Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 543
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 121(22): e2401591121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38787877

ABSTRACT

The sodium (Na+) leak channel (NALCN) is a member of the four-domain voltage-gated cation channel family that includes the prototypical voltage-gated sodium and calcium channels (NaVs and CaVs, respectively). Unlike NaVs and CaVs, which have four lateral fenestrations that serve as routes for lipophilic compounds to enter the central cavity to modulate channel function, NALCN has bulky residues (W311, L588, M1145, and Y1436) that block these openings. Structural data suggest that occluded fenestrations underlie the pharmacological resistance of NALCN, but functional evidence is lacking. To test this hypothesis, we unplugged the fenestrations of NALCN by substituting the four aforementioned residues with alanine (AAAA) and compared the effects of NaV, CaV, and NALCN blockers on both wild-type (WT) and AAAA channels. Most compounds behaved in a similar manner on both channels, but phenytoin and 2-aminoethoxydiphenyl borate (2-APB) elicited additional, distinct responses on AAAA channels. Further experiments using single alanine mutants revealed that phenytoin and 2-APB enter the inner cavity through distinct fenestrations, implying structural specificity to their modes of access. Using a combination of computational and functional approaches, we identified amino acid residues critical for 2-APB activity, supporting the existence of drug binding site(s) within the pore region. Intrigued by the activity of 2-APB and its analogues, we tested compounds containing the diphenylmethane/amine moiety on WT channels. We identified clinically used drugs that exhibited diverse activity, thus expanding the pharmacological toolbox for NALCN. While the low potencies of active compounds reiterate the pharmacological resistance of NALCN, our findings lay the foundation for rational drug design to develop NALCN modulators with refined properties.


Subject(s)
Phenytoin , Binding Sites , Humans , Phenytoin/metabolism , Phenytoin/pharmacology , Boron Compounds/chemistry , Boron Compounds/pharmacology , Boron Compounds/metabolism , Ion Channels/metabolism , Ion Channels/genetics , HEK293 Cells , Animals , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/chemistry , Membrane Proteins
2.
Eur J Nucl Med Mol Imaging ; 51(6): 1703-1712, 2024 May.
Article in English | MEDLINE | ID: mdl-38191817

ABSTRACT

PURPOSE: Boramino acids are a class of amino acid biomimics that replace the carboxylate group with trifluoroborate and can achieve the 18F-labeled positron emission tomography (PET) and boron neutron capture therapy (BNCT) with identical chemical structure. METHODS: This study reports a trifluoroborate-derived boronophenylalanine (BBPA), a derived boronophenylalanine (BPA) for BNCT, as a promising PET tracer for tumor imaging. RESULTS: Competition inhibition assays in cancer cells suggested the cell accumulation of [18F]BBPA is through large neutral amino acid transporter type-1 (LAT-1). Of note, [18F]BBPA is a pan-cancer probe that shows notable tumor uptake in B16-F10 tumor-bearing mice. In the patients with gliomas and metastatic brain tumors, [18F]BBPA-PET shows good tumor uptake and notable tumor-to-normal brain ratio (T/N ratio, 18.7 ± 5.5, n = 11), higher than common amino acid PET tracers. The [18F]BBPA-PET quantitative parameters exhibited no difference in diverse contrast-enhanced status (P = 0.115-0.687) suggesting the [18F]BBPA uptake was independent from MRI contrast-enhancement. CONCLUSION: This study outlines a clinical trial with [18F]BBPA to achieve higher tumor-specific accumulation for PET, provides a potential technique for brain tumor diagnosis, and might facilitate the BNCT of brain tumors.


Subject(s)
Boron Compounds , Brain Neoplasms , Fluorine Radioisotopes , Phenylalanine , Positron Emission Tomography Computed Tomography , Radioactive Tracers , Animals , Female , Humans , Mice , Boron Compounds/analysis , Boron Compounds/metabolism , Boron Compounds/pharmacokinetics , Boron Neutron Capture Therapy , Brain/diagnostic imaging , Brain/metabolism , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Fluorine Radioisotopes/analysis , Fluorine Radioisotopes/metabolism , Fluorine Radioisotopes/pharmacokinetics , Healthy Volunteers , Large Neutral Amino Acid-Transporter 1/metabolism , Magnetic Resonance Imaging , Melanoma, Experimental , Mice, Inbred C57BL , Molecular Probes/analysis , Molecular Probes/metabolism , Molecular Probes/pharmacokinetics , Phenylalanine/analogs & derivatives , Phenylalanine/analysis , Phenylalanine/metabolism , Phenylalanine/pharmacokinetics , Positron Emission Tomography Computed Tomography/methods , Xenograft Model Antitumor Assays
3.
Mol Cell ; 62(2): 284-294, 2016 04 21.
Article in English | MEDLINE | ID: mdl-27105118

ABSTRACT

Natural variations in gene expression provide a mechanism for multiple phenotypes to arise in an isogenic bacterial population. In particular, a sub-group termed persisters show high tolerance to antibiotics. Previously, their formation has been attributed to cell dormancy. Here we demonstrate that bacterial persisters, under ß-lactam antibiotic treatment, show less cytoplasmic drug accumulation as a result of enhanced efflux activity. Consistently, a number of multi-drug efflux genes, particularly the central component TolC, show higher expression in persisters. Time-lapse imaging and mutagenesis studies further establish a positive correlation between tolC expression and bacterial persistence. The key role of efflux systems, among multiple biological pathways involved in persister formation, indicates that persisters implement a positive defense against antibiotics prior to a passive defense via dormancy. Finally, efflux inhibitors and antibiotics together effectively attenuate persister formation, suggesting a combination strategy to target drug tolerance.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/metabolism , Boron Compounds/pharmacology , Drug Resistance, Bacterial , Escherichia coli Proteins/metabolism , Escherichia coli/drug effects , Membrane Transport Proteins/metabolism , Penicillins/pharmacology , Anti-Bacterial Agents/metabolism , Bacterial Outer Membrane Proteins/genetics , Biological Transport , Boron Compounds/metabolism , Colony Count, Microbial , Dose-Response Relationship, Drug , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Genotype , High-Throughput Nucleotide Sequencing , Membrane Transport Proteins/genetics , Microbial Viability/drug effects , Mutation , Optical Imaging , Penicillins/metabolism , Phenotype , Time Factors , Up-Regulation
4.
Angew Chem Int Ed Engl ; 63(29): e202404286, 2024 07 15.
Article in English | MEDLINE | ID: mdl-38712936

ABSTRACT

Perhalogenated closo-borates represent a new class of membrane carriers. They owe this activity to their chaotropicity, which enables the transport of hydrophilic molecules across model membranes and into living cells. The transport efficiency of this new class of cluster carriers depends on a careful balance between their affinity to membranes and cargo, which varies with chaotropicity. However, the structure-activity parameters that define chaotropic transport remain to be elucidated. Here, we have studied the modulation of chaotropic transport by decoupling the halogen composition from the boron core size. The binding affinity between perhalogenated decaborate and dodecaborate clusters carriers was quantified with different hydrophilic model cargos, namely a neutral and a cationic peptide, phalloidin and (KLAKLAK)2. The transport efficiency, membrane-lytic properties, and cellular toxicity, as obtained from different vesicle and cell assays, increased with the size and polarizability of the clusters. These results validate the chaotropic effect as the driving force behind the membrane transport propensity of boron clusters. This work advances our understanding of the structural features of boron cluster carriers and establishes the first set of rational design principles for chaotropic membrane transporters.


