Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38.395
Filter
1.
Cell Commun Signal ; 22(1): 306, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831454

ABSTRACT

BACKGROUND: Dysregulation in histone acetylation, a significant epigenetic alteration closely associated with major pathologies including cancer, promotes tumorigenesis, inactivating tumor-suppressor genes and activating oncogenic pathways. AMP-activated protein kinase (AMPK) is a cellular energy sensor that regulates a multitude of biological processes. Although a number of studies have identified the mechanisms by which AMPK regulates cancer growth, the underlying epigenetic mechanisms remain unknown. METHODS: The impact of metformin, an AMPK activator, on cervical cancer was evaluated through assessments of cell viability, tumor xenograft model, pan-acetylation analysis, and the role of the AMPK-PCAF-H3K9ac signaling pathway. Using label-free quantitative acetylproteomics and chromatin immunoprecipitation-sequencing (ChIP) technology, the activation of AMPK-induced H3K9 acetylation was further investigated. RESULTS: In this study, we found that metformin, acting as an AMPK agonist, activates AMPK, thereby inhibiting the proliferation of cervical cancer both in vitro and in vivo. Mechanistically, AMPK activation induces H3K9 acetylation at epigenetic level, leading to chromatin remodeling in cervical cancer. This also enhances the binding of H3K9ac to the promoter regions of multiple tumor suppressor genes, thereby promoting their transcriptional activation. Furthermore, the absence of PCAF renders AMPK activation incapable of inducing H3K9 acetylation. CONCLUSIONS: In conclusion, our findings demonstrate that AMPK mediates the inhibition of cervical cancer growth through PCAF-dependent H3K9 acetylation. This discovery not only facilitates the clinical application of metformin but also underscores the essential role of PCAF in AMPK activation-induced H3K9 hyperacetylation.


Subject(s)
AMP-Activated Protein Kinases , Cell Proliferation , Histones , Metformin , Uterine Cervical Neoplasms , p300-CBP Transcription Factors , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/genetics , Humans , Acetylation/drug effects , Female , Histones/metabolism , AMP-Activated Protein Kinases/metabolism , Cell Proliferation/drug effects , Animals , p300-CBP Transcription Factors/metabolism , p300-CBP Transcription Factors/genetics , Metformin/pharmacology , Mice , Mice, Nude , Cell Line, Tumor , Enzyme Activation/drug effects
2.
Braz Oral Res ; 38: e034, 2024.
Article in English | MEDLINE | ID: mdl-38747821

ABSTRACT

The aim of this study was to investigate the effect of acid challenge on the activation of matrix metalloproteinases (MMPs) in the Dentinoenamel junction of primary and permanent teeth submitted to radiotherapy. For this purpose, a total of 178 dental fragments obtained from molars were used, and randomly divided into 2 groups (primary and permanent teeth) / 4 experimental subgroups (irradiated and non-irradiated, demineralized and non-demineralized). The fragments were exposed to radiation, with a dose fraction of 2 Gy, for 5 consecutive days, until a total dose of 60 Gy was reached, with a total of 30 cycles, for 6 weeks. To determine the activity of MMPs on the dentinoenamel junction (DEJ), in situ zymography assays on 0.6mm dental fragments were performed. To assess whether MMP activity would be impacted by an acidic environment, the fragments were placed in a demineralizing solution (pH of 4.8). The finding was that irradiation activated MMPs in DEJ and these effects were more evident in permanent when compared with primary teeth. When the effect of an acid challenge on MMPs activity was investigated, demineralization was observed not to increase MMPs activity in non-irradiated teeth, but it did increase MMPs activity in irradiated teeth. In conclusion, an acid challenge was found to exacerbate activation of MMPs in DEJ of permanent teeth submitted to irradiation, but not in primary teeth.


Subject(s)
Matrix Metalloproteinases , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/radiation effects , Matrix Metalloproteinases/analysis , Humans , Time Factors , Tooth, Deciduous/radiation effects , Tooth, Deciduous/drug effects , Dentin/radiation effects , Dentin/drug effects , Dentin/enzymology , Dentition, Permanent , Random Allocation , Hydrogen-Ion Concentration , Tooth Demineralization , Statistics, Nonparametric , Analysis of Variance , Reference Values , Enzyme Activation/radiation effects , Enzyme Activation/drug effects
3.
Sci Total Environ ; 933: 173100, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38735330

ABSTRACT

Microplastics (MPs) are emerging pollutants of terrestrial ecosystems. The impacts of MP particle size on terrestrial systems remain unclear. The current study aimed to investigate the effects of six particle sizes (i.e., 4500, 1500, 500, 50, 5, and 0.5 µm) of polyethylene (PE) and polyvinyl chloride (PVC) on soil respiration, enzyme activity, bacteria, fungi, protists, and seed germination. MPs significantly promoted soil respiration, and the stimulating effects of PE were the strongest for medium and small-sized (0.5-1500 µm) particles, while those of PVC were the strongest for small particle sizes (0.5-50 µm). Large-sized (4500 µm) PE and all sizes of PVC significantly improved soil urease activity, while medium-sized (1500 µm) PVC significantly improved soil invertase activity. MPs altered the soil microbial community diversity, and the effects were especially pronounced for medium and small-sized (0.5-1500 µm) particles of PE and PVC on bacteria and fungi and small-sized (0.5 µm) particles of PE on protists. The impacts of MPs on bacteria and fungi were greater than on protists. The seed germination rate of Brassica chinensis decreased gradually with the decrease in PE MPs particle size. Therefore, to reduce the impact of MPs on soil ecosystems, effective measures should be taken to avoid the transformation of MPs into smaller particles in soil environmental management.


Subject(s)
Germination , Microbiota , Microplastics , Particle Size , Soil Microbiology , Soil Pollutants , Microplastics/pharmacology , Soil Pollutants/pharmacology , Germination/drug effects , Soil/chemistry , Soil/parasitology , Bacteria/drug effects , Bacteria/enzymology , Fungi/drug effects , Fungi/enzymology , Eukaryota/drug effects , Eukaryota/enzymology , Enzyme Activation/drug effects , Microbiota/drug effects , Biodiversity , Brassica/drug effects , Brassica/growth & development
4.
Dis Model Mech ; 17(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38747698

ABSTRACT

Diabetic nephropathy (DN), as a complication of diabetes, is a substantial healthcare challenge owing to the high risk of morbidity and mortality involved. Although significant progress has been made in understanding the pathogenesis of DN, more efficient models are required to develop new therapeutics. Here, we created a DN model in zebrafish by crossing diabetic Tg(acta1:dnIGF1R-EGFP) and proteinuria-tracing Tg(l-fabp::VDBP-GFP) lines, named zMIR/VDBP. Overfed adult zMIR/VDBP fish developed severe hyperglycemia and proteinuria, which were not observed in wild-type zebrafish. Renal histopathology revealed human DN-like characteristics, such as glomerular basement membrane thickening, foot process effacement and glomerular sclerosis. Glomerular dysfunction was restored upon calorie restriction. RNA sequencing analysis demonstrated that DN zebrafish kidneys exhibited transcriptional patterns similar to those seen in human DN pathogenesis. Notably, the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway was activated, a phenomenon observed in the early phase of human DN. In addition, metformin improved hyperglycemia and proteinuria in DN zebrafish by modulating Akt phosphorylation. Our results indicate that zMIR/VDBP fish are suitable for elucidating the mechanisms underlying human DN and could be a powerful tool for therapeutic discovery.


Subject(s)
Diabetic Nephropathies , Disease Models, Animal , Hyperglycemia , Proteinuria , Proto-Oncogene Proteins c-akt , Signal Transduction , Zebrafish , Animals , Hyperglycemia/complications , Hyperglycemia/pathology , Proto-Oncogene Proteins c-akt/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/metabolism , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Humans , Phosphorylation/drug effects , Animals, Genetically Modified , Metformin/pharmacology , Metformin/therapeutic use , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Kidney/pathology , Kidney/drug effects , Kidney/metabolism , Kidney Glomerulus/pathology , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Enzyme Activation/drug effects
6.
Nature ; 629(8010): 184-192, 2024 May.
Article in English | MEDLINE | ID: mdl-38600378

ABSTRACT

Glucocorticoids represent the mainstay of therapy for a broad spectrum of immune-mediated inflammatory diseases. However, the molecular mechanisms underlying their anti-inflammatory mode of action have remained incompletely understood1. Here we show that the anti-inflammatory properties of glucocorticoids involve reprogramming of the mitochondrial metabolism of macrophages, resulting in increased and sustained production of the anti-inflammatory metabolite itaconate and consequent inhibition of the inflammatory response. The glucocorticoid receptor interacts with parts of the pyruvate dehydrogenase complex whereby glucocorticoids provoke an increase in activity and enable an accelerated and paradoxical flux of the tricarboxylic acid (TCA) cycle in otherwise pro-inflammatory macrophages. This glucocorticoid-mediated rewiring of mitochondrial metabolism potentiates TCA-cycle-dependent production of itaconate throughout the inflammatory response, thereby interfering with the production of pro-inflammatory cytokines. By contrast, artificial blocking of the TCA cycle or genetic deficiency in aconitate decarboxylase 1, the rate-limiting enzyme of itaconate synthesis, interferes with the anti-inflammatory effects of glucocorticoids and, accordingly, abrogates their beneficial effects during a diverse range of preclinical models of immune-mediated inflammatory diseases. Our findings provide important insights into the anti-inflammatory properties of glucocorticoids and have substantial implications for the design of new classes of anti-inflammatory drugs.


Subject(s)
Anti-Inflammatory Agents , Glucocorticoids , Inflammation , Macrophages , Mitochondria , Succinates , Animals , Female , Humans , Male , Mice , Anti-Inflammatory Agents/pharmacology , Carboxy-Lyases/metabolism , Carboxy-Lyases/antagonists & inhibitors , Citric Acid Cycle/drug effects , Citric Acid Cycle/genetics , Cytokines/immunology , Cytokines/metabolism , Glucocorticoids/pharmacology , Glucocorticoids/metabolism , Hydro-Lyases/deficiency , Hydro-Lyases/genetics , Inflammation/drug therapy , Inflammation/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Receptors, Glucocorticoid/metabolism , Succinates/metabolism , Enzyme Activation/drug effects
7.
Eur J Pharmacol ; 972: 176558, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38614382

ABSTRACT

Inhibitors of polo-like kinase (PLK) are currently being evaluated as anticancer drugs. However, the molecular mechanism of PLK inhibitor-induced cell death is not fully understood. In this study, we found that GW843682X and BI2536, two inhibitors of PLK1, significantly induced cell death in multiple type cells. The induction of cell death was related to the preferring expression of PLK1. However, in human umbilical vascular endothelial cells (HUVEC) and human colorectal carcinoma cells, which expressed higher levels of both PLK1 and PLK2, PLK1 inhibitors induced very low levels of cell death. Clinical analysis reveals PLK1 presence in 26 of 30 NPC tumor tissues. In in vivo NPC lung metastasis nude mouse models, PLK1 inhibitors decreased NPC progress. Mechanistically, the PLK1 inhibitor did not activate p53, and the cell death was not reversed by p53 inhibition. Moreover, PLK1 inhibitor-induced cell death was PARP- and caspase-independent. Although PLK1 inhibitors induced down-regulation of calpain inhibitor calpastatin and calpain was activated by PLK1 inhibition, calpain blocking did not reverse cell death induced by PLK1 inhibitors, suggesting the non-involvement of calpain. Surprisingly, we found that PLK1 inhibitors induced the activation of proteasome, and the treatment of cells with PLK1 inhibitors reduced the levels of ubiquitinated proteins. And proteasome inhibitors reversed cell death induced by PLK1 inhibitors in various cell types in which PLK1 was preferentially expressed. Moreover, PLK1 inhibition reversed the degradation of proteins including p53, caspase 8, PARP and calpastatin. These results suggest that the activation of proteasome is critical for cell death induced by PLK1 inhibition.


Subject(s)
Cell Cycle Proteins , Cell Death , Polo-Like Kinase 1 , Proteasome Endopeptidase Complex , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Humans , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Animals , Proteasome Endopeptidase Complex/metabolism , Cell Death/drug effects , Mice , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Mice, Nude , Pteridines/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Calpain/antagonists & inhibitors , Calpain/metabolism , Enzyme Activation/drug effects , Xenograft Model Antitumor Assays , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology
8.
Chem Biol Interact ; 394: 110995, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38583854

ABSTRACT

Small molecule-driven ERK activation is known to induce autophagy and ferroptosis in cancer cells. Herein the effect of cannabidiol (CBD), a phytochemical derived from Cannabis sativa, on ERK-driven autophagy and ferroptosis has been demonstrated in glioblastoma (GBM) cells (U87 and U373 cells). CBD imparted significant cytotoxicity in GBM cells, induced activation of ERK (not JNK and p38), and increased intracellular reactive oxygen species (ROS) levels. It increased the autophagy-related proteins such as LC3 II, Atg7, and Beclin-1 and modulated the expression of ferroptosis-related proteins such as glutathione peroxidase 4 (GPX4), SLC7A11, and TFRC. CBD significantly elevated the endoplasmic reticulum stress, ROS, and iron load, and decreased GSH levels. Inhibitors of autophagy (3-MA) and ferroptosis (Fer-1) had a marginal effect on CBD-induced autophagy/ferroptosis. Treatment with N-acetyl-cysteine (antioxidant) or PD98059 (ERK inhibitor) partly reverted the CBD-induced autophagy/ferroptosis by decreasing the activation of ERK and the production of ROS. Overall, CBD induced autophagy and ferroptosis through the activation of ERK and generation of ROS in GBM cells.


Subject(s)
Autophagy , Cannabidiol , Ferroptosis , Glioblastoma , Reactive Oxygen Species , Humans , Autophagy/drug effects , Beclin-1/metabolism , Cannabidiol/pharmacology , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Ferroptosis/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/drug therapy , MAP Kinase Signaling System/drug effects , Reactive Oxygen Species/metabolism
9.
Food Chem ; 448: 139082, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38537544

ABSTRACT

ß-galactosidase (lactase) is commercially important as a dietary supplement to alleviate the symptoms of lactose intolerance. This work investigated a unique activation of CMP (carboxymethylated (1 â†’ 3)-ß-d-glucan) on lactase and its mechanism by comparing it with carboxymethyl chitosan (CMCS), an inhibitor of lactase. The results illustrated that the secondary and tertiary structures of lactase were altered and its active sites exposed after complexation with CMP, and dissociation of lactase aggregates was also observed. These changes favored better accessibility of the substrate to the active sites of lactase, resulting in a maximum increase of 60.5 % in lactase activity. Furthermore, the hydrophobic and electrostatic interactions with lactase caused by the carboxymethyl group of CMP were shown to be crucial for its activation ability. Thus, the improvement of lactase activity and stability by CMP shown here is important for the development of new products in the food and pharmaceutical industries.


Subject(s)
Chitosan/analogs & derivatives , Hydrophobic and Hydrophilic Interactions , Static Electricity , beta-Galactosidase , beta-Glucans , beta-Galactosidase/chemistry , beta-Galactosidase/metabolism , beta-Glucans/chemistry , beta-Glucans/pharmacology , Chitosan/chemistry , Enzyme Stability , Kinetics , Enzyme Activation/drug effects
10.
Int J Biol Macromol ; 266(Pt 2): 131065, 2024 May.
Article in English | MEDLINE | ID: mdl-38521329

ABSTRACT

Protein C inhibitor (PCI) maintains hemostasis by inhibiting both procoagulant and anticoagulant serine proteases, and plays important roles in coagulation, fibrinolysis, reproduction, and anti-angiogenesis. The reactive site loop of PCI traps and irreversibly inhibits the proteases like APC (activating protein C), thrombin (FIIa) and factor Xa (FXa). Previous studies on antithrombin (ATIII) had identified Tyr253 and Glu255 as functional exosites that interact and aid in the inhibition of factor IXa and FXa. Presence of exosite in PCI is not known, however a sequence comparison with the PCI from different vertebrate species and ATIII identified Glu239 to be absolutely conserved. PCI residues analogous to ATIII exosite residues were mutated to R238A and E239A. Purified variant PCI in the presence of heparin (10 µg/ml) showed a 2-4 fold decrease in the rate of inhibition of the proteases. However, the stoichiometry of inhibition of FIIa, APC, and FXa by native PCI, R238A and E239A variants were found to be close to 1.0, which also indicated the formation of stable complexes based on SDS-PAGE and western blot analysis with thrombin and APC. Our findings revealed the possible presence of an exosite in PCI that influences the protease inhibition rates.


Subject(s)
Heparin , Protein C Inhibitor , Serine Proteases , Protein C Inhibitor/chemistry , Protein C Inhibitor/metabolism , Heparin/chemistry , Heparin/pharmacology , Humans , Serine Proteases/metabolism , Serine Proteases/chemistry , Thrombin/metabolism , Protein C/metabolism , Protein C/chemistry , Factor Xa/metabolism , Factor Xa/chemistry , Amino Acid Sequence , Enzyme Activation/drug effects
11.
Nature ; 629(8014): 1182-1191, 2024 May.
Article in English | MEDLINE | ID: mdl-38480881

ABSTRACT

G-protein-coupled receptors (GPCRs) activate heterotrimeric G proteins by stimulating guanine nucleotide exchange in the Gα subunit1. To visualize this mechanism, we developed a time-resolved cryo-EM approach that examines the progression of ensembles of pre-steady-state intermediates of a GPCR-G-protein complex. By monitoring the transitions of the stimulatory Gs protein in complex with the ß2-adrenergic receptor at short sequential time points after GTP addition, we identified the conformational trajectory underlying G-protein activation and functional dissociation from the receptor. Twenty structures generated from sequential overlapping particle subsets along this trajectory, compared to control structures, provide a high-resolution description of the order of main events driving G-protein activation in response to GTP binding. Structural changes propagate from the nucleotide-binding pocket and extend through the GTPase domain, enacting alterations to Gα switch regions and the α5 helix that weaken the G-protein-receptor interface. Molecular dynamics simulations with late structures in the cryo-EM trajectory support that enhanced ordering of GTP on closure of the α-helical domain against the nucleotide-bound Ras-homology domain correlates with α5 helix destabilization and eventual dissociation of the G protein from the GPCR. These findings also highlight the potential of time-resolved cryo-EM as a tool for mechanistic dissection of GPCR signalling events.


Subject(s)
Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gs , Receptors, Adrenergic, beta-2 , Humans , Binding Sites , GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/drug effects , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein alpha Subunits, Gs/ultrastructure , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Models, Molecular , Molecular Dynamics Simulation , Protein Binding , Receptors, Adrenergic, beta-2/metabolism , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/ultrastructure , Time Factors , Enzyme Activation/drug effects , Protein Domains , Protein Structure, Secondary , Signal Transduction/drug effects
12.
FEBS J ; 291(9): 1944-1957, 2024 May.
Article in English | MEDLINE | ID: mdl-38335056

ABSTRACT

The transmembrane receptor for advanced glycation end products (RAGE) is a signaling receptor for many damage- and pathogen-associated molecules. Activation of RAGE is associated with inflammation and an increase in reactive oxygen species (ROS) production. Although several sources of ROS have been previously suggested, how RAGE induces ROS production is still unclear, considering the multiple targets of pathogen-associated molecules. Here, using acute brain slices and primary co-culture of cortical neurons and astrocytes, we investigated the effects of a range of synthetic peptides corresponding to the fragments of the RAGE V-domain on redox signaling. We found that the synthetic fragment (60-76) of the RAGE V-domain induces activation of ROS production in astrocytes and neurons from the primary co-culture and acute brain slices. This effect occurred through activation of RAGE and could be blocked by a RAGE inhibitor. Activation of RAGE by the synthetic fragment stimulates ROS production in NADPH oxidase (NOX). This RAGE-induced NOX activation produced only minor decreases in glutathione levels and increased the rate of lipid peroxidation, although it also reduced basal and ß-amyloid induced cell death in neurons and astrocytes. Thus, specific activation of RAGE induces redox signaling through NOX, which can be a part of a cell protective mechanism.


Subject(s)
Astrocytes , Coculture Techniques , NADPH Oxidases , Neurons , Reactive Oxygen Species , Receptor for Advanced Glycation End Products , Astrocytes/metabolism , Astrocytes/drug effects , Neurons/metabolism , Neurons/drug effects , Animals , Receptor for Advanced Glycation End Products/metabolism , Receptor for Advanced Glycation End Products/genetics , Reactive Oxygen Species/metabolism , NADPH Oxidases/metabolism , NADPH Oxidases/genetics , Neuroprotection , Cells, Cultured , Oxidation-Reduction , Signal Transduction , Mice , Lipid Peroxidation/drug effects , Rats , Enzyme Activation/drug effects , Glutathione/metabolism
13.
J Nat Med ; 78(1): 208-215, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38063995

ABSTRACT

Recently, the number of patients diagnosed with dementia has increased. The World Health Organization (WHO) estimates that 50 million patients suffer from dementia. Although several therapeutic strategies have been proposed, currently, there is no curative approach for treating dementia. Neurodegeneration is an irreversible process. As this disease gradually progresses over 15-20 years, a low-cost and sustainable method for preventing these diseases is desired. Cacao nib is consumed in many countries, and a recent clinical study indicated that cocoa intake upregulates brain-derived neurotrophic factor (BDNF), which plays a significant role in memory formation and neuronal cell survival. In the present study, neural cells were treated with cacao nib extract or the 17 characteristic components of cacao nib. Treatment with Cacao nib extract upregulates BDNF mRNA expression. In addition, cacao nib extract elicits the phosphorylation of cAMP-response-element-binding protein (CREB), which regulates the transcription of BDNF. Among the 17 species screened, isovaleraldehyde (IVA), also known as an aroma component of cacao nibs extract, improved BDNF mRNA expression without SH-SY5Y cell toxicity. IVA also promoted CREB phosphorylation through a cAMP-dependent protein kinase (PKA)-dependent mechanism. In conclusion, IVA could be responsible for the BDNF upregulation effect of cacao nib, and IVA upregulated BDNF expression via the PKA-CREB axis.


Subject(s)
Aldehydes , Brain-Derived Neurotrophic Factor , Cyclic AMP Response Element-Binding Protein , Neuroprotective Agents , Up-Regulation , Neuroprotective Agents/pharmacology , Aldehydes/pharmacology , Up-Regulation/drug effects , Brain-Derived Neurotrophic Factor/genetics , Humans , Cell Line, Tumor , Cacao/chemistry , Plant Extracts/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Signal Transduction/drug effects , Enzyme Activation/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism
14.
Hear Res ; 441: 108919, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38043402

ABSTRACT

Auditory neuropathy spectrum disorder (ANSD) is a hearing impairment involving disruptions to inner hair cells (IHCs), ribbon synapses, spiral ganglion neurons (SGNs), and/or the auditory nerve itself. The outcomes of cochlear implants (CI) for ANSD are variable and dependent on the location of lesion sites. Discovering a potential therapeutic agent for ANSD remains an urgent requirement. Here, 293T stable transfection cell lines and patient induced pluripotent stem cells (iPSCs)-derived auditory neurons carrying the apoptosis inducing factor (AIF) p.R422Q variant were used to pursue a therapeutic regent for ANSD. Nicotinamide adenine dinucleotide (NADH) is a main electron donor in the electron transport chain (ETC). In 293T stable transfection cells with the p.R422Q variant, NADH treatment improved AIF dimerization, rescued mitochondrial dysfunctions, and decreased cell apoptosis. The effects of NADH were further confirmed in patient iPSCs-derived neurons. The relative level of AIF dimers was increased to 150.7 % (P = 0.026) from 59.2 % in patient-neurons upon NADH treatment. Such increased AIF dimerization promoted the mitochondrial import of coiled-coil-helix-coiled-coil-helix domain-containing protein 4 (CHCHD4), which further restored mitochondrial functions. Similarly, the content of mitochondrial calcium (mCa2+) was downregulated from 136.7 % to 102.3 % (P = 0.0024) in patient-neurons upon NADH treatment. Such decreased mCa2+ levels inhibited calpain activity, ultimately reducing the percentage of apoptotic cells from 30.5 % to 21.1 % (P = 0.021). We also compared the therapeutic effects of gene correction and NADH treatment on hereditary ANSD. NADH treatment had comparable restorative effects on functions of ANSD patient-specific cells to that of gene correction. Our findings offer evidence of the molecular mechanisms of ANSD and introduce NADH as a potential therapeutic agent for ANSD therapy.


Subject(s)
Apoptosis Inducing Factor , Apoptosis , Hearing Loss, Central , NAD , Sensory Receptor Cells , Hearing Loss, Central/genetics , Hearing Loss, Central/metabolism , Hearing Loss, Central/physiopathology , Apoptosis/drug effects , NAD/pharmacology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Dimerization , Mitochondria/drug effects , HEK293 Cells , Mitochondrial Precursor Protein Import Complex Proteins/metabolism , Calcium/metabolism , Reactive Oxygen Species/metabolism , Calpain/metabolism , Enzyme Activation/drug effects , Genotype , Humans , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism
15.
Cell Rep ; 42(12): 113535, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38060450

ABSTRACT

The phosphoinositide 3-kinase p110α is an essential mediator of insulin signaling and glucose homeostasis. We interrogated the human serine, threonine, and tyrosine kinome to search for novel regulators of p110α and found that the Hippo kinases phosphorylate p110α at T1061, which inhibits its activity. This inhibitory state corresponds to a conformational change of a membrane-binding domain on p110α, which impairs its ability to engage membranes. In human primary hepatocytes, cancer cell lines, and rodent tissues, activation of the Hippo kinases MST1/2 using forskolin or epinephrine is associated with phosphorylation of T1061 and inhibition of p110α, impairment of downstream insulin signaling, and suppression of glycolysis and glycogen synthesis. These changes are abrogated when MST1/2 are genetically deleted or inhibited with small molecules or if the T1061 is mutated to alanine. Our study defines an inhibitory pathway of PI3K signaling and a link between epinephrine and insulin signaling.


Subject(s)
Protein Serine-Threonine Kinases , Humans , Animals , Mice , Cell Line , Mice, Inbred C57BL , Male , Female , Epinephrine/pharmacology , Enzyme Activation/drug effects , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Gene Deletion , Colforsin/pharmacology , Insulin/metabolism , Phosphorylation/drug effects , Hippo Signaling Pathway/drug effects , Hippo Signaling Pathway/genetics
16.
J Biol Chem ; 299(12): 105369, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37865311

ABSTRACT

Cardiac MyBP-C (cMyBP-C) interacts with actin and myosin to fine-tune cardiac muscle contractility. Phosphorylation of cMyBP-C, which reduces the binding of cMyBP-C to actin and myosin, is often decreased in patients with heart failure (HF) and is cardioprotective in model systems of HF. Therefore, cMyBP-C is a potential target for HF drugs that mimic its phosphorylation and/or perturb its interactions with actin or myosin. We labeled actin with fluorescein-5-maleimide (FMAL) and the C0-C2 fragment of cMyBP-C (cC0-C2) with tetramethylrhodamine (TMR). We performed two complementary high-throughput screens (HTS) on an FDA-approved drug library, to discover small molecules that specifically bind to cMyBP-C and affect its interactions with actin or myosin, using fluorescence lifetime (FLT) detection. We first excited FMAL and detected its FLT, to measure changes in fluorescence resonance energy transfer (FRET) from FMAL (donor) to TMR (acceptor), indicating binding. Using the same samples, we then excited TMR directly, using a longer wavelength laser, to detect the effects of compounds on the environmentally sensitive FLT of TMR, to identify compounds that bind directly to cC0-C2. Secondary assays, performed on selected modulators with the most promising effects in the primary HTS assays, characterized the specificity of these compounds for phosphorylated versus unphosphorylated cC0-C2 and for cC0-C2 versus C1-C2 of fast skeletal muscle (fC1-C2). A subset of identified compounds modulated ATPase activity in cardiac and/or skeletal myofibrils. These assays establish the feasibility of the discovery of small-molecule modulators of the cMyBP-C-actin/myosin interaction, with the ultimate goal of developing therapies for HF.


Subject(s)
Carrier Proteins , Drug Discovery , Heart Failure , Myofibrils , Small Molecule Libraries , Humans , Actins/metabolism , Drug Discovery/methods , Heart Failure/drug therapy , Heart Failure/metabolism , Myocardium/metabolism , Myosins/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Small Molecule Libraries/pharmacology , Drug Evaluation, Preclinical , Myofibrils/drug effects , Carrier Proteins/metabolism , Biosensing Techniques , Adenosine Triphosphatases/metabolism , Muscle, Skeletal/metabolism , Recombinant Proteins/metabolism , Enzyme Activation/drug effects , Fluorescence Resonance Energy Transfer
17.
J Biol Chem ; 299(12): 105363, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37863262

ABSTRACT

Metformin is among the most prescribed medications worldwide and the first-line therapy for type 2 diabetes. However, gastrointestinal side effects are common and can be dose limiting. The total daily metformin dose frequently reaches several grams, and poor absorption results in high intestinal drug concentrations. Here, we report that metformin inhibits the activity of enteropeptidase and other digestive enzymes at drug concentrations predicted to occur in the human duodenum. Treatment of mouse gastrointestinal tissue with metformin reduces enteropeptidase activity; further, metformin-treated mice exhibit reduced enteropeptidase activity, reduced trypsin activity, and impaired protein digestion within the intestinal lumen. These results indicate that metformin-induced protein maldigestion could contribute to the gastrointestinal side effects and other impacts of this widely used drug.


Subject(s)
Enteropeptidase , Metformin , Proteolysis , Animals , Humans , Mice , Diabetes Mellitus, Type 2/drug therapy , Enteropeptidase/metabolism , Metformin/adverse effects , Metformin/pharmacology , Metformin/therapeutic use , Proteolysis/drug effects , Enzyme Activation/drug effects , Gastrointestinal Tract/enzymology , Trypsin/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use
18.
J Biol Chem ; 299(12): 105366, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37863264

ABSTRACT

Hypoxic responses in plants involve Plant Cysteine Oxidases (PCOs). They catalyze the N-terminal cysteine oxidation of Ethylene Response Factors VII (ERF-VII) in an oxygen-dependent manner, leading to their degradation via the cysteine N-degron pathway (Cys-NDP) in normoxia. In hypoxia, PCO activity drops, leading to the stabilization of ERF-VIIs and subsequent hypoxic gene upregulation. Thus far, no chemicals have been described to specifically inhibit PCO enzymes. In this work, we devised an in vivo pipeline to discover Cys-NDP effector molecules. Budding yeast expressing AtPCO4 and plant-based ERF-VII reporters was deployed to screen a library of natural-like chemical scaffolds and was further combined with an Arabidopsis Cys-NDP reporter line. This strategy allowed us to identify three PCO inhibitors, two of which were shown to affect PCO activity in vitro. Application of these molecules to Arabidopsis seedlings led to an increase in ERF-VII stability, induction of anaerobic gene expression, and improvement of tolerance to anoxia. By combining a high-throughput heterologous platform and the plant model Arabidopsis, our synthetic pipeline provides a versatile system to study how the Cys-NDP is modulated. Its first application here led to the discovery of at least two hypoxia-mimicking molecules with the potential to impact plant tolerance to low oxygen stress.


Subject(s)
Arabidopsis Proteins , Cysteine Dioxygenase , Enzyme Inhibitors , Small Molecule Libraries , Humans , Arabidopsis/drug effects , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cysteine/metabolism , Cysteine Dioxygenase/antagonists & inhibitors , Cysteine Dioxygenase/metabolism , Gene Expression Regulation, Plant/drug effects , Oxygen/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Drug Evaluation, Preclinical/methods , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Seedlings/drug effects , Anaerobiosis , Degrons , Enzyme Activation/drug effects , Recombinant Proteins/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology
19.
J Biol Chem ; 299(8): 104889, 2023 08.
Article in English | MEDLINE | ID: mdl-37286041

ABSTRACT

Human neutrophil elastase (HNE) plays a pivotal role in innate immunity, inflammation, and tissue remodeling. Aberrant proteolytic activity of HNE contributes to organ destruction in various chronic inflammatory diseases including emphysema, asthma, and cystic fibrosis. Therefore, elastase inhibitors could alleviate the progression of these disorders. Here, we used the systematic evolution of ligands by exponential enrichment to develop ssDNA aptamers that specifically target HNE. We determined the specificity of the designed inhibitors and their inhibitory efficacy against HNE using biochemical and in vitro methods, including an assay of neutrophil activity. Our aptamers inhibit the elastinolytic activity of HNE with nanomolar potency and are highly specific for HNE and do not target other tested human proteases. As such, this study provides lead compounds suitable for the evaluation of their tissue-protective potential in animal models.


Subject(s)
Aptamers, Nucleotide , Leukocyte Elastase , Serine Proteinase Inhibitors , Humans , Cystic Fibrosis/drug therapy , Emphysema/drug therapy , Leukocyte Elastase/antagonists & inhibitors , Neutrophils/drug effects , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/pharmacology , Serine Proteinase Inhibitors/therapeutic use , Aptamers, Nucleotide/chemical synthesis , Aptamers, Nucleotide/pharmacology , Aptamers, Nucleotide/therapeutic use , Sensitivity and Specificity , Enzyme Activation/drug effects , Proteolysis/drug effects , Cells, Cultured
20.
J Biol Chem ; 299(4): 104595, 2023 04.
Article in English | MEDLINE | ID: mdl-36898579

ABSTRACT

The integrated stress response (ISR) is an important mechanism by which cells confer protection against environmental stresses. Central to the ISR is a collection of related protein kinases that monitor stress conditions, such as Gcn2 (EIF2AK4) that recognizes nutrient limitations, inducing phosphorylation of eukaryotic translation initiation factor 2 (eIF2). Gcn2 phosphorylation of eIF2 lowers bulk protein synthesis, conserving energy and nutrients, coincident with preferential translation of stress-adaptive gene transcripts, such as that encoding the Atf4 transcriptional regulator. While Gcn2 is central for cell protection to nutrient stress and its depletion in humans leads to pulmonary disorders, Gcn2 can also contribute to the progression of cancers and facilitate neurological disorders during chronic stress. Consequently, specific ATP-competitive inhibitors of Gcn2 protein kinase have been developed. In this study, we report that one such Gcn2 inhibitor, Gcn2iB, can activate Gcn2, and we probe the mechanism by which this activation occurs. Low concentrations of Gcn2iB increase Gcn2 phosphorylation of eIF2 and enhance Atf4 expression and activity. Of importance, Gcn2iB can activate Gcn2 mutants devoid of functional regulatory domains or with certain kinase domain substitutions derived from Gcn2-deficient human patients. Other ATP-competitive inhibitors can also activate Gcn2, although there are differences in their mechanisms of activation. These results provide a cautionary note about the pharmacodynamics of eIF2 kinase inhibitors in therapeutic applications. Compounds designed to be kinase inhibitors that instead directly activate Gcn2, even loss of function variants, may provide tools to alleviate deficiencies in Gcn2 and other regulators of the ISR.


Subject(s)
Eukaryotic Initiation Factor-2 , Protein Serine-Threonine Kinases , Humans , Adenosine Triphosphate/metabolism , Enzyme Activation/drug effects , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...