Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.786
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 21(7): 727-735, 2020 07.
Article in English | MEDLINE | ID: mdl-32541831

ABSTRACT

Stimulator-of-interferon genes (STING) is vital for sensing cytosolic DNA and initiating innate immune responses against microbial infection and tumors. Redox homeostasis is the balance of oxidative and reducing reactions present in all living systems. Yet, how the intracellular redox state controls STING activation is unclear. Here, we show that cellular redox homeostasis maintained by glutathione peroxidase 4 (GPX4) is required for STING activation. GPX4 deficiency enhanced cellular lipid peroxidation and thus specifically inhibited the cGAS-STING pathway. Concordantly, GPX4 deficiency inhibited herpes simplex virus-1 (HSV-1)-induced innate antiviral immune responses and promoted HSV-1 replication in vivo. Mechanistically, GPX4 inactivation increased production of lipid peroxidation, which led to STING carbonylation at C88 and inhibited its trafficking from the endoplasmic reticulum (ER) to the Golgi complex. Thus, cellular stress-induced lipid peroxidation specifically attenuates the STING DNA-sensing pathway, suggesting that GPX4 facilitates STING activation by maintaining redox homeostasis of lipids.


Subject(s)
Herpes Simplex/immunology , Membrane Proteins/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Animals , Carbolines/pharmacology , Cells, Cultured , DNA, Viral/immunology , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Female , Fibroblasts , Golgi Apparatus/metabolism , HEK293 Cells , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Homeostasis/immunology , Humans , Immunity, Innate , Lipid Peroxidation/genetics , Lipid Peroxidation/immunology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Membrane Proteins/immunology , Mice , Mice, Knockout , Nucleotidyltransferases/metabolism , Oxidation-Reduction , Oximes/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Primary Cell Culture , Protein Carbonylation/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Sulfonamides/pharmacology , THP-1 Cells , Virus Replication/immunology
2.
Nature ; 629(8013): 824-829, 2024 May.
Article in English | MEDLINE | ID: mdl-38720081

ABSTRACT

Enzymes play an increasingly important role in improving the benignity and efficiency of chemical production, yet the diversity of their applications lags heavily behind chemical catalysts as a result of the relatively narrow range of reaction mechanisms of enzymes. The creation of enzymes containing non-biological functionalities facilitates reaction mechanisms outside nature's canon and paves the way towards fully programmable biocatalysis1-3. Here we present a completely genetically encoded boronic-acid-containing designer enzyme with organocatalytic reactivity not achievable with natural or engineered biocatalysts4,5. This boron enzyme catalyses the kinetic resolution of hydroxyketones by oxime formation, in which crucial interactions with the protein scaffold assist in the catalysis. A directed evolution campaign led to a variant with natural-enzyme-like enantioselectivities for several different substrates. The unique activation mode of the boron enzyme was confirmed using X-ray crystallography, high-resolution mass spectrometry (HRMS) and 11B NMR spectroscopy. Our study demonstrates that genetic-code expansion can be used to create evolvable enantioselective enzymes that rely on xenobiotic catalytic moieties such as boronic acids and access reaction mechanisms not reachable through catalytic promiscuity of natural or engineered enzymes.


Subject(s)
Biocatalysis , Boronic Acids , Enzymes , Protein Engineering , Boronic Acids/chemistry , Boronic Acids/metabolism , Crystallography, X-Ray , Directed Molecular Evolution , Enzymes/chemistry , Enzymes/metabolism , Enzymes/genetics , Ketones/chemistry , Ketones/metabolism , Kinetics , Models, Molecular , Oximes/chemistry , Oximes/metabolism , Substrate Specificity , Nuclear Magnetic Resonance, Biomolecular , Mass Spectrometry , Xenobiotics/chemistry , Xenobiotics/metabolism
3.
Mol Cell ; 81(11): 2290-2302.e7, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33831358

ABSTRACT

Cancer cells adapt their metabolism to support elevated energetic and anabolic demands of proliferation. Folate-dependent one-carbon metabolism is a critical metabolic process underpinning cellular proliferation supplying carbons for the synthesis of nucleotides incorporated into DNA and RNA. Recent research has focused on the nutrients that supply one-carbons to the folate cycle, particularly serine. Tryptophan is a theoretical source of one-carbon units through metabolism by IDO1, an enzyme intensively investigated in the context of tumor immune evasion. Using in vitro and in vivo pancreatic cancer models, we show that IDO1 expression is highly context dependent, influenced by attachment-independent growth and the canonical activator IFNγ. In IDO1-expressing cancer cells, tryptophan is a bona fide one-carbon donor for purine nucleotide synthesis in vitro and in vivo. Furthermore, we show that cancer cells release tryptophan-derived formate, which can be used by pancreatic stellate cells to support purine nucleotide synthesis.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Pancreatic Neoplasms/genetics , Pancreatic Stellate Cells/metabolism , Tumor Escape/drug effects , Allografts , Animals , Antineoplastic Agents/pharmacology , Carbon/immunology , Carbon/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/mortality , Cell Line, Tumor , Formates/immunology , Formates/metabolism , Gene Expression Regulation, Neoplastic , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Mice, Nude , Oximes/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/immunology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/immunology , Serine/immunology , Serine/metabolism , Serine/pharmacology , Signal Transduction , Sulfonamides/pharmacology , Tryptophan/immunology , Tryptophan/metabolism , Tryptophan/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
4.
PLoS Genet ; 20(3): e1011179, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38437227

