Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.989
Filter
1.
Protein Sci ; 33(7): e5067, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38864716

ABSTRACT

The N-degron pathway determines the half-life of proteins by selectively destabilizing the proteins bearing N-degrons. N-terminal glutamine amidohydrolase 1 (NTAQ1) plays an essential role in the arginine N-degron (Arg/N-degron) pathway as an initializing enzyme via the deamidation of the N-terminal (Nt) glutamine (Gln). However, the Nt-serine-bound conformation of hNTAQ1 according to the previously identified crystal structure suggests the possibility of other factors influencing the recognition of Nt residues by hNTAQ1. Hence, in the current study, we aimed to further elucidate the substrate recognition of hNTAQ1; specifically, we explored 12 different substrate-binding conformations of hNTAQ1 depending on the subsequent residue of Nt-Gln. Results revealed that hNTAQ1 primarily interacts with the protein Nt backbone, instead of the side chain, for substrate recognition. Here, we report that the Nt backbone of proteins appears to be a key component of hNTAQ1 function and is the main determinant of substrate recognition. Moreover, not all second residues from Nt-Gln, but rather distinctive and charged residues, appeared to aid in detecting substrate recognition. These new findings define the substrate-recognition process of hNTAQ1 and emphasize the importance of the subsequent Gln residue in the Nt-Gln degradation system. Our extensive structural and biochemical analyses provide insights into the substrate specificity of the N-degron pathway and shed light on the mechanism underlying hNTAQ1 substrate recognition. An improved understanding of the protein degradation machinery could aid in developing therapies to promote overall health through enhanced protein regulation, such as targeted protein therapies.


Subject(s)
Arginine , Humans , Substrate Specificity , Arginine/chemistry , Arginine/metabolism , Models, Molecular , Glutamine/metabolism , Glutamine/chemistry , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Amidohydrolases/genetics , Protein Conformation , Proteolysis , Degrons
2.
ACS Sens ; 9(6): 3387-3393, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38850514

ABSTRACT

Fatty acid amide hydrolase (FAAH) plays a crucial role in the metabolism of the endocannabinoid system by hydrolyzing a series of bioactive amides, whose abnormal levels are associated with neuronal disorders including Alzheimer's disease (AD). However, due to the lack of suitable quantitative sensing tools, real-time and accurate monitoring of the activity of FAAH in living systems remains unresolved. Herein, a novel enzyme-activated near-infrared two-photon ratiometric fluorescent probe (CANP) based on a naphthylvinylpyridine monofluorophore is successfully developed, in which the electron-withdrawing amide moiety is prone to be hydrolyzed to an electron-donating amine group under the catalysis of FAAH, leading to the activation of the intramolecular charge transfer process and the emergence of a new 80 nm red-shifted emission, thereby achieving a ratiometric luminescence response. Benefiting from the high selectivity, high sensitivity, and ratiometric response to FAAH, the probe CANP is successfully used to quantitatively monitor and image the FAAH levels in living neurons, by which an amyloid ß (Aß)-induced upregulation of endogenous FAAH activity is observed. Similar increases in FAAH activity are found in various brain regions of AD model mice, indicating a potential fatty acid amide metabolite-involved pathway for the pathological deterioration of AD. Moreover, our quantitative FAAH inhibition experiments further demonstrate the great value of CANP as an efficient visual probe for in situ and precise assessment of FAAH inhibitors in complex living systems, assisting the discovery of FAAH-related therapeutic agents.


Subject(s)
Amidohydrolases , Brain , Fluorescent Dyes , Neurons , Amidohydrolases/metabolism , Animals , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Brain/diagnostic imaging , Brain/metabolism , Neurons/metabolism , Mice , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/analysis , Humans , Pyridines/chemistry , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Photons
3.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891799

ABSTRACT

Emerging research links the endocannabinoid system to gut microbiota, influencing nociception, mood, and immunity, yet the molecular interactions remain unclear. This study focused on the effects of probiotics on ECS markers-cannabinoid receptor type 2 (CB2) and fatty acid amide hydrolase (FAAH)-in dancers, a group selected due to their high exposure to physical and psychological stress. In a double-blind, placebo-controlled trial (ClinicalTrials.gov NCT05567653), 15 dancers were assigned to receive either a 12-week regimen of Lactobacillus helveticus Rosell-52 and Bifidobacterium longum Rosell-17 or a placebo (PLA: n = 10, PRO: n = 5). There were no significant changes in CB2 (probiotic: 0.55 to 0.29 ng/mL; placebo: 0.86 to 0.72 ng/mL) or FAAH levels (probiotic: 5.93 to 6.02 ng/mL; placebo: 6.46 to 6.94 ng/mL; p > 0.05). A trend toward improved sleep quality was observed in the probiotic group, while the placebo group showed a decline (PRO: from 1.4 to 1.0; PLA: from 0.8 to 1.2; p = 0.07841). No other differences were noted in assessed outcomes (pain and fatigue). Probiotic supplementation showed no significant impact on CB2 or FAAH levels, pain, or fatigue but suggested potential benefits for sleep quality, suggesting an area for further research.


Subject(s)
Amidohydrolases , Endocannabinoids , Fatigue , Pain , Probiotics , Sleep , Humans , Probiotics/therapeutic use , Endocannabinoids/metabolism , Female , Double-Blind Method , Fatigue/metabolism , Adult , Male , Pain/drug therapy , Sleep/drug effects , Sleep/physiology , Amidohydrolases/metabolism , Young Adult , Receptor, Cannabinoid, CB2/metabolism , Gastrointestinal Microbiome/drug effects , Adolescent
4.
Drug Des Devel Ther ; 18: 2143-2167, 2024.
Article in English | MEDLINE | ID: mdl-38882045

ABSTRACT

Over the past decade, the idea of targeting the endocannabinoid system to treat anxiety disorders has received increasing attention. Previous studies focused more on developing cannabinoid receptor agonists or supplementing exogenous cannabinoids, which are prone to various adverse effects due to their strong pharmacological activity and poor receptor selectivity, limiting their application in clinical research. Endocannabinoid hydrolase inhibitors are considered to be the most promising development strategies for the treatment of anxiety disorders. More recent efforts have emphasized that inhibition of two major endogenous cannabinoid hydrolases, monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH), indirectly activates cannabinoid receptors by increasing endogenous cannabinoid levels in the synaptic gap, circumventing receptor desensitization resulting from direct enhancement of endogenous cannabinoid signaling. In this review, we comprehensively summarize the anxiolytic effects of MAGL and FAAH inhibitors and their potential pharmacological mechanisms, highlight reported novel inhibitors or natural products, and provide an outlook on future directions in this field.


Subject(s)
Amidohydrolases , Anti-Anxiety Agents , Endocannabinoids , Enzyme Inhibitors , Monoacylglycerol Lipases , Humans , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/chemistry , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Monoacylglycerol Lipases/antagonists & inhibitors , Monoacylglycerol Lipases/metabolism , Animals , Endocannabinoids/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Anxiety Disorders/drug therapy , Anxiety Disorders/metabolism
5.
Cell Death Dis ; 15(6): 399, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849335

ABSTRACT

The loss of dopaminergic neurons in the substantia nigra is a hallmark of pathology in Parkinson's disease (PD). Dimethylarginine dimethylaminohydrolase-1 (DDAH-1) is the critical enzyme responsible for the degradation of asymmetric dimethylarginine (ADMA) which inhibits nitric oxide (NO) synthase and has been implicated in neurodegeneration. Mitochondrial dysfunction, particularly in the mitochondria-associated endoplasmic reticulum membrane (MAM), plays a critical role in this process, although the specific molecular target has not yet been determined. This study aims to examine the involvement of DDAH-1 in the nigrostriatal dopaminergic pathway and PD pathogenesis. The distribution of DDAH-1 in the brain and its colocalization with dopaminergic neurons were observed. The loss of dopaminergic neurons and aggravated locomotor disability after rotenone (ROT) injection were showed in the DDAH-1 knockout rat. L-arginine (ARG) and NO donors were employed to elucidate the role of NO respectively. In vitro, we investigated the effects of DDAH-1 knockdown or overexpression on cell viability and mitochondrial functions, as well as modulation of ADMA/NO levels using ADMA or ARG. MAM formation was assessed by the Mitofusin2 oligomerization and the mitochondrial ubiquitin ligase (MITOL) phosphorylation. We found that DDAH-1 downregulation resulted in enhanced cell death and mitochondrial dysfunctions, accompanied by elevated ADMA and reduced NO levels. However, the recovered NO level after the ARG supplement failed to exhibit a protective effect on mitochondrial functions and partially restored cell viability. DDAH-1 overexpression prevented ROT toxicity, while ADMA treatment attenuated these protective effects. The declines of MAM formation in ROT-treated cells were exacerbated by DDAH-1 downregulation via reduced MITOL phosphorylation, which was reversed by DDAH-1 overexpression. Together, the abundant expression of DDAH-1 in nigral dopaminergic neurons may exert neuroprotective effects by maintaining MAM formation and mitochondrial function probably via ADMA, indicating the therapeutic potential of targeting DDAH-1 for PD.


Subject(s)
Amidohydrolases , Arginine , Dopaminergic Neurons , Endoplasmic Reticulum , Mitochondria , Nitric Oxide , Parkinson Disease , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Animals , Amidohydrolases/metabolism , Amidohydrolases/genetics , Mitochondria/metabolism , Mitochondria/drug effects , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/genetics , Arginine/metabolism , Arginine/analogs & derivatives , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/drug effects , Rats , Nitric Oxide/metabolism , Male , Rats, Sprague-Dawley , Humans , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/genetics , Rotenone/pharmacology , Mitochondrial Proteins/metabolism , Mitochondria Associated Membranes
6.
Sci Rep ; 14(1): 14900, 2024 06 28.
Article in English | MEDLINE | ID: mdl-38942903

ABSTRACT

Eukaryotic cells can synthesize formyl-methionine (fMet)-containing proteins not only in mitochondria but also in the cytosol to some extent. Our previous study revealed substantial upregulation of N-terminal (Nt)-fMet-containing proteins in the cytosol of SW480 colorectal cancer cells. However, the functional and pathophysiological implications remain unclear. Here, we demonstrated that removal of the Nt-formyl moiety of Nt-fMet-containing proteins (via expressing Escherichia coli PDF peptide deformylase) resulted in a dramatic increase in the proliferation of SW480 colorectal cancer cells. This proliferation coincided with the acquisition of cancer stem cell features, including reduced cell size, enhanced self-renewal capacity, and elevated levels of the cancer stem cell surface marker CD24 and pluripotent transcription factor SOX2. Furthermore, deformylation of Nt-fMet-containing proteins promoted the tumorigenicity of SW480 colorectal cancer cells in an in vivo xenograft mouse model. Taken together, these findings suggest that cytosolic deformylation has a tumor-enhancing effect, highlighting its therapeutic potential for cancer treatment.


Subject(s)
Amidohydrolases , Cell Proliferation , Cytosol , Neoplastic Stem Cells , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Cytosol/metabolism , Mice , Cell Line, Tumor , Amidohydrolases/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , CD24 Antigen/metabolism , SOXB1 Transcription Factors/metabolism , Disease Progression , Methionine/metabolism , Methionine/analogs & derivatives
7.
Appl Microbiol Biotechnol ; 108(1): 392, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38910173

ABSTRACT

In the last decades, biocatalysis has offered new perspectives for the synthesis of (chiral) amines, which are essential building blocks for pharmaceuticals, fine and bulk chemicals. In this regard, amidases have been employed due to their broad substrate scope and their independence from expensive cofactors. To expand the repertoire of amidases, tools for their rapid identification and characterization are greatly demanded. In this work an ultra-high throughput growth selection assay based on the production of the folate precursor p-aminobenzoic acid (PABA) is introduced to identify amidase activity. PABA-derived amides structurally mimic the broad class of commonly used chromogenic substrates derived from p-nitroaniline. This suggests that the assay should be broadly applicable for the identification of amidases. Unlike conventional growth selection assays that rely on substrates as nitrogen or carbon source, our approach requires PABA in sub-nanomolar concentrations, making it exceptionally sensitive and ideal for engineering campaigns that aim at enhancing amidase activities from minimally active starting points, for example. The presented assay offers flexibility in the adjustment of sensitivity to suit project-specific needs using different expression systems and fine-tuning with the antimetabolite sulfathiazole. Application of this PABA-based assay facilitates the screening of millions of enzyme variants on a single agar plate within two days, without the need for laborious sample preparation or expensive instruments, with transformation efficiency being the only limiting factor. KEY POINTS: • Ultra-high throughput assay (tens of millions on one agar plate) for amidase screening • High sensitivity by coupling selection to folate instead of carbon or nitrogen source • Highly adjustable in terms of sensitivity and expression of the engineering target.


Subject(s)
4-Aminobenzoic Acid , Amidohydrolases , High-Throughput Screening Assays , Amidohydrolases/metabolism , Amidohydrolases/genetics , High-Throughput Screening Assays/methods , 4-Aminobenzoic Acid/metabolism , 4-Aminobenzoic Acid/chemistry , Substrate Specificity , Escherichia coli/genetics , Escherichia coli/enzymology , Escherichia coli/metabolism
8.
Proc Natl Acad Sci U S A ; 121(25): e2321890121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38857388

ABSTRACT

In bacteria, attenuation of protein-tyrosine phosphorylation occurs during oxidative stress. The main described mechanism behind this effect is the H2O2-triggered conversion of bacterial phospho-tyrosines to protein-bound 3,4-dihydroxyphenylalanine. This disrupts the bacterial tyrosine phosphorylation-based signaling network, which alters the bacterial polysaccharide biosynthesis. Herein, we report an alternative mechanism, in which oxidative stress leads to a direct inhibition of bacterial protein-tyrosine kinases (BY-kinases). We show that DefA, a minor peptide deformylase, inhibits the activity of BY-kinase PtkA when Bacillus subtilis is exposed to oxidative stress. High levels of PtkA activity are known to destabilize B. subtilis pellicle formation, which leads to higher sensitivity to oxidative stress. Interaction with DefA inhibits both PtkA autophosphorylation and phosphorylation of its substrate Ugd, which is involved in exopolysaccharide formation. Inactivation of defA drastically reduces the capacity of B. subtilis to cope with oxidative stress, but it does not affect the major oxidative stress regulons PerR, OhrR, and Spx, indicating that PtkA inhibition is the main pathway for DefA involvement in this stress response. Structural analysis identified DefA residues Asn95, Tyr150, and Glu152 as essential for interaction with PtkA. Inhibition of PtkA depends also on the presence of a C-terminal α-helix of DefA, which resembles PtkA-interacting motifs from known PtkA activators, TkmA, SalA, and MinD. Loss of either the key interacting residues or the inhibitory helix of DefA abolishes inhibition of PtkA in vitro and impairs postoxidative stress recovery in vivo, confirming the involvement of these structural features in the proposed mechanism.


Subject(s)
Bacillus subtilis , Bacterial Proteins , Oxidative Stress , Bacillus subtilis/metabolism , Bacillus subtilis/genetics , Phosphorylation , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Protein-Tyrosine Kinases/metabolism , Hydrogen Peroxide/metabolism , Amidohydrolases/metabolism
9.
J Hazard Mater ; 473: 134716, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38797074

ABSTRACT

Ochratoxin A (OTA) is a toxic secondary metabolite that widely contaminates agro-products and poses a significant dietary risk to human health. Previously, a carboxypeptidase CP4 was characterized for OTA degradation in Lysobacter sp. CW239, but the degradation activity was much lower than its host strain CW239. In this study, an amidohydrolase ADH2 was screened for OTA hydrolysis in this strain. The result showed that 50 µg/L OTA was completely degraded by 1.0 µg/mL rADH2 within 5 min, indicating ultra-efficient activity. Meanwhile, the two hydrolases (i.e., CP4 and ADH2) in the strain CW239 showed the same degradation manner, which transformed the OTA to ochratoxin α (OTα) and l-ß-phenylalanine. Gene mutants (Δcp4, Δadh2 and Δcp4-adh2) testing result showed that OTA was co-degraded by carboxypeptidase CP4 and amidohydrolase ADH2, and the two hydrolases are sole agents in strain CW239 for OTA degradation. Hereinto, the ADH2 was the overwhelming efficient hydrolase, and the two types of hydrolases co-degraded OTA in CW239 by synergistic effect. The results of this study are highly significant to ochratoxin A contamination control during agro-products production and postharvest.


Subject(s)
Lysobacter , Ochratoxins , Ochratoxins/metabolism , Ochratoxins/toxicity , Lysobacter/metabolism , Lysobacter/genetics , Amidohydrolases/metabolism , Amidohydrolases/genetics , Carboxypeptidases/metabolism , Carboxypeptidases/genetics , Hydrolases/metabolism , Hydrolases/genetics
10.
Int J Biol Macromol ; 270(Pt 2): 132281, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38740150

ABSTRACT

DapE is a Zn2+-metallohydrolase recognized as a drug target for bacterial control. It is a homodimer that requires the exchange of interface strands by an induced fit essential for catalysis. Identifying novel anti-DapE agents requires greater structural details. Most of the characterized DapEs are from the Gram-negative group. Here, two high-resolution DapE crystal structures from Enterococcus faecium are presented for the first time with novel aspects. A loosened enzyme intermediate between the open and closed conformations is observed. Substrates may bind to loose state, subsequently it closes, where hydrolysis occurs, and finally, the change to the open state leads to the release of the products. Mutation of His352 suggests a role, along with His194, in the oxyanion stabilization in the mono-metalated Zn2+ isoform, while in the di-metalated isoform, the metal center 2 complements it function. An aromatic-π box potentially involved in the interaction of DapE with other proteins, and a peptide flip could determine the specificity in the Gram-positive ArgE/DapE group. Finally, details of two extra-catalytic cavities whose geometry changes depending on the conformational state of the enzyme are presented. These cavities could be a target for developing non-competitive agents that trap the enzyme in an inactive state.


Subject(s)
Bacterial Proteins , Enterococcus faecium , Enterococcus faecium/enzymology , Substrate Specificity , Ligands , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Models, Molecular , Protein Conformation , Zinc/chemistry , Zinc/metabolism , Catalytic Domain , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Amidohydrolases/genetics , Crystallography, X-Ray , Amino Acid Sequence , Protein Binding
11.
Biomacromolecules ; 25(6): 3607-3619, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38776179

ABSTRACT

Studying how synthetic polymer assemblies respond to sequential enzymatic stimuli can uncover intricate interactions in biological systems. Using amidase- and esterase-responsive PEG-based diblock (DBA) and triblock amphiphiles (TBAs), we created two distinct formulations: amidase-responsive DBA with esterase-responsive TBA and vice versa. We studied their cascade responses to the two enzymes and the sequence of their introduction. These formulations underwent cascade mesophase transitions upon the addition of the DBA-degrading enzyme, transitioning from (i) coassembled micelles to (ii) triblock-based hydrogel, and ultimately to (iii) dissolved polymers when exposed to the TBA hydrolyzing enzyme. The specific pathway of the two mesophase transitions depended on the compositions of the formulations and the enzyme introduction sequence. The results highlight the potential for designing polymeric formulations with programmable multistep enzymatic responses, mimicking the complex behavior of biological macromolecules.


Subject(s)
Polyethylene Glycols , Polyethylene Glycols/chemistry , Micelles , Esterases/chemistry , Esterases/metabolism , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Phase Transition , Polymers/chemistry , Hydrogels/chemistry
12.
mBio ; 15(6): e0094324, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38727244

ABSTRACT

Bile acids play a critical role in the emulsification of dietary lipids, a critical step in the primary function of the small intestine, which is the digestion and absorption of food. Primary bile acids delivered into the small intestine are conjugated to enhance functionality, in part, by increasing aqueous solubility and preventing passive diffusion of bile acids out of the gut lumen. Bile acid function can be disrupted by the gut microbiota via the deconjugation of primary bile acids by bile salt hydrolases (BSHs), leading to their conversion into secondary bile acids through the expression of bacterial bile acid-inducible genes, a process often observed in malabsorption due to small intestinal bacterial overgrowth. By modeling the small intestinal microbiota in vitro using human small intestinal ileostomy effluent as the inocula, we show here that the infusion of physiologically relevant levels of oxygen, normally found in the proximal small intestine, reduced deconjugation of primary bile acids, in part, through the expansion of bacterial taxa known to have a low abundance of BSHs. Further recapitulating the small intestinal bile acid composition of the small intestine, limited conversion of primary into secondary bile acids was observed. Remarkably, these effects were preserved among four separate communities, each inoculated with a different small intestinal microbiota, despite a high degree of taxonomic variability under both anoxic and aerobic conditions. In total, these results provide evidence for a previously unrecognized role that the oxygenated environment of the small intestine plays in the maintenance of normal digestive physiology. IMPORTANCE: Conjugated primary bile acids are produced by the liver and exist at high concentrations in the proximal small intestine, where they are critical for proper digestion. Deconjugation of these bile acids with subsequent transformation via dehydroxylation into secondary bile acids is regulated by the colonic gut microbiota and reduces their digestive function. Using an in vitro platform modeling the small intestinal microbiota, we analyzed the ability of this community to transform primary bile acids and studied the effect of physiological levels of oxygen normally found in the proximal small intestine (5%) on this metabolic process. We found that oxygenation of the small intestinal microbiota inhibited the deconjugation of primary bile acids in vitro. These findings suggest that luminal oxygen levels normally found in the small intestine may maintain the optimal role of bile acids in the digestive process by regulating bile acid conversion by the gut microbiota.


Subject(s)
Bile Acids and Salts , Gastrointestinal Microbiome , Intestine, Small , Oxygen , Bile Acids and Salts/metabolism , Humans , Intestine, Small/microbiology , Intestine, Small/metabolism , Oxygen/metabolism , Bacteria/metabolism , Bacteria/genetics , Bacteria/classification , Amidohydrolases
13.
Drug Metab Pharmacokinet ; 56: 101007, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38797091

ABSTRACT

Arylacetamide deacetylase (AADAC) is involved in drug hydrolysis and lipid metabolism. In 23 human liver samples, no significant correlation was observed between AADAC mRNA (19.7-fold variation) and protein levels (137.6-fold variation), suggesting a significant contribution of post-transcriptional regulation to AADAC expression. The present study investigated whether AADAC is regulated by microRNA in the human liver and elucidate its biological significance. Computational analysis predicted two potential miR-222-3p recognition elements in the 3'-untranslated region (UTR) of AADAC. Luciferase assay revealed that the miR-222-3p recognition element was functional in downregulating AADAC expression. In HEK293 cells transfected with an AADAC expression plasmid containing 3'-UTR, miR-222-3p overexpression decreased AADAC protein level and activity, whereas miR-222-3p inhibition increased them. Similar results were observed in human hepatoma-derived Huh-1 cells endogenously expressing AADAC and HepaSH cells that are hepatocytes from chimeric mice with humanized livers. In individual human liver samples, AADAC protein levels inversely correlated with miR-222-3p levels. Overexpression of miR-222-3p resulted in increased lipid accumulation in Huh-1 cells, which was reversed by AADAC overexpression. In contrast, miR-222-3p inhibition decreased lipid accumulation, which was reversed by AADAC knockdown. In conclusion, we found that hepatic AADAC was downregulated by miR-222-3p, resulting in decreased drug hydrolysis and increased lipid accumulation.


Subject(s)
Down-Regulation , Lipid Metabolism , MicroRNAs , Animals , Humans , Mice , Amidohydrolases/metabolism , Amidohydrolases/genetics , Carboxylic Ester Hydrolases , HEK293 Cells , Hydrolysis , Lipid Metabolism/genetics , Liver/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism
14.
Acta Biochim Pol ; 71: 12299, 2024.
Article in English | MEDLINE | ID: mdl-38721302

ABSTRACT

This report describes a comprehensive approach to local random mutagenesis of the E. coli Ntn-amidohydrolase EcAIII, and supplements the results published earlier for the randomization series RDM1. Here, random mutagenesis was applied in the center of the EcAIII molecule, i.e., in the region important for substrate binding and its immediate neighborhood (series RDM2, RDM3, RDM7), in the vicinity of the catalytic threonine triplet (series RDM4, RDM5, RDM6), in the linker region (series RDM8), and in the sodium-binding (stabilization) loop (series RDM9). The results revealed that the majority of the new EcAIII variants have abolished or significantly reduced rate of autoprocessing, even if the mutation was not in a highly conserved sequence and structure regions. AlphaFold-predicted structures of the mutants suggest the role of selected residues in the positioning of the linker and stabilization of the scissile bond in precisely correct orientation, enabling the nucleophilic attack during the maturation process. The presented data highlight the details of EcAIII geometry that are important for the autoproteolytic maturation and for the catalytic mechanism in general, and can be treated as a guide for protein engineering experiments with other Ntn-hydrolases.


Subject(s)
Amidohydrolases , Escherichia coli , Mutagenesis , Amidohydrolases/genetics , Amidohydrolases/metabolism , Amidohydrolases/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Models, Molecular , Amino Acid Sequence , Mutation
15.
Cells ; 13(10)2024 May 13.
Article in English | MEDLINE | ID: mdl-38786051

ABSTRACT

The inhibition of endocannabinoid hydrolysis by enzymatic inhibitors may interfere with mechanisms underlying migraine-related pain. The dual FAAH/MAGL inhibitor AKU-005 shows potent inhibitory activity in vitro. Here, we assessed the effect of AKU-005 in a migraine animal model based on nitroglycerin (NTG) administration. Male rats were treated with AKU-005 (0.5 mg/kg, i.p.) or vehicle 3 h after receiving NTG (10 mg/kg, i.p.) or NTG vehicle. One hour later, rats were subjected to the open field test followed by the orofacial formalin test. At the end of the test, we collected serum samples for assessing calcitonin gene-related peptide (CGRP) levels as well as meninges, trigeminal ganglia, and brain areas to assess mRNA levels of CGRP and pro-inflammatory cytokines, and endocannabinoid and related lipid levels. AKU-005 reduced NTG-induced hyperalgesia during the orofacial formalin test but did not influence NTG-induced changes in the open field test. It significantly reduced serum levels of CGRP, CGRP, and pro-inflammatory cytokine mRNA levels in the meninges, trigeminal ganglia, and central areas. Surprisingly, AKU-005 caused no change in endocannabinoids and related lipids in the regions evaluated. The present findings suggest that AKU-005 may have anti-migraine effects by reducing CGRP synthesis and release and the associated inflammatory events. This effect, however, does not seem mediated via an interference with the endocannabinoid pathway.


Subject(s)
Amidohydrolases , Calcitonin Gene-Related Peptide , Hyperalgesia , Trigeminal Ganglion , Animals , Male , Hyperalgesia/drug therapy , Rats , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Amidohydrolases/genetics , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/blood , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/metabolism , Rats, Sprague-Dawley , Monoacylglycerol Lipases/antagonists & inhibitors , Monoacylglycerol Lipases/metabolism , Endocannabinoids/metabolism , Nitroglycerin/pharmacology , Disease Models, Animal , Cytokines/metabolism , Cytokines/blood , Migraine Disorders/drug therapy , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Oligopeptides , Salivary Proteins and Peptides
16.
Sci Rep ; 14(1): 10952, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38740850

ABSTRACT

It is recognized as a promising therapeutic strategy for cocaine use disorder to develop an efficient enzyme which can rapidly convert cocaine to physiologically inactive metabolites. We have designed and discovered a series of highly efficient cocaine hydrolases, including CocH5-Fc(M6) which is the currently known as the most efficient cocaine hydrolase with both the highest catalytic activity against (-)-cocaine and the longest biological half-life in rats. In the present study, we characterized the time courses of protein appearance, pH, structural integrity, and catalytic activity against cocaine in vitro and in vivo of a CocH5-Fc(M6) bulk drug substance produced in a bioreactor for its in vitro and in vivo stability after long-time storage under various temperatures (- 80, - 20, 4, 25, or 37 °C). Specifically, all the tested properties of the CocH5-Fc(M6) protein did not significantly change after the protein was stored at any of four temperatures including - 80, - 20, 4, and 25 °C for ~ 18 months. In comparison, at 37 °C, the protein was less stable, with a half-life of ~ 82 days for cocaine hydrolysis activity. Additionally, the in vivo studies further confirmed the linear elimination PK profile of CocH5-Fc(M6) with an elimination half-life of ~ 9 days. All the in vitro and in vivo data on the efficacy and stability of CocH5-Fc(M6) have consistently demonstrated that CocH5-Fc(M6) has the desired in vitro and in vivo stability as a promising therapeutic candidate for treatment of cocaine use disorder.


Subject(s)
Cocaine , Enzyme Stability , Animals , Cocaine/metabolism , Rats , Hydrolysis , Hydrogen-Ion Concentration , Male , Half-Life , Temperature , Amidohydrolases/metabolism , Carboxylic Ester Hydrolases , Recombinant Proteins
17.
Biomed Pharmacother ; 175: 116677, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38701570

ABSTRACT

The current pharmacological approaches to multiple sclerosis (MS) target its inflammatory and autoimmune components, but effective treatments to foster remyelination and axonal repair are still lacking. We therefore selected two targets known to be involved in MS pathogenesis: N-acylethanolamine-hydrolyzing acid amidase (NAAA) and glycogen synthase kinase-3ß (GSK-3ß). We tested whether inhibiting these targets exerted a therapeutic effect against experimental autoimmune encephalomyelitis (EAE), an animal model of MS. The combined inhibition of NAAA and GSK-3ß by two selected small-molecule compounds, ARN16186 (an NAAA inhibitor) and AF3581 (a GSK-3ß inhibitor), effectively mitigated disease progression, rescuing the animals from paralysis and preventing a worsening of the pathology. The complementary activity of the two inhibitors reduced the infiltration of immune cells into the spinal cord and led to the formation of thin myelin sheaths around the axons post-demyelination. Specifically, the inhibition of NAAA and GSK-3ß modulated the over-activation of NF-kB and STAT3 transcription factors in the EAE-affected mice and induced the nuclear translocation of ß-catenin, reducing the inflammatory insult and promoting the remyelination process. Overall, this work demonstrates that the dual-targeting of key aspects responsible for MS progression could be an innovative pharmacological approach to tackle the pathology.


Subject(s)
Amidohydrolases , Encephalomyelitis, Autoimmune, Experimental , Glycogen Synthase Kinase 3 beta , Mice, Inbred C57BL , Multiple Sclerosis , Animals , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Mice , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Female , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , NF-kappa B/metabolism , Enzyme Inhibitors/pharmacology , Myelin Sheath/metabolism , Myelin Sheath/drug effects
18.
Article in English | MEDLINE | ID: mdl-38765527

ABSTRACT

Objective: To examine whether the DDAH2 promoter polymorphisms -1415G/A (rs2272592), -1151A/C (rs805304) and -449G/C (rs805305), and their haplotypes, are associated with PE compared with normotensive pregnant women, and whether they affect ADMA levels in these groups. Methods: A total of 208 pregnant women were included in the study and classified as early-onset (N=57) or late-onset PE (N =49), and as normotensive pregnant women (N = 102). Results: Pregnant with early-onset PE carrying the GC and GG genotypes for the DDAH2 -449G/C polymorphism had increased ADMA levels (P=0.01). No association of DDAH2 polymorphisms with PE in single-locus analysis was found. However, the G-C-G haplotype was associated with the risk for late-onset PE. Conclusion: It is suggested that DDAH2 polymorphisms could affect ADMA levels in PE, and that DDAH2 haplotypes may affect the risk for PE.


Subject(s)
Amidohydrolases , Arginine , Haplotypes , Polymorphism, Genetic , Pre-Eclampsia , Humans , Female , Amidohydrolases/genetics , Pre-Eclampsia/genetics , Pre-Eclampsia/blood , Pregnancy , Adult , Arginine/analogs & derivatives , Arginine/blood , Arginine/genetics , Young Adult
19.
Mar Drugs ; 22(5)2024 May 07.
Article in English | MEDLINE | ID: mdl-38786603

ABSTRACT

Naturally occurring echinocandin B and FR901379 are potent antifungal lipopeptides featuring a cyclic hexapeptide nucleus and a fatty acid side chain. They are the parent compounds of echinocandin drugs for the treatment of severe fungal infections caused by the Candida and Aspergilla species. To minimize hemolytic toxicity, the native fatty acid side chains in these drug molecules are replaced with designer acyl side chains. The deacylation of the N-acyl side chain is, therefore, a crucial step for the development and manufacturing of echinocandin-type antibiotics. Echinocandin E (ECE) is a novel echinocandin congener with enhanced stability generated via the engineering of the biosynthetic machinery of echinocandin B (ECB). In the present study, we report the discovery of the first echinocandin E acylase (ECEA) using the enzyme similarity tool (EST) for enzymatic function mining across protein families. ECEA is derived from Streptomyces sp. SY1965 isolated from a sediment collected from the Mariana Trench. It was cloned and heterologously expressed in S. lividans TK24. The resultant TKecea66 strain showed efficient cleavage activity of the acyl side chain of ECE, showing promising applications in the development of novel echinocandin-type therapeutics. Our results also provide a showcase for harnessing the essentially untapped biodiversity from the hadal ecosystems for the discovery of functional molecules.


Subject(s)
Antifungal Agents , Echinocandins , Streptomyces , Streptomyces/enzymology , Streptomyces/genetics , Echinocandins/chemistry , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Amidohydrolases/metabolism , Fungal Proteins
20.
Toxins (Basel) ; 16(5)2024 May 09.
Article in English | MEDLINE | ID: mdl-38787069

ABSTRACT

The fungal cell wall serves as the primary interface between fungi and their external environment, providing protection and facilitating interactions with the surroundings. Chitin is a vital structural element in fungal cell wall. Chitin deacetylase (CDA) can transform chitin into chitosan through deacetylation, providing various biological functions across fungal species. Although this modification is widespread in fungi, the biological functions of CDA enzymes in Aspergillus flavus remain largely unexplored. In this study, we aimed to investigate the biofunctions of the CDA family in A. flavus. The A. flavus genome contains six annotated putative chitin deacetylases. We constructed knockout strains targeting each member of the CDA family, including Δcda1, Δcda2, Δcda3, Δcda4, Δcda5, and Δcda6. Functional analyses revealed that the deletion of CDA family members neither significantly affects the chitin content nor exhibits the expected chitin deacetylation function in A. flavus. However, the Δcda6 strain displayed distinct phenotypic characteristics compared to the wild-type (WT), including an increased conidia count, decreased mycelium production, heightened aflatoxin production, and impaired seed colonization. Subcellular localization experiments indicated the cellular localization of CDA6 protein within the cell wall of A. flavus filaments. Moreover, our findings highlight the significance of the CBD1 and CBD2 structural domains in mediating the functional role of the CDA6 protein. Overall, we analyzed the gene functions of CDA family in A. flavus, which contribute to a deeper understanding of the mechanisms underlying aflatoxin contamination and lay the groundwork for potential biocontrol strategies targeting A. flavus.


Subject(s)
Aflatoxins , Amidohydrolases , Aspergillus flavus , Aspergillus flavus/genetics , Aspergillus flavus/enzymology , Aspergillus flavus/metabolism , Amidohydrolases/genetics , Amidohydrolases/metabolism , Aflatoxins/biosynthesis , Aflatoxins/metabolism , Aflatoxins/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Chitin/metabolism , Cell Wall/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL