble epoxide hydrolase inhibitors counteract microglia phenotypic changes induced by monomeric C-reactive protein: New perspectives against neuroinflammation in Alzheimer's disease

    Get PDF
    Resumen del trabajo presentado en el XII Simposi de Neurobiologia, Societat Catalana de Biologia, celebrado en Barcelona (España), los días 7 y 8 de junio de 2022Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide. Neuroinflammation is a crucial neuropathological trait in AD, although the underlying mechanisms are not clarified. We previously proposed the monomeric C-reactive protein (mCRP), which is generated by activation and further disaggregation of blood CRP, as a trigger of AD neuropathology after cerebrovascular damage. mCRP is a potent pro-inflammatory agent found deposited in AD brain tissue and proved to induce AD-like dementia and tau and amyloid neuropathology in experimental models. Here we analysed the phenotypic changes induced by mCRP in microglia, the main effector cells of the inflammatory response. Then we tested the protective action of inhibiting the soluble epoxide hydrolase enzyme (sEH), a promising druggable target in AD. sEH inhibitors (sEHi) increase intracellular levels of anti-inflammatory epoxyeicosatrienoic acids (EETs). BV2 microglial cells were incubated with 50 or 100 µM mCRP for 24 h. Leading molecules of chemical families of newly synthesized sEHi (UB-JML-99 and UB-JM-39) were used for protective assays. mCRP activated the nitric oxide pathway as shown by increased release of nitric oxide and higher gene expression of iNOS, whereas sEHi agents blocked these effects. mCRP increased the release of TNF¿ into the media and the expression of pro-inflammatory cytokines and chemokines. Furthermore, mCRP modified the epigenetic microglial phenotype suggesting a dysregulated pattern. sEHi generally inhibited damaging effects of mCRP with a higher potency than the standard compound TPPU. Overall we demonstrated that mCRP directly activates microglia inflammatory pathways and may contribute to the triggering and progression of AD. Moreover, sEHi were confirmed as effective therapeutic molecules against neuroinflammation in an AD scenario.Agencia Estatal de Investigación (España); Ministerio de Ciencia, Innovación y Universidades (España); Generalitat de Cataluny
    corecore