ABSTRACT

Recent human genome-wide association studies have identified common missense variants in MARC1, p.Ala165Thr and p.Met187Lys, associated with lower hepatic fat, reduction in liver enzymes and protection from most causes of cirrhosis. Using an exome-wide association study we recapitulated earlier MARC1 p.Ala165Thr and p.Met187Lys findings in 540,000 individuals from five ancestry groups. We also discovered novel rare putative loss of function variants in MARC1 with a phenotype similar to MARC1 p.Ala165Thr/p.Met187Lys variants. In vitro studies of recombinant human MARC1 protein revealed Ala165Thr substitution causes protein instability and aberrant localization in hepatic cells, suggesting MARC1 inhibition or deletion may lead to hepatoprotection. Following this hypothesis, we generated Marc1 knockout mice and evaluated the effect of Marc1 deletion on liver phenotype. Unexpectedly, our study found that whole-body Marc1 deficiency in mouse is not protective against hepatic triglyceride accumulation, liver inflammation or fibrosis. In attempts to explain the lack of the observed phenotype, we discovered that Marc1 plays only a minor role in mouse liver while its paralogue Marc2 is the main Marc family enzyme in mice. Our findings highlight the major difference in MARC1 physiological function between human and mouse.


Subject(s)
Genome-Wide Association Study , Oximes , Animals , Humans , Mice , Liver Cirrhosis
5.
Genes Dev ; 33(17-18): 1236-1251, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31416966

ABSTRACT

Tumors display increased uptake and processing of nutrients to fulfill the demands of rapidly proliferating cancer cells. Seminal studies have shown that the proto-oncogene MYC promotes metabolic reprogramming by altering glutamine uptake and metabolism in cancer cells. How MYC regulates the metabolism of other amino acids in cancer is not fully understood. Using high-performance liquid chromatography (HPLC)-tandem mass spectrometry (LC-MS/MS), we found that MYC increased intracellular levels of tryptophan and tryptophan metabolites in the kynurenine pathway. MYC induced the expression of the tryptophan transporters SLC7A5 and SLC1A5 and the enzyme arylformamidase (AFMID), involved in the conversion of tryptophan into kynurenine. SLC7A5, SLC1A5, and AFMID were elevated in colon cancer cells and tissues, and kynurenine was significantly greater in tumor samples than in the respective adjacent normal tissue from patients with colon cancer. Compared with normal human colonic epithelial cells, colon cancer cells were more sensitive to the depletion of tryptophan. Blocking enzymes in the kynurenine pathway caused preferential death of established colon cancer cells and transformed colonic organoids. We found that only kynurenine and no other tryptophan metabolite promotes the nuclear translocation of the transcription factor aryl hydrocarbon receptor (AHR). Blocking the interaction between AHR and kynurenine with CH223191 reduced the proliferation of colon cancer cells. Therefore, we propose that limiting cellular kynurenine or its downstream targets could present a new strategy to reduce the proliferation of MYC-dependent cancer cells.


Subject(s)
Colonic Neoplasms/physiopathology , Kynurenine/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tryptophan/metabolism , Amino Acid Transport System ASC/genetics , Antineoplastic Agents/pharmacology , Arylformamidase/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Humans , Indoles/pharmacology , Kynurenine/genetics , Large Neutral Amino Acid-Transporter 1/genetics , Minor Histocompatibility Antigens/genetics , Oximes/pharmacology , Proto-Oncogene Mas , Sulfonamides/pharmacology
6.
Genes Dev ; 33(17-18): 1252-1264, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31395740

ABSTRACT

Although MAX is regarded as an obligate dimerization partner for MYC, its function in normal development and neoplasia is poorly defined. We show that B-cell-specific deletion of Max has a modest effect on B-cell development but completely abrogates Eµ-Myc-driven lymphomagenesis. While Max loss affects only a few hundred genes in normal B cells, it leads to the global down-regulation of Myc-activated genes in premalignant Eµ-Myc cells. We show that the balance between MYC-MAX and MNT-MAX interactions in B cells shifts in premalignant B cells toward a MYC-driven transcriptional program. Moreover, we found that MAX loss leads to a significant reduction in MYC protein levels and down-regulation of direct transcriptional targets, including regulators of MYC stability. This phenomenon is also observed in multiple cell lines treated with MYC-MAX dimerization inhibitors. Our work uncovers a layer of Myc autoregulation critical for lymphomagenesis yet partly dispensable for normal development.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Lymphoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Active Transport, Cell Nucleus , Animals , Carcinogenesis/drug effects , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Gene Deletion , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Kynurenine/genetics , Kynurenine/metabolism , Lymphoma/physiopathology , Mice , Organoids/growth & development , Organoids/physiopathology , Oximes/pharmacology , Sulfonamides/pharmacology
7.
Nature ; 585(7823): 141-145, 2020 09.
Article in English | MEDLINE | ID: mdl-32641835