Subject(s)
Boron , Boron/chemistry , Boron/metabolism , Humans , Biological Transport , Boron Compounds/chemistry , Boron Compounds/metabolism , Hydrophobic and Hydrophilic Interactions , Borates/chemistry , Borates/metabolism
5.
Turk J Med Sci ; 53(3): 619-629, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37476906

ABSTRACT

BACKGROUND: Gastrointestinal health is essential for maintaining a healthy lifestyle. Improving nutrient absorption and energy metabolism are the critical targets for intestinal health. This study aimed to determine the effects of different boron (B) derivatives on nutrient digestibility, intestinal nutrient transporters, and lipid metabolism in rats. METHODS: Twenty-one rats were allocated to three groups (n = 7) as follows: (i) Control, (ii) Sodium pentaborate pentahydrate (SPP), and (iii) boric acid (BA). The rats were fed a chow diet (AIN-93M) and supplemented with 8 mg/kg elemental B from SPP (45.2 mg/kg BW) and BA (42.7 mg/kg BW) via oral gavage every other day for 12 weeks. The nutrient digestibility of rats in each group was measured using the indigestible indicator (chromium oxide, Cr2 O3, 0.20%). At the end of the experiment, animals were decapitated by cervical dislocation and jejunum, and liver samples were taken from each animal. The nutrient transporters and lipid-regulated transcription factors were determined by RT-PCR. RESULTS: The nutrient digestibility (except for ash) was increased by SPP and BA supplementation (p < 0.05). SPP and BA-supplemented rats had higher jejunal glucose transporter 1 (GLUT1), GLUT2, GLUT5, sodium-dependent glucose transporter 1 (SGLT1), fatty acid transport protein-1 (FATP1), and FATP4 mRNA expression levels compared to nonsupplemented rats (p < 0.0001). BA-supplemented rats had remarkably higher peroxisome proliferator-activated receptor gamma (PPARγ) levels than nonsupplemented rats (p < 0.0001). In contrast, sterol regulatory element-binding protein 1c (SREBP-1c), liver X receptor alpha (LxR-α), and fatty acid synthase (FAS) levels decreased by SPP supplementation compared to other groups (p < 0.05). DISCUSSION: SPP and BA administration enhanced nutrient digestibility, intestinal nutrient transporters, and liver lipid metabolism in rats.


Subject(s)
Intestines , Lipid Metabolism , Rats , Animals , Glucose Transporter Type 1/metabolism , Liver , Boron Compounds/metabolism , Boron Compounds/pharmacology
6.
Chembiochem ; 23(3): e202100366, 2022 02 04.
Article in English | MEDLINE | ID: mdl-34636113

ABSTRACT

Hydrogen peroxide is the most stable reactive oxygen species generated endogenously, participating in numerous physiological processes and abnormal pathological conditions. Mounting evidence suggests that a higher level of H2 O2 exists in various disease conditions. Thus, H2 O2 functions as an ideal target for site-specific bioimaging and therapeutic targeting. The unique reactivity of organoborons with H2 O2 provides a method for developing chemoselective molecules for biological and biomedical applications. This review highlights the design and application of boron-derived molecules for H2 O2 detection, and the utility of boron moieties toward masking reactive compounds leading to the development of metal prochelators and prodrugs for selectively delivering an active species at the target sites with elevated H2 O2 levels. Additionally, the emergence of H2 O2 -responsive theranostic agents consisting of both therapeutic and diagnostic moieties in one integrated system are discussed. The purpose of this review is to provide a better understanding of the role of boron-derived molecules toward biological and pharmacological applications.


Subject(s)
Boron Compounds/chemistry , Hydrogen Peroxide/analysis , Boron Compounds/chemical synthesis , Boron Compounds/metabolism , Molecular Structure
7.
PLoS Pathog ; 16(11): e1008932, 2020 11.
Article in English | MEDLINE | ID: mdl-33141865

ABSTRACT

Livestock diseases caused by Trypanosoma congolense, T. vivax and T. brucei, collectively known as nagana, are responsible for billions of dollars in lost food production annually. There is an urgent need for novel therapeutics. Encouragingly, promising antitrypanosomal benzoxaboroles are under veterinary development. Here, we show that the most efficacious subclass of these compounds are prodrugs activated by trypanosome serine carboxypeptidases (CBPs). Drug-resistance to a development candidate, AN11736, emerged readily in T. brucei, due to partial deletion within the locus containing three tandem copies of the CBP genes. T. congolense parasites, which possess a larger array of related CBPs, also developed resistance to AN11736 through deletion within the locus. A genome-scale screen in T. brucei confirmed CBP loss-of-function as the primary mechanism of resistance and CRISPR-Cas9 editing proved that partial deletion within the locus was sufficient to confer resistance. CBP re-expression in either T. brucei or T. congolense AN11736-resistant lines restored drug-susceptibility. CBPs act by cleaving the benzoxaborole AN11736 to a carboxylic acid derivative, revealing a prodrug activation mechanism. Loss of CBP activity results in massive reduction in net uptake of AN11736, indicating that entry is facilitated by the concentration gradient created by prodrug metabolism.


Subject(s)
Boron Compounds/metabolism , Carboxypeptidases/metabolism , Trypanocidal Agents/metabolism , Trypanosoma brucei brucei/enzymology , Trypanosoma congolense/enzymology , Trypanosoma vivax/enzymology , Trypanosomiasis, African/veterinary , Valine/analogs & derivatives , Animals , Carboxylic Acids/metabolism , Drug Resistance , Female , Livestock , Mice , Parasitemia/veterinary , Prodrugs/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/drug effects , Trypanosoma congolense/drug effects , Trypanosoma vivax/drug effects , Trypanosomiasis, African/drug therapy , Trypanosomiasis, African/parasitology , Valine/metabolism
8.
Cell Mol Life Sci ; 78(3): 1113-1129, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32607595

ABSTRACT

Protein homeostasis is essential for life in eukaryotes. Organisms respond to proteotoxic stress by activating heat shock transcription factors (HSFs), which play important roles in cytoprotection, longevity and development. Of six human HSFs, HSF1 acts as a proteostasis guardian regulating stress-induced transcriptional responses, whereas HSF2 has a critical role in development, in particular of brain and reproductive organs. Unlike HSF1, that is a stable protein constitutively expressed, HSF2 is a labile protein and its expression varies in different tissues; however, the mechanisms regulating HSF2 expression remain poorly understood. Herein we demonstrate that the proteasome inhibitor anticancer drug bortezomib (Velcade), at clinically relevant concentrations, triggers de novo HSF2 mRNA transcription in different types of cancers via HSF1 activation. Similar results were obtained with next-generation proteasome inhibitors ixazomib and carfilzomib, indicating that induction of HSF2 expression is a general response to proteasome dysfunction. HSF2-promoter analysis, electrophoretic mobility shift assays, and chromatin immunoprecipitation studies unexpectedly revealed that HSF1 is recruited to a heat shock element located at 1.397 bp upstream from the transcription start site in the HSF2-promoter. More importantly, we found that HSF1 is critical for HSF2 gene transcription during proteasome dysfunction, representing an interesting example of transcription factor involved in controlling the expression of members of the same family. Moreover, bortezomib-induced HSF2 was found to localize in the nucleus, interact with HSF1, and participate in bortezomib-mediated control of cancer cell migration. The results shed light on HSF2-expression regulation, revealing a novel level of HSF1/HSF2 interplay that may lead to advances in pharmacological modulation of these fundamental transcription factors.


Subject(s)
Heat Shock Transcription Factors/metabolism , Heat-Shock Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Transcription Factors/metabolism , Boron Compounds/chemistry , Boron Compounds/metabolism , Bortezomib/chemistry , Bortezomib/metabolism , Bortezomib/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/metabolism , Electrophoretic Mobility Shift Assay , Glycine/analogs & derivatives , Glycine/chemistry , Glycine/metabolism , Heat Shock Transcription Factors/antagonists & inhibitors , Heat Shock Transcription Factors/genetics , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Humans , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/chemistry , Proteasome Inhibitors/chemistry , Proteasome Inhibitors/metabolism , Proteasome Inhibitors/pharmacology , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Initiation Site , Transcription, Genetic
9.
Reprod Domest Anim ; 57(2): 196-199, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34748661

ABSTRACT

C11-BODIPY581/591 is a fluorescent probe that has been successfully used to evaluate lipid peroxidation in different species, but it has not been completely studied in the dog. Thus, the aim of the present study was to assess lipid peroxidation of dog spermatozoa using C11-BODIPY581/591 and compare different positive controls of the technique. Twenty-four ejaculates were collected from 8 adult male dogs. Routine seminal characteristics were evaluated in raw semen. Lipid peroxidation evaluation was performed as described in other species. Samples were divided in three aliquots, exposed to UV radiation, incubated with hydrogen peroxide or left without treatment (control). Lipid peroxidation was significantly greater only in UV-exposed samples than in the control ones (91 ± 6% vs. 8.3 ± 3.5%, p Ë‚ .01). In conclusion, C11-BODIPY581/591 is useful to evaluate lipid peroxidation of dog spermatozoa and UV radiation is a good promoter of membrane oxidation, so irradiated samples can be used as a positive control of this technique.


Subject(s)
Fluorescent Dyes , Spermatozoa , Animals , Boron Compounds/metabolism , Dogs , Fluorescent Dyes/metabolism , Lipid Peroxidation , Male , Spermatozoa/metabolism
10.
Chembiochem ; 22(18): 2741-2761, 2021 09 14.
Article in English | MEDLINE | ID: mdl-33939874

ABSTRACT

This review describes recent progress in the design and development of inhibitors of human carbonic anhydrase IX (CA IX) based on space-filling carborane and cobalt bis(dicarbollide) clusters. CA IX enzyme is known to play a crucial role in cancer cell proliferation and metastases. The new class of potent and selective CA IX inhibitors combines the structural motif of a bulky inorganic cluster with an alkylsulfamido or alkylsulfonamido anchor group for Zn2+ ion in the enzyme active site. Detailed structure-activity relationship (SAR) studies of a large series containing 50 compounds uncovered structural features of the cluster-containing inhibitors that are important for efficient and selective inhibition of CA IX activity. Preclinical evaluation of selected compounds revealed low toxicity, favorable pharmacokinetics and ability to reduce tumor growth. Cluster-containing inhibitors of CA IX can thus be considered as promising candidates for drug development and/or for combination therapy in boron neutron capture therapy (BNCT).


Subject(s)
Boron Compounds/chemistry , Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase Inhibitors/chemistry , Binding Sites , Boron Compounds/metabolism , Boron Compounds/therapeutic use , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/metabolism , Carbonic Anhydrase Inhibitors/therapeutic use , Humans , Molecular Dynamics Simulation , Neoplasms/drug therapy , Organometallic Compounds/chemistry , Structure-Activity Relationship , Sulfonamides/chemistry
11.
BMC Cancer ; 21(1): 72, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33446132

ABSTRACT

BACKGROUND: p-Boronophenylalanine (10BPA) is a powerful 10B drug used in current clinical trials of BNCT. For BNCT to be successful, a high (500 mg/kg) dose of 10BPA must be administered over a few hours. Here, we report BNCT efficacy after rapid, ultralow-dose administration of either tumor vasculature-specific annexin A1-targeting IFLLWQR (IF7)-conjugated 10BPA or borocaptate sodium (10BSH). METHODS: (1) IF7 conjugates of either 10B drugs intravenously injected into MBT2 bladder tumor-bearing mice and biodistribution of 10B in tumors and normal organs analyzed by prompt gamma-ray analysis. (2) Therapeutic effect of IF7-10B drug-mediated BNCT was assessed by either MBT2 bladder tumor bearing C3H/He mice and YTS-1 tumor bearing nude mice. RESULTS: Intravenous injection of IF7C conjugates of either 10B drugs into MBT2 bladder tumor-bearing mice promoted rapid 10B accumulation in tumor and suppressed tumor growth. Moreover, multiple treatments at ultralow (10-20 mg/kg) doses of IF7-10B drug-mediated BNCT significantly suppressed tumor growth in a mouse model of human YTS-1 bladder cancer, with increased Anxa1 expression in tumors and infiltration by CD8-positive lymphocytes. CONCLUSIONS: We conclude that IF7 serves as an efficient 10B delivery vehicle by targeting tumor tissues via the tumor vasculature and could serve as a relevant vehicle for BNCT drugs.


Subject(s)
Annexin A1/metabolism , Boron Compounds/administration & dosage , Boron Neutron Capture Therapy/methods , Neovascularization, Pathologic/radiotherapy , Peptide Fragments/metabolism , Phenylalanine/analogs & derivatives , Urinary Bladder Neoplasms/radiotherapy , Animals , Apoptosis , Boron Compounds/chemistry , Boron Compounds/metabolism , Cell Proliferation , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Nude , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phenylalanine/administration & dosage , Phenylalanine/chemistry , Phenylalanine/metabolism , Tumor Cells, Cultured , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
12.
Org Biomol Chem ; 19(10): 2203-2212, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33496698

ABSTRACT

Here were report the combination of biocompatible click chemistry of ω-azidosphinganine with fluorescence microscopy and mass spectrometry as a powerful tool to elaborate the sphingolipid metabolism. The azide probe was efficiently synthesized over 13 steps starting from l-serine in an overall yield of 20% and was used for live-cell fluorescence imaging of the endoplasmic reticulum in living cells by bioorthogonal click reaction with a DBCO-labeled fluorophore revealing that the incorporated analogue is mainly localized in the endoplasmic membrane like the endogenous species. A LC-MS(/MS)-based microsomal in vitro assay confirmed that ω-azidosphinganine mimics the natural species enabling the identification and analysis of metabolic breakdown products of sphinganine as a key starting intermediate in the complex sphingolipid biosynthetic pathways. Furthermore, the sphinganine-fluorophore conjugate after click reaction was enzymatically tolerated to form its dihydroceramide and ceramide metabolites. Thus, ω-azidosphinganine represents a useful biofunctional tool for metabolic investigations both by in vivo fluorescence imaging of the sphingolipid subcellular localization in the ER and by in vitro high-resolution mass spectrometry analysis. This should reveal novel insights of the molecular mechanisms sphingolipids and their processing enzymes have e.g. in infection.


Subject(s)
Azides/metabolism , Sphingolipids/analysis , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Animals , Azides/chemical synthesis , Boron Compounds/chemical synthesis , Boron Compounds/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Click Chemistry , Endoplasmic Reticulum/metabolism , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Sphingolipids/biosynthesis
13.
Bioorg Chem ; 117: 105410, 2021 12.
Article in English | MEDLINE | ID: mdl-34700109

ABSTRACT

Cholesterol is not only a major component of the cell membrane, but also plays an important role in a wide range of biological processes and pathologies. It is therefore crucial to develop appropriate tools for visualizing intracellular cholesterol transport. Here, we describe new cationic analogues of BODIPY-Cholesterol (TopFluor-Cholesterol, TF-Chol), which combine a positive charge on the sterol side chain and a BODIPY group connected via a C-4 linker. In contrast to TF-Chol, the new analogues TF-1 and TF-3 possessing acetyl groups on the A ring (C-3 position on steroid) internalized much faster and displayed slightly different levels of intracellular localization. Their applicability for cholesterol monitoring was indicated by the fact that they strongly label compartments with accumulated cholesterol in cells carrying a mutation of the Niemann-Pick disease-associated cholesterol transporter, NPC1.


Subject(s)
Boron Compounds/analysis , Cholesterol/analysis , Biological Transport , Boron Compounds/chemical synthesis , Boron Compounds/chemistry , Boron Compounds/metabolism , Cell Line , Cholesterol/analogs & derivatives , Cholesterol/chemical synthesis , Cholesterol/metabolism , Humans , Optical Imaging
14.
Bioorg Chem ; 106: 104467, 2021 01.
Article in English | MEDLINE | ID: mdl-33223201

ABSTRACT

Donor-Acceptor type BODIPYs with strong absorption and fluorescence in the red region (550-800 nm) are reported. The aromatic groups like N-butylcarbazole/ N-butylphenothiazine/ benzothiadiazole were attached to the C-8 position of the BODIPY core with furan or thiophene spacers. TD-DFT studies indicated significant charge distribution between C-8 aromatic heterocycles and BODIPY core in all the molecules. The in-vitro studies of the N-butylcarbazole substituted BODIPYs indicated significant localization in the endoplasmic reticulum and lysosomes of the cancer cells. The BODIPYs showed decent cytotoxicity after 48 h incubation period (14.9 to 31.8 µM) in HeLa and A549 cancer cells, indicating their potential application as theranostic agents.


Subject(s)
Boron Compounds/pharmacology , Fluorescent Dyes/pharmacology , Heterocyclic Compounds/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Boron Compounds/chemical synthesis , Boron Compounds/metabolism , Boron Compounds/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Density Functional Theory , Drug Screening Assays, Antitumor , Endoplasmic Reticulum/metabolism , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Fluorescent Dyes/toxicity , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/metabolism , Heterocyclic Compounds/toxicity , Humans , Lysosomes/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Models, Chemical , Precision Medicine
15.
Angew Chem Int Ed Engl ; 60(36): 19912-19920, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34227724

ABSTRACT

Developing Type-I photosensitizers is considered as an efficient approach to overcome the deficiency of traditional photodynamic therapy (PDT) for hypoxic tumors. However, it remains a challenge to design photosensitizers for generating reactive oxygen species by the Type-I process. Herein, we report a series of α,ß-linked BODIPY dimers and a trimer that exclusively generate superoxide radical (O2-. ) by the Type-I process upon light irradiation. The triplet formation originates from an effective excited-state relaxation from the initially populated singlet (S1 ) to triplet (T1 ) states via an intermediate triplet (T2 ) state. The low reduction potential and ultralong lifetime of the T1 state facilitate the efficient generation of O2-. by inter-molecular charge transfer to molecular oxygen. The energy gap of T1 -S0 is smaller than that between 3 O2 and 1 O2 thereby precluding the generation of singlet oxygen by the Type-II process. The trimer exhibits superior PDT performance under the hypoxic environment.


Subject(s)
Boron Compounds/metabolism , Neoplasms/metabolism , Photochemotherapy , Photosensitizing Agents/metabolism , Singlet Oxygen/metabolism , Superoxides/metabolism , Boron Compounds/chemistry , Boron Compounds/therapeutic use , Humans , Light , Molecular Structure , Neoplasms/drug therapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Singlet Oxygen/chemistry , Superoxides/chemistry
16.
J Am Chem Soc ; 142(11): 4970-4974, 2020 03 18.
Article in English | MEDLINE | ID: mdl-32115942

ABSTRACT

Photoactivation of bioactive molecules allows manipulation of cellular processes with high spatiotemporal precision. The recent emergence of visible-light excitable photoprotecting groups has the potential to further expand the established utility of the photoactivation strategy in biological applications by offering higher tissue penetration, diminished phototoxicity, and compatibility with other light-dependent techniques. Nevertheless, a critical barrier to such applications remains the significant hydrophobicity of most visible-light excitable photocaging groups. Here, we find that applying the conventional 2,6-sulfonation to meso-methyl BODIPY photocages is incompatible with their photoreaction due to an increase in the excited state barrier for photorelease. We present a simple, remote sulfonation solution to BODIPY photocages that imparts water solubility and provides control over cellular permeability while retaining their favorable spectroscopic and photoreaction properties. Peripherally disulfonated BODIPY photocages are cell impermeable, making them useful for modulation of cell-surface receptors, while monosulfonated BODIPY retains the ability to cross the cellular membrane and can modulate intracellular targets. This new approach is generalizable for controlling BODIPY localization and was validated by sensitization of mammalian cells and neurons by visible-light photoactivation of signaling molecules.


Subject(s)
Alkanesulfonates/metabolism , Boron Compounds/metabolism , Fluorescent Dyes/metabolism , Alkanesulfonates/chemical synthesis , Alkanesulfonates/radiation effects , Animals , Boron Compounds/chemical synthesis , Boron Compounds/radiation effects , Cell Membrane/metabolism , Dopamine/chemistry , Dopamine/pharmacology , Drug Carriers/chemical synthesis , Drug Carriers/metabolism , Drug Carriers/radiation effects , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/radiation effects , HEK293 Cells , Hippocampus/drug effects , Histamine/chemistry , Histamine/pharmacology , Humans , Light , Microscopy, Confocal , Microscopy, Fluorescence , Molecular Structure , Neurons/drug effects , Rats , Solubility
17.
Anal Chem ; 92(24): 16051-16057, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33211958

ABSTRACT

Clinical identification of the pathogenic bacterium Moraxella catarrhalis in cultures relies on the detection of bacterial butyrate esterase (C4-esterase) using a coumarin-based fluorogenic substrate, 4-methylumbelliferyl butyrate. However, this classical probe may give false-positive responses because of its poor stability and lack of specificity. Here, we report a new colorimetric and fluorogenic probe design employing a meso-ester-substituted boron dipyrromethene (BODIPY) dye for the specific detection of C4-esterase activity expressed by M. catarrhalis. This new probe has resistance to nonspecific hydrolysis that is far superior to the classical probe and also selectively responds to esterase with rapid colorimetric and fluorescence signal changes and large "turn-on" ratios. The probe was successfully applied to the specific detection of M. catarrhalis with high sensitivity.


Subject(s)
Acinetobacter calcoaceticus/enzymology , Acinetobacter calcoaceticus/isolation & purification , Carboxylic Ester Hydrolases/metabolism , Colorimetry/methods , Fluorescent Dyes/metabolism , Boron Compounds/chemistry , Boron Compounds/metabolism , Fluorescent Dyes/chemistry , Limit of Detection , Time Factors
18.
Anal Chem ; 92(1): 1236-1244, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31779312

ABSTRACT

In recent years, inorganic biomimetic nanozymes that mimic the activity of natural biological enzymes have attracted extensive research interest, and some mimic enzymes have been successfully applied in the fields of biosensing, catalysis, and oncotherapy. Herein, we report the preparation and mechanism study of a novel nanocomposite, Cu2+-modified hexagonal boron nitride nanosheets-supported subnanometer gold nanoparticles (Au NPs/Cu2+-BNNS). Interestingly, our investigation reveals that Cu2+-BNNS exhibits strong peroxidase mimetic nanoenzyme activity, while Au NPs/Cu2+-BNNS exhibits excellent oxidase-like activity, that is, it can catalyze the oxidation reaction of the substrate in the absence of an oxidant such as H2O2. For example, Au NPs/Cu2+-BNNS can efficiently and selectively oxidize 3,3',5,5'-tetramethylbenzidine (TMB) and 3,3'-dimethylbiphenyl-4,4'-diamine (OT) coloration without the presence of horseradish peroxidase (HRP) and H2O2. It is worthy to note that AuNPs/Cu2+-BNNS-induced TMB coloration only takes 4 min to reach the platform, while the conventional HRP-H2O2 system takes more than 30 min to reach the platform. Further mechanism study shows that the zeta potential, oxidation potential, and steric hindrance of the oxidative chromogenic substrate determine the selectivity of oxidation coloration, while the oxidase-like properties of Au NPs/Cu2+-BNNS are derived from reactive oxygen species generated by the adsorbed oxygen, and Cu2+ ion can synergistically promote the oxidation process. Compared with conventional biological enzymes, Au NPs/Cu2+-BNNS has the advantages of being HRP free and H2O2 free, having high efficiency, low cost, and good stability, and is successfully demonstrated for the detection of carcinoembryonic antigen (a universal cancer biomarker) and H2S (the third gaseous signal molecule).


Subject(s)
Boron Compounds/metabolism , Copper/metabolism , Gold/metabolism , Horseradish Peroxidase/metabolism , Nanostructures/chemistry , Biomarkers, Tumor/analysis , Boron Compounds/chemistry , Carcinoembryonic Antigen/analysis , Copper/chemistry , Gold/chemistry , Horseradish Peroxidase/chemistry , Hydrogen Sulfide/analysis , Oxidation-Reduction , Particle Size , Surface Properties
19.
Chembiochem ; 21(8): 1129-1135, 2020 04 17.
Article in English | MEDLINE | ID: mdl-31702868

ABSTRACT

Misfolding and aggregation of amyloid ß1-42 peptide (Aß1-42) play a central role in the pathogenesis of