Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Eur J Med Chem ; 264: 115971, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38071795

ABSTRACT

Pharmacological inhibition of dihydrofolate reductase (DHFR) is an established approach for treating a variety of human diseases, including foreign infections and cancer. However, treatment with classic DHFR inhibitors, such as methotrexate (MTX), are associated with negative side-effects and resistance mechanisms that have prompted the search for alternatives. The DHFR inhibitor pyrimethamine (Pyr) has compelling anti-cancer activity in in vivo models, but lacks potency compared to MTX, thereby requiring higher concentrations to induce therapeutic responses. The purpose of this work was to investigate structural analogues of Pyr to improve its in vitro and cellular activity. A series of 36 Pyr analogues were synthesized and tested in a sequence of in vitro and cell-based assays to monitor their DHFR inhibitory activity, cellular target engagement, and impact on breast cancer cell viability. Ten top compounds were identified, two of which stood out as potential lead candidates, 32 and 34. These functionalized Pyr analogues potently engaged DHFR in cells, at concentrations as low as 1 nM and represent promising DHFR inhibitors that could be further explored as potential anti-cancer agents.


Subject(s)
Antineoplastic Agents , Folic Acid Antagonists , Neoplasms , Humans , Pyrimethamine/pharmacology , Folic Acid Antagonists/pharmacology , Folic Acid Antagonists/chemistry , Methotrexate/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Biology , Tetrahydrofolate Dehydrogenase/chemistry
2.
Metabolites ; 13(2)2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36837770

ABSTRACT

Dihydrofolate reductase (DHFR) is an established anti-cancer drug target whose inhibition disrupts folate metabolism and STAT3-dependent gene expression. Cycloguanil was proposed as a DHFR inhibitor in the 1950s and is the active metabolite of clinically approved plasmodium DHFR inhibitor Proguanil. The Cycloguanil scaffold was explored to generate potential cancer therapies in the 1970s. Herein, current computational and chemical biology techniques were employed to re-investigate the anti-cancer activity of Cycloguanil and related compounds. In silico modeling was employed to identify promising Cycloguanil analogues from NCI databases, which were cross-referenced with NCI-60 Human Tumor Cell Line Screening data. Using target engagement assays, it was found that these compounds engage DHFR in cells at sub-nanomolar concentrations; however, growth impairments were not observed until higher concentrations. Folinic acid treatment rescues the viability impairments induced by some, but not all, Cycloguanil analogues, suggesting these compounds may have additional targets. Cycloguanil and its most promising analogue, NSC127159, induced similar metabolite profiles compared to established DHFR inhibitors Methotrexate and Pyrimethamine while also blocking downstream signaling, including STAT3 transcriptional activity. These data confirm that Cycloguanil and its analogues are potent inhibitors of human DHFR, and their anti-cancer activity may be worth further investigation.

3.
Amino Acids ; 55(2): 215-233, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36454288

ABSTRACT

Protein arginine N-methyltransferases (PRMTs) have emerged as important actors in the eukaryotic stress response with implications in human disease, aging, and cell signaling. Intracellular free methylarginines contribute to cellular stress through their interaction with nitric oxide synthase (NOS). The arginine-dependent production of nitric oxide (NO), which is strongly inhibited by methylarginines, serves as a protective small molecule against oxidative stress in eukaryotic cells. NO signaling is highly conserved between higher and lower eukaryotes, although a canonical NOS homologue has yet to be identified in yeast. Since stress signaling pathways are well conserved among eukaryotes, yeast is an ideal model organism to study the implications of PRMTs and methylarginines during stress. We sought to explore the roles and fates of methylarginines in Saccharomyces cerevisiae. We starved methyltransferase-, autophagy-, and permease-related yeast knockouts by incubating them in water and monitored methylarginine production. We found that under starvation, methylarginines are expelled from yeast cells. We found that autophagy-deficient cells have an impaired ability to efflux methylarginines, which suggests that methylarginine-containing proteins are degraded via autophagy. For the first time, we determine that yeast take up methylarginines less readily than arginine, and we show that methylarginines impact yeast NO production. This study reveals that yeast circumvent a potential methylarginine toxicity by expelling them after autophagic degradation of arginine-modified proteins.


Subject(s)
Nitric Oxide , Saccharomyces cerevisiae , Humans , omega-N-Methylarginine/metabolism , omega-N-Methylarginine/pharmacology , Saccharomyces cerevisiae/metabolism , Nitric Oxide/metabolism , Arginine/metabolism , Nitric Oxide Synthase/metabolism , Nutrients
4.
Biomaterials ; 289: 121735, 2022 10.
Article in English | MEDLINE | ID: mdl-36055815

ABSTRACT

A small molecule drug with poor aqueous solubility can be conjugated to a hydrophilic polymer like poly(ethylene glycol) (PEG) to form an amphiphilic polymer-drug conjugate that self-assembles to form nanoparticles (NPs) with improved solubility and enhanced efficacy. This strategy has been extensively applied to improve the delivery of several small molecule drugs. However, very few reports have succeeded to tune the rate of drug release from these NPs. To the best of our knowledge, there have been no reports of utilizing click and steric hindrance chemistry to modulate the drug release of self-assembling polymer-drug conjugates. In this study, we utilized click chemistry to conjugate methoxy-PEG (mPEG) to an anti-tumor drug, paclitaxel (PTX). A focused library of PTX-Rx-mPEG (x = 0, 1, 2) conjugates were synthesized with different chemical modalities next to the cleavable ester bond to study the effect of increasing steric hindrance on the self-assembly process and the physicochemical properties of the resulting PTX-NPs. PTX-R0-mPEG had no added steric hindrance (x = 0; minimal), PTX-R1-mPEG consisted of two methyl groups (x = 1: moderate), and PTX-R2-mPEG consisted of a phenyl group (x = 2: significant). Drug release studies showed that PTX-NPs released PTX at a decreased rate with increasing steric hindrance. Pharmacokinetic studies showed that the AUC of released PTX from the moderate-release PTX-R1-NP was approximately 20-, 6-, and 3-fold higher than that from free PTX, PTX-R0-NP and PTX-R2-NP, respectively. As a result, among these different PTX formulations, PTX-R1-NP showed superior efficacy in inducing tumor regression and prolonging the animal survival. The tumors treated with PTX-R1-NP displayed the lowest tumor progression markers (Ki68 and CD31) and the highest apoptotic marker (TUNEL) compared to the others. This work emphasizes the importance of taking a systematic approach in designing self-assembling polymer drug conjugates and highlights the potential of utilizing steric hindrance as a tool to tune the drug release rate from such systems.


Subject(s)
Antineoplastic Agents , Nanoparticles , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival , Drug Carriers/chemistry , Drug Delivery Systems , Drug Liberation , Esters , Nanoparticles/chemistry , Paclitaxel/therapeutic use , Polyethylene Glycols/chemistry , Polymers/chemistry
5.
J Biol Chem ; 298(2): 101531, 2022 02.
Article in English | MEDLINE | ID: mdl-34953855

ABSTRACT

Cancer is often characterized by aberrant gene expression patterns caused by the inappropriate activation of transcription factors. Signal transducer and activator of transcription 3 (STAT3) is a key transcriptional regulator of many protumorigenic processes and is persistently activated in many types of human cancer. However, like many transcription factors, STAT3 has proven difficult to target clinically. To address this unmet clinical need, we previously developed a cell-based assay of STAT3 transcriptional activity and performed an unbiased and high-throughput screen of small molecules known to be biologically active in humans. We identified the antimicrobial drug pyrimethamine as a novel and specific inhibitor of STAT3 transcriptional activity. Here, we show that pyrimethamine does not significantly affect STAT3 phosphorylation, nuclear translocation, or DNA binding at concentrations sufficient to inhibit STAT3 transcriptional activity, suggesting a potentially novel mechanism of inhibition. To identify the direct molecular target of pyrimethamine and further elucidate the mechanism of action, we used a new quantitative proteome profiling approach called proteome integral solubility alteration coupled with a metabolomic analysis. We identified human dihydrofolate reductase as a target of pyrimethamine and demonstrated that the STAT3-inhibitory effects of pyrimethamine are the result of a deficiency in reduced folate downstream of dihydrofolate reductase inhibition, implicating folate metabolism in the regulation of STAT3 transcriptional activity. This study reveals a previously unknown regulatory node of the STAT3 pathway that may be important for the development of novel strategies to treat STAT3-driven cancers.


Subject(s)
Anti-Infective Agents , Pyrimethamine , STAT3 Transcription Factor , Tetrahydrofolate Dehydrogenase , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Cell Line, Tumor , Folic Acid/metabolism , Humans , Proteome/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism
6.
J Neurochem ; 159(4): 742-761, 2021 11.
Article in English | MEDLINE | ID: mdl-34216036

ABSTRACT

Protein arginine methyltransferases (PRMTs) are a family of enzymes involved in gene regulation and protein/histone modifications. PRMT8 is primarily expressed in the central nervous system, specifically within the cellular membrane and synaptic vesicles. Recently, PRMT8 has been described to play key roles in neuronal signaling such as a regulator of dendritic arborization, synaptic function and maturation, and neuronal differentiation and plasticity. Here, we examined the role of PRMT8 in response to hypoxia-induced stress in brain metabolism. Our results from liquid chromatography mass spectrometry, mitochondrial oxygen consumption rate, and protein analyses indicate that PRMT8(-/-) knockout mice presented with altered membrane phospholipid composition, decreased mitochondrial stress capacity, and increased neuroinflammatory markers, such as tumor necrosis factor alpha and ionized calcium binding adaptor molecule 1 (Iba1, a specific marker for microglia/macrophage activation) after hypoxic stress. Furthermore, adenovirus-based overexpression of PRMT8 reversed the changes in membrane phospholipid composition, mitochondrial stress capacity, and neuroinflammatory markers. Together, our findings establish PRMT8 as an important regulatory component of membrane phospholipid composition, short-term memory function, mitochondrial function, and neuroinflammation in response to hypoxic stress.


Subject(s)
Energy Metabolism/genetics , Hypoxia/genetics , Mitochondria/genetics , Mitochondria/metabolism , Neuroinflammatory Diseases/genetics , Protein-Arginine N-Methyltransferases/genetics , Animals , Calcium-Binding Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Cytokines/analysis , Cytokines/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Neural Stem Cells , Oxygen Consumption , Phospholipids/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
Methods ; 175: 10-23, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31726226

ABSTRACT

Protein arginine N-methyltransferases (PRMTs) are a family of 9 enzymes that catalyze mono- or di-methylation of arginine residues using S-adenosyl-l-methionine (SAM). Arginine methylation is an important post-translational modification that can regulate the activity and structure of target proteins. Altered PRMT activity can lead to a variety of health issues including neurodevelopmental disease, autoimmune disorders, cancer, and cardiovascular disease. Thus, developing a robust mechanistic understanding of PRMT function may provide insight into these various disease states and enable the development of potential therapeutic agents. Although PRMTs have been studied for nearly two decades, a consensus regarding the mechanism of action for this class of enzymes has remained noticeably elusive. To address this shortcoming, differential scanning fluorimetry (DSF) was used to gain mechanistic insight into the order of PRMT substrate and cofactor binding. This methodology confirms that PRMT cofactor binding precedes target substrate binding and supports the use of DSF to study bisubstrate enzymatic reaction mechanisms.


Subject(s)
Arginine/chemistry , Calorimetry, Differential Scanning/methods , Coenzymes/chemistry , Enzyme Assays/methods , Fluorometry/methods , Protein-Arginine N-Methyltransferases/chemistry , S-Adenosylmethionine/chemistry , Arginine/metabolism , Catalysis , Dimerization , Fluorescent Dyes/chemistry , Gene Expression , Kinetics , Methylation , Protein Conformation, alpha-Helical , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/isolation & purification , Protein-Arginine N-Methyltransferases/metabolism , Recombinant Proteins , S-Adenosylmethionine/metabolism , Substrate Specificity , Temperature
8.
Biochim Biophys Acta Proteins Proteom ; 1867(3): 306-316, 2019 03.
Article in English | MEDLINE | ID: mdl-30342239

ABSTRACT

Protein arginine N-methyltransferase (PRMT) kinetic parameters have been catalogued over the past fifteen years for eight of the nine mammalian enzyme family members. Like the majority of methyltransferases, these enzymes employ the highly ubiquitous cofactor S-adenosyl-l-methionine as a co-substrate to methylate arginine residues in peptidic substrates with an approximately 4-µM median KM. The median values for PRMT turnover number (kcat) and catalytic efficiency (kcat/KM) are 0.0051 s-1 and 708 M-1 s-1, respectively. When comparing PRMT metrics to entries found in the BRENDA database, we find that while PRMTs exhibit high substrate affinity relative to other enzyme-substrate pairs, PRMTs display largely lower kcat and kcat/KM values. We observe that kinetic parameters for PRMTs and arginine demethylase activity from dual-functioning lysine demethylases are statistically similar, paralleling what the broader enzyme families in which they belong reveal, and adding to the evidence in support of arginine methylation reversibility.


Subject(s)
Arginine/chemistry , Protein-Arginine N-Methyltransferases/chemistry , Kinetics , Methylation
9.
Chembiochem ; 19(1): 85-99, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29112789

ABSTRACT

Arginine methylation is a prevalent post-translational modification in eukaryotic cells. Two significant debates exist within the field: do these enzymes dimethylate their substrates in a processive or distributive manner, and do these enzymes operate using a random or sequential method of bisubstrate binding? We revealed that human protein arginine N-methyltransferase 1 (PRMT1) enzyme kinetics are dependent on substrate sequence. Further, peptides containing an Nη-hydroxyarginine generally demonstrated substrate inhibition and had improved KM values, which evoked a possible role in inhibitor design. We also revealed that the perceived degree of enzyme processivity is a function of both cofactor and enzyme concentration, suggesting that previous conclusions about PRMT sequential methyl transfer mechanisms require reassessment. Finally, we demonstrated a sequential ordered Bi-Bi kinetic mechanism for PRMT1, based on steady-state kinetic analysis. Together, our data indicate a PRMT1 mechanism of action and processivity that might also extend to other functionally and structurally conserved PRMTs.


Subject(s)
Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Arginine/metabolism , Biocatalysis , Humans , Kinetics , Mass Spectrometry , Methylation , Peptides/analysis , Peptides/chemistry , Protein-Arginine N-Methyltransferases/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Repressor Proteins/genetics , Substrate Specificity
10.
Childs Nerv Syst ; 20(2): 100-3, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14556029

ABSTRACT

BACKGROUND: Arachnoid cysts are a relatively common incidental finding on CT scans of the brain. They most commonly occur in the middle cranial fossa, where familial occurrence has rarely been reported. Posterior fossa arachnoid cysts are more unusual. CASE HISTORIES: We report the presence of quadrigeminal cistern arachnoid cysts in siblings.


Subject(s)
Arachnoid Cysts/pathology , Family Health , Skull Base Neoplasms/pathology , Arachnoid Cysts/complications , Arachnoid Cysts/surgery , Child, Preschool , Cranial Fossa, Posterior , Female , Humans , Hydrocephalus/etiology , Hydrocephalus/pathology , Hydrocephalus/surgery , Infant , Magnetic Resonance Imaging/methods , Male , Neurologic Examination/methods , Skull Base Neoplasms/complications , Tomography, X-Ray Computed/methods , Ventriculoperitoneal Shunt/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...