ABSTRACT

The transient receptor potential ion channel TRPA1 is expressed by primary afferent nerve fibres, in which it functions as a low-threshold sensor for structurally diverse electrophilic irritants, including small volatile environmental toxicants and endogenous algogenic lipids1. TRPA1 is also a 'receptor-operated' channel whose activation downstream of metabotropic receptors elicits inflammatory pain or itch, making it an attractive target for novel analgesic therapies2. However, the mechanisms by which TRPA1 recognizes and responds to electrophiles or cytoplasmic second messengers remain unknown. Here we use strutural studies and electrophysiology to show that electrophiles act through a two-step process in which modification of a highly reactive cysteine residue (C621) promotes reorientation of a cytoplasmic loop to enhance nucleophilicity and modification of a nearby cysteine (C665), thereby stabilizing the loop in an activating configuration. These actions modulate two restrictions controlling ion permeation, including widening of the selectivity filter to enhance calcium permeability and opening of a canonical gate at the cytoplasmic end of the pore. We propose a model to explain functional coupling between electrophile action and these control points. We also characterize a calcium-binding pocket that is highly conserved across TRP channel subtypes and accounts for all aspects of calcium-dependent TRPA1 regulation, including potentiation, desensitization and activation by metabotropic receptors. These findings provide a structural framework for understanding how a broad-spectrum irritant receptor is controlled by endogenous and exogenous agents that elicit or exacerbate pain and itch.


Subject(s)
Calcium/metabolism , Calcium/pharmacology , Ion Channel Gating/drug effects , TRPA1 Cation Channel/chemistry , TRPA1 Cation Channel/metabolism , Amino Acid Sequence , Cysteine/metabolism , Electric Conductivity , Humans , Iodoacetamide/pharmacology , Models, Molecular , Mutation , Oximes/pharmacology , TRPA1 Cation Channel/genetics
8.
Circulation ; 150(4): 302-316, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38695173

ABSTRACT

BACKGROUND: The ubiquitin-proteasome system regulates protein degradation and the development of pulmonary arterial hypertension (PAH), but knowledge about the role of deubiquitinating enzymes in this process is limited. UCHL1 (ubiquitin carboxyl-terminal hydrolase 1), a deubiquitinase, has been shown to reduce AKT1 (AKT serine/threonine kinase 1) degradation, resulting in higher levels. Given that AKT1 is pathological in pulmonary hypertension, we hypothesized that UCHL1 deficiency attenuates PAH development by means of reductions in AKT1. METHODS: Tissues from animal pulmonary hypertension models as well as human pulmonary artery endothelial cells from patients with PAH exhibited increased vascular UCHL1 staining and protein expression. Exposure to LDN57444, a UCHL1-specific inhibitor, reduced human pulmonary artery endothelial cell and smooth muscle cell proliferation. Across 3 preclinical PAH models, LDN57444-exposed animals, Uchl1 knockout rats (Uchl1-/-), and conditional Uchl1 knockout mice (Tie2Cre-Uchl1fl/fl) demonstrated reduced right ventricular hypertrophy, right ventricular systolic pressures, and obliterative vascular remodeling. Lungs and pulmonary artery endothelial cells isolated from Uchl1-/- animals exhibited reduced total and activated Akt with increased ubiquitinated Akt levels. UCHL1-silenced human pulmonary artery endothelial cells displayed reduced lysine(K)63-linked and increased K48-linked AKT1 levels. RESULTS: Supporting experimental data, we found that rs9321, a variant in a GC-enriched region of the UCHL1 gene, is associated with reduced methylation (n=5133), increased UCHL1 gene expression in lungs (n=815), and reduced cardiac index in patients (n=796). In addition, Gadd45α (an established demethylating gene) knockout mice (Gadd45α-/-) exhibited reduced lung vascular UCHL1 and AKT1 expression along with attenuated hypoxic pulmonary hypertension. CONCLUSIONS: Our findings suggest that UCHL1 deficiency results in PAH attenuation by means of reduced AKT1, highlighting a novel therapeutic pathway in PAH.


Subject(s)
Mice, Knockout , Proto-Oncogene Proteins c-akt , Ubiquitin Thiolesterase , Animals , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/deficiency , Ubiquitin Thiolesterase/metabolism , Humans , Mice , Rats , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Male , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/enzymology , Rats, Sprague-Dawley , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/etiology , Vascular Remodeling , Cells, Cultured , Cell Proliferation , Mice, Inbred C57BL , Indoles , Oximes
9.
J Virol ; 98(8): e0061824, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39023323

ABSTRACT

Zika virus (ZIKV) is a re-emerging mosquito-borne flavivirus that has been associated with congenital neurological defects in fetuses born to infected mothers. At present, no vaccine or antiviral therapy is available to combat this devastating disease. Repurposing drugs that target essential host factors exploited by viruses is an attractive therapeutic approach. Here, we screened a panel of clinically approved small-molecule kinase inhibitors for their antiviral effects against a clinical isolate of ZIKV and thoroughly characterized their mechanisms of action. We found that the Raf kinase inhibitors Dabrafenib and Regorafenib potently impair the replication of ZIKV, but not that of its close relative dengue virus. Time-of-addition experiments showed that both inhibitors target ZIKV infection at post-entry steps. We found that Dabrafenib, but not Regorafenib, interfered with ZIKV genome replication by impairing both negative- and positive-strand RNA synthesis. Regorafenib, on the other hand, altered steady-state viral protein levels, viral egress, and blocked NS1 secretion. We also observed Regorafenib-induced ER fragmentation in ZIKV-infected cells, which might contribute to its antiviral effects. Because these inhibitors target different steps of the ZIKV infection cycle, their use in combination therapy may amplify their antiviral effects which could be further explored for future therapeutic strategies against ZIKV and possibly other flaviviruses. IMPORTANCE: There is an urgent need to develop effective therapeutics against re-emerging arboviruses associated with neurological disorders like Zika virus (ZIKV). We identified two FDA-approved kinase inhibitors, Dabrafenib and Regorafenib, as potent inhibitors of contemporary ZIKV strains at distinct stages of infection despite overlapping host targets. Both inhibitors reduced viral titers by ~1 to 2 log10 (~10-fold to 100-fold) with minimal cytotoxicity. Furthermore, we show that Dabrafenib inhibits ZIKV RNA replication whereas Regorafenib inhibits ZIKV translation and egress. Regorafenib has the added benefit of limiting NS1 secretion, which contributes to the pathogenesis and disease progression of several flaviviruses. Because these inhibitors affect distinct post-entry steps of ZIKV infection, their therapeutic potential may be amplified by combination therapy and likely does not require prophylactic administration. This study provides further insight into ZIKV-host interactions and has implications for the development of novel antivirals against ZIKV and possibly other flaviviruses.


Subject(s)
Antiviral Agents , Imidazoles , Oximes , Phenylurea Compounds , Protein Kinase Inhibitors , Pyridines , Virus Replication , Zika Virus Infection , Zika Virus , Virus Replication/drug effects , Oximes/pharmacology , Zika Virus/drug effects , Pyridines/pharmacology , Humans , Imidazoles/pharmacology , Zika Virus Infection/virology , Zika Virus Infection/drug therapy , Chlorocebus aethiops , Animals , Phenylurea Compounds/pharmacology , Vero Cells , Antiviral Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Cell Line , Dengue Virus/drug effects
10.
Bioinformatics ; 40(Supplement_1): i160-i168, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38940147

ABSTRACT

MOTIVATION: Predicting cancer drug response requires a comprehensive assessment of many mutations present across a tumor genome. While current drug response models generally use a binary mutated/unmutated indicator for each gene, not all mutations in a gene are equivalent. RESULTS: Here, we construct and evaluate a series of predictive models based on leading methods for quantitative mutation scoring. Such methods include VEST4 and CADD, which score the impact of a mutation on gene function, and CHASMplus, which scores the likelihood a mutation drives cancer. The resulting predictive models capture cellular responses to dabrafenib, which targets BRAF-V600 mutations, whereas models based on binary mutation status do not. Performance improvements generalize to other drugs, extending genetic indications for PIK3CA, ERBB2, EGFR, PARP1, and ABL1 inhibitors. Introducing quantitative mutation features in drug response models increases performance and mechanistic understanding. AVAILABILITY AND IMPLEMENTATION: Code and example datasets are available at https://github.com/pgwall/qms.


Subject(s)
Antineoplastic Agents , Mutation , Neoplasms , Humans , Neoplasms/genetics , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Imidazoles/pharmacology , Oximes/pharmacology , Computational Biology/methods
11.
Blood ; 141(9): 996-1006, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36108341

ABSTRACT

BRAF V600E is the key oncogenic driver mutation in hairy cell leukemia (HCL). We report the efficacy and safety of dabrafenib plus trametinib in patients with relapsed/refractory BRAF V600E mutation-positive HCL. This open-label, phase 2 study enrolled patients with BRAF V600E mutation-positive HCL refractory to first-line treatment with a purine analog or relapsed after ≥2 prior lines of treatment. Patients received dabrafenib 150 mg twice daily plus trametinib 2 mg once daily until disease progression, unacceptable toxicity, or death. The primary endpoint was investigator-assessed objective response rate (ORR) per criteria adapted from National Comprehensive Cancer Network-Consensus Resolution guidelines. Secondary endpoints included duration of response (DOR), progression-free survival (PFS), overall survival (OS), and safety. Fifty-five patients with BRAF V600E mutation-positive HCL were enrolled. The investigator-assessed ORR was 89.0% (95% confidence interval, 77.8%-95.9%); 65.5% of patients had a complete response (without minimal residual disease [MRD]: 9.1% [negative immunohistochemistry of bone marrow {BM} biopsy], 12.7% [negative BM aspirate flow cytometry {FC}], 16.4% [negative immunohistochemistry and/or FC results]; with MRD, 49.1%), and 23.6% had a partial response. The 24-month DOR was 97.7% with 24-month PFS and OS rates of 94.4% and 94.5%, respectively. The most common treatment-related adverse events were pyrexia (58.2%), chills (47.3%), and hyperglycemia (40.0%). Dabrafenib plus trametinib demonstrated durable responses with a manageable safety profile consistent with previous observations in other indications and should be considered as a rituximab-free therapeutic option for patients with relapsed/refractory BRAF V600E mutation-positive HCL. This trial is registered at www.clinicaltrials.gov as #NCT02034110.


Subject(s)
Leukemia, Hairy Cell , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Leukemia, Hairy Cell/drug therapy , Leukemia, Hairy Cell/genetics , Pyridones/adverse effects , Pyrimidinones/adverse effects , Oximes/adverse effects , Mutation , Antineoplastic Combined Chemotherapy Protocols/adverse effects
12.
Plant J ; 116(1): 187-200, 2023 10.
Article in English | MEDLINE | ID: mdl-37366635

ABSTRACT

Phenylpropanoids are specialized metabolites derived from phenylalanine. Glucosinolates are defense compounds derived mainly from methionine and tryptophan in Arabidopsis. It was previously shown that the phenylpropanoid pathway and glucosinolate production are metabolically linked. The accumulation of indole-3-acetaldoxime (IAOx), the precursor of tryptophan-derived glucosinolates, represses phenylpropanoid biosynthesis through accelerated degradation of phenylalanine ammonia lyase (PAL). As PAL functions at the entry point of the phenylpropanoid pathway, which produces indispensable specialized metabolites such as lignin, aldoxime-mediated phenylpropanoid repression is detrimental to plant survival. Although methionine-derived glucosinolates in Arabidopsis are abundant, any impact of aliphatic aldoximes (AAOx) derived from aliphatic amino acids such as methionine on phenylpropanoid production remains unclear. Here, we investigate the impact of AAOx accumulation on phenylpropanoid production using Arabidopsis aldoxime mutants, ref2 and ref5. REF2 and REF5 metabolize aldoximes to respective nitrile oxides redundantly, but with different substrate specificities. ref2 and ref5 mutants have decreased phenylpropanoid contents due to the accumulation of aldoximes. As REF2 and REF5 have high substrate specificity toward AAOx and IAOx, respectively, it was assumed that ref2 accumulates AAOx, not IAOx. Our study indicates that ref2 accumulates both AAOx and IAOx. Removing IAOx partially restored phenylpropanoid content in ref2, but not to the wild-type level. However, when AAOx biosynthesis was silenced, phenylpropanoid production and PAL activity in ref2 were completely restored, suggesting an inhibitory effect of AAOx on phenylpropanoid production. Further feeding studies revealed that the abnormal growth phenotype commonly observed in Arabidopsis mutants lacking AAOx production is a consequence of methionine accumulation.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Glucosinolates/metabolism , Tryptophan/metabolism , Oximes/metabolism , Phenylalanine Ammonia-Lyase/metabolism , Plant Development , Methionine/metabolism , Gene Expression Regulation, Plant
13.
J Neurochem ; 168(4): 355-369, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37429600

ABSTRACT

This review presents recent studies of the chemical and molecular regulators of acetylcholine (ACh) signaling and the complexity of the small molecule and RNA regulators of those mechanisms that control cholinergic functioning in health and disease. The underlying structural, neurochemical, and transcriptomic concepts, including basic and translational research and clinical studies, shed new light on how these processes inter-change under acute states, age, sex, and COVID-19 infection; all of which modulate ACh-mediated processes and inflammation in women and men and under diverse stresses. The aspect of organophosphorus (OP) compound toxicity is discussed based on the view that despite numerous studies, acetylcholinesterase (AChE) is still a vulnerable target in OP poisoning because of a lack of efficient treatment and the limitations of oxime-assisted reactivation of inhibited AChE. The over-arching purpose of this review is thus to discuss mechanisms of cholinergic signaling dysfunction caused by OP pesticides, OP nerve agents, and anti-cholinergic medications; and to highlight new therapeutic strategies to combat both the acute and chronic effects of these chemicals on the cholinergic and neuroimmune systems. Furthermore, OP toxicity was examined in view of cholinesterase inhibition and beyond in order to highlight improved small molecules and RNA therapeutic strategies and assess their predicted pitfalls to reverse the acute toxicity and long-term deleterious effects of OPs.


Subject(s)
Cholinesterase Reactivators , Female , Humans , Cholinesterase Reactivators/chemistry , Cholinesterase Inhibitors/pharmacology , Acetylcholinesterase/chemistry , Organophosphorus Compounds , Oximes/chemistry , Oximes/pharmacology , Oximes/therapeutic use , Acetylcholine , RNA
14.
J Neurochem ; 168(4): 370-380, 2024 Apr.
Article in English | MEDLINE | ID: mdl-36786545

ABSTRACT

Millions of individuals globally suffer from inadvertent, occupational or self-harm exposures from organophosphate (OP) insecticides, significantly impacting human health. Similar to nerve agents, insecticides are neurotoxins that target and inhibit acetylcholinesterase (AChE) in central and peripheral synapses in the cholinergic nervous system. Post-exposure therapeutic countermeasures generally include administration of atropine with an oxime to reactivate the OP-inhibited AChE. However, animal model studies and recent clinical trials using insecticide-poisoned individuals have shown minimal clinical benefits of the currently approved oximes and their efficacy as antidotes has been debated. Currently used oximes either reactivate poorly, do not readily cross the blood-brain barrier (BBB), or are rapidly cleared from the circulation and must be repeatedly administered. Zwitterionic oximes of unbranched and simplified structure, for example RS194B, have been developed that efficiently cross the BBB resulting in reactivation of OP-inhibited AChE and dramatic reversal of severe clinical symptoms in mice and macaques exposed to OP insecticides or nerve agents. Thus, a single IM injection of RS194B has been shown to rapidly restore blood AChE and butyrylcholinesterase (BChE) activity, reverse cholinergic symptoms, and prevent death in macaques following lethal inhaled sarin and paraoxon exposure. The present macaque studies extend these findings and assess the ability of post-exposure RS194B treatment to counteract oral poisoning by highly toxic diethylphosphorothioate insecticides such as parathion and chlorpyrifos. These OPs require conversion by P450 in the liver of the inactive thions to the active toxic oxon forms, and once again demonstrated RS194B efficacy to reactivate and alleviate clinical symptoms within 60 mins of a single IM administration. Furthermore, when delivered orally, the Tmax of RS194B at 1-2 h was in the same range as those administered IM but were maintained in the circulation for longer periods greatly facilitating the use of RS194B as a non-invasive treatment, especially in isolated rural settings.


Subject(s)
Acetamides , Chlorpyrifos , Cholinesterase Reactivators , Insecticides , Nerve Agents , Parathion , Animals , Mice , Acetylcholinesterase/chemistry , Butyrylcholinesterase/chemistry , Chlorpyrifos/toxicity , Cholinesterase Inhibitors/chemistry , Cholinesterase Reactivators/chemistry , Cholinesterase Reactivators/pharmacology , Insecticides/toxicity , Macaca , Organophosphorus Compounds/toxicity , Oximes/pharmacology , Oximes/chemistry , Oximes/therapeutic use , Parathion/adverse effects , Parathion/toxicity
15.
J Neurochem ; 168(3): 288-302, 2024 03.
Article in English | MEDLINE | ID: mdl-38275215

ABSTRACT

An increase in tau acetylation at K274 and K281 and abnormal mitochondrial dynamics have been observed in the brains of Alzheimer's disease (AD) patients. Here, we constructed three types of tau plasmids, TauKQ (acetylated tau mutant, by mutating its K274/K281 into glutamine to mimic disease-associated lysine acetylation), TauKR (non-acetylated tau mutant, by mutating its K274/K281 into arginine), and TauWT (wild-type human full-length tau). By transfecting these tau plasmids in HEK293 cells, we found that TauWT and TauKR induced mitochondrial fusion by increasing the level of mitochondrial fusion proteins. Conversely, TauKQ induced mitochondrial fission by reducing mitochondrial fusion proteins, exacerbating mitochondrial dysfunction and apoptosis. BGP-15 ameliorated TauKQ-induced mitochondrial dysfunction and apoptosis by improving mitochondrial dynamics. Our findings suggest that acetylation of K274/281 represents an important post-translational modification site regulating mitochondrial dynamics, and that BGP-15 holds potential as a therapeutic agent for mitochondria-associated diseases such as AD.


Subject(s)
Alzheimer Disease , Mitochondrial Diseases , Oximes , Piperidines , Humans , Acetylation , Alzheimer Disease/metabolism , Apoptosis , HEK293 Cells , Mitochondrial Dynamics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , tau Proteins/genetics , tau Proteins/metabolism
16.
Cancer ; 130(10): 1784-1796, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38261444

ABSTRACT

BACKGROUND: Aberrant PI3K/AKT signaling in BRAF-mutant cancers contributes to resistance to BRAF inhibitors. The authors examined dual MAPK and PI3K pathway inhibition in patients who had BRAF-mutated solid tumors (ClinicalTrials.gov identifier NCT01902173). METHODS: Patients with BRAF V600E/V600K-mutant solid tumors received oral dabrafenib at 150 mg twice daily with dose escalation of oral uprosertib starting at 50 mg daily, or, in the triplet cohorts, with dose escalation of both oral trametinib starting at 1.5 mg daily and oral uprosertib starting at 25 mg daily. Dose-limiting toxicities (DLTs) were assessed within the first 56 days of treatment. Radiographic responses were assessed at 8-week intervals. RESULTS: Twenty-seven patients (22 evaluable) were enrolled in parallel doublet and triplet cohorts. No DLTs were observed in the doublet cohorts (N = 7). One patient had a DLT at the maximum administered dose of triplet therapy (dabrafenib 150 mg twice daily and trametinib 2 mg daily plus uprosertib 75 mg daily). Three patients in the doublet cohorts had partial responses (including one who had BRAF inhibitor-resistant melanoma). Two patients in the triplet cohorts had a partial response, and one patient had an unconfirmed partial response. Pharmacokinetic data suggested reduced dabrafenib and dabrafenib metabolite exposure in patients who were also exposed to both trametinib and uprosertib, but not in whose who were exposed to uprosertib without trametinib. CONCLUSIONS: Concomitant inhibition of both the MAPK and PI3K-AKT pathways for the treatment of BRAF-mutated cancers was well tolerated, leading to objective responses, but higher level drug-drug interactions affected exposure to dabrafenib and its metabolites.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Imidazoles , Mutation , Neoplasms , Oximes , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins c-akt , Pyridones , Pyrimidinones , Humans , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Female , Male , Middle Aged , Aged , Adult , Pyridones/administration & dosage , Pyridones/adverse effects , Pyrimidinones/administration & dosage , Pyrimidinones/adverse effects , Pyrimidinones/therapeutic use , Imidazoles/administration & dosage , Imidazoles/therapeutic use , Imidazoles/adverse effects , Imidazoles/pharmacokinetics , Proto-Oncogene Proteins c-akt/metabolism , Oximes/administration & dosage , Oximes/adverse effects , Oximes/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Aged, 80 and over , Molecular Targeted Therapy
17.
Angiogenesis ; 27(3): 441-460, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38700584

ABSTRACT

Current treatments of brain arteriovenous malformation (BAVM) are associated with considerable risks and at times incomplete efficacy. Therefore, a clinically consistent animal model of BAVM is urgently needed to investigate its underlying biological mechanisms and develop innovative treatment strategies. Notably, existing mouse models have limited utility due to heterogenous and untypical phenotypes of AVM lesions. Here we developed a novel mouse model of sporadic BAVM that is consistent with clinical manifestations in humans. Mice with BrafV600E mutations in brain ECs developed BAVM closely resembled that of human lesions. This strategy successfully induced BAVMs in mice across different age groups and within various brain regions. Pathological features of BAVM were primarily dilated blood vessels with reduced vascular wall stability, accompanied by spontaneous hemorrhage and neuroinflammation. Single-cell sequencing revealed differentially expressed genes that were related to the cytoskeleton, cell motility, and intercellular junctions. Early administration of Dabrafenib was found to be effective in slowing the progression of BAVMs; however, its efficacy in treating established BAVM lesions remained uncertain. Taken together, our proposed approach successfully induced BAVM that closely resembled human BAVM lesions in mice, rendering the model suitable for investigating the pathogenesis of BAVM and assessing potential therapeutic strategies.


Subject(s)
Intracranial Arteriovenous Malformations , Proto-Oncogene Proteins B-raf , Animals , Intracranial Arteriovenous Malformations/genetics , Intracranial Arteriovenous Malformations/pathology , Intracranial Arteriovenous Malformations/metabolism , Mice , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Mutation/genetics , Disease Models, Animal , Humans , Oximes/pharmacology , Imidazoles/pharmacology , Brain/pathology , Brain/metabolism , Brain/blood supply , Endothelium, Vascular/pathology , Endothelium, Vascular/metabolism , Mice, Transgenic , Mice, Inbred C57BL
18.
Br J Cancer ; 131(5): 843-851, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38971952

ABSTRACT

BACKGROUND: Therapeutic drug monitoring (TDM) - performing dose adjustments based on measured drug levels and established pharmacokinetic (PK) targets - could optimise treatment with drugs that show large interpatient variability in exposure. We evaluated the feasibility of TDM for multiple oral targeted therapies. Here we report on drugs for which routine TDM is not feasible. METHODS: We evaluated drug cohorts from the Dutch Pharmacology Oncology Group - TDM study. Based on PK levels taken at pre-specified time points, PK-guided interventions were performed. Feasibility of TDM was evaluated, and based on the success and practicability of TDM, cohorts could be closed. RESULTS: For 10 out of 24 cohorts TDM was not feasible and inclusion was closed. A high incidence of adverse events resulted in closing the cabozantinib, dabrafenib/trametinib, everolimus, regorafenib and vismodegib cohort. The enzalutamide and erlotinib cohorts were closed because almost all PK levels were above target. Other, non-pharmacological reasons led to closing the palbociclib, olaparib and tamoxifen cohort. CONCLUSIONS: Although TDM could help personalising treatment for many drugs, the above-mentioned reasons can influence its feasibility, usefulness and clinical applicability. Therefore, routine TDM is not advised for cabozantinib, dabrafenib/trametinib, enzalutamide, erlotinib, everolimus, regorafenib and vismodegib. Nonetheless, TDM remains valuable for individual clinical decisions.


Subject(s)
Drug Monitoring , Neoplasms , Pyridines , Humans , Drug Monitoring/methods , Prospective Studies , Neoplasms/drug therapy , Pyridines/pharmacokinetics , Pyridines/administration & dosage , Feasibility Studies , Administration, Oral , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Molecular Targeted Therapy , Imidazoles/pharmacokinetics , Imidazoles/administration & dosage , Anilides/pharmacokinetics , Anilides/administration & dosage , Male , Everolimus/pharmacokinetics , Everolimus/administration & dosage , Female , Oximes/pharmacokinetics , Oximes/administration & dosage , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacokinetics , Phenylthiohydantoin/administration & dosage , Nitriles/pharmacokinetics , Nitriles/administration & dosage , Phenylurea Compounds , Pyridones , Pyrimidinones , Piperazines , Benzamides
19.
Ann Oncol ; 35(8): 739-746, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38754780

ABSTRACT

BACKGROUND: Neoadjuvant dabrafenib plus trametinib has a high pathological response rate and impressive short-term survival in patients with resectable stage III melanoma. We report 5-year outcomes from the phase II NeoCombi trial. PATIENTS AND METHODS: NeoCombi (NCT01972347) was a single-arm, open-label, single-centre, phase II trial. Eligible patients were adults (aged ≥18 years) with histologically confirmed, resectable, RECIST-measurable, American Joint Committee on Cancer seventh edition clinical stage IIIB-C BRAF V600E/K-mutant melanoma and Eastern Cooperative Oncology Group performance status ≤1. Patients received 52 weeks of treatment with dabrafenib 150 mg (orally twice per day) plus trametinib 2 mg (orally once per day), with complete resection of the pre-therapy tumour bed at week 12. RESULTS: Between 20 August 2014 and 19 April 2017, 35 patients were enrolled. At data cut-off (17 August 2021), the median follow-up was 60 months [95% confidence interval (CI) 56-72 months]. Overall, 21 of 35 (60%) patients recurred, including 12 (57%) with first recurrence in locoregional sites (followed by later distant recurrence in 6) and 9 (43%) with first recurrence in distant sites, including 3 in the brain. Most recurrences occurred within 2 years, with no recurrences beyond 3 years. At 5 years, recurrence-free survival (RFS) was 40% (95% CI 27% to 60%), distant metastasis-free survival (DMFS) was 57% (95% CI 42% to 76%), and overall survival was 80% (95% CI 67% to 94%). Five-year survival outcomes were stratified by pathological response: RFS was 53% with pathological complete response (pCR) versus 28% with non-pCR (P = 0.087), DMFS was 59% versus 55% (P = 0.647), and overall survival was 88% versus 71% (P = 0.205), respectively. CONCLUSIONS: Neoadjuvant dabrafenib plus trametinib has high pathological response rates in clinical stage III melanoma, but low rates of RFS, similar to those achieved with adjuvant targeted therapy alone. Patients with a pCR to dabrafenib plus trametinib still had a high risk of recurrence, unlike that seen with immunotherapy where recurrences are rare.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Imidazoles , Melanoma , Neoadjuvant Therapy , Neoplasm Staging , Oximes , Pyridones , Pyrimidinones , Humans , Oximes/administration & dosage , Melanoma/drug therapy , Melanoma/pathology , Melanoma/mortality , Pyrimidinones/administration & dosage , Pyridones/administration & dosage , Imidazoles/administration & dosage , Female , Male , Middle Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aged , Adult , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin Neoplasms/mortality , Follow-Up Studies
20.
J Pharmacol Exp Ther ; 388(2): 536-545, 2024 01 17.
Article in English | MEDLINE | ID: mdl-37652710

ABSTRACT

Phosgene oxime (CX), categorized as a vesicating chemical threat agent, causes effects that resemble an urticant or nettle agent. CX is an emerging potential threat agent that can be deployed alone or with other chemical threat agents to enhance their toxic effects. Studies on CX-induced skin toxicity, injury progression, and related biomarkers are largely unknown. To study the physiologic changes, skin clinical lesions and their progression, skin exposure of SKH-1 and C57BL/6 mice was carried out with vapor from 10 µl CX for 0.5-minute or 1.0-minute durations using a designed exposure system for consistent CX vapor exposure. One-minute exposure caused sharp (SKH-1) or sustained (C57BL/6) decrease in respiratory and heart rate, leading to mortality in both mouse strains. Both exposures caused immediate blanching, erythema with erythematous ring (wheel) and edema, and an increase in skin bifold thickness. Necrosis was also observed in the 0.5-minute CX exposure group. Both mouse strains showed comparative skin clinical lesions upon CX exposure; however, skin bifold thickness and erythema remained elevated up to 14 days postexposure in SKH-1 mice but not in C57BL/6 mice. Our data suggest that CX causes immediate changes in the physiologic parameters and gross skin lesions resembling urticaria, which could involve mast cell activation and intense systemic toxicity. This novel study recorded and compared the progression of skin injury to establish clinical biomarkers of CX dermal exposure in both the sexes of two murine strains relevant for skin and systemic injury studies and therapeutic target identification. SIGNIFICANCE STATEMENT: Phosgene oxime (CX), categorized as a vesicating agent, is considered as a potent chemical weapon and is of high military and terrorist threat interest since it produces rapid onset of severe injury as an urticant. However, biomarkers of clinical relevance related to its toxicity and injury progression are not studied. Data from this study provide useful clinical markers of CX skin toxicity in mouse models using a reliable CX exposure system for future mechanistic and efficacy studies.


Subject(s)
Chemical Warfare Agents , Mustard Gas , Phosgene , Animals , Mice , Phosgene/toxicity , Disease Models, Animal , Mustard Gas/toxicity , Mice, Inbred C57BL , Skin , Irritants/toxicity , Erythema/chemically induced , Erythema/pathology , Biomarkers , Oximes/toxicity , Chemical Warfare Agents/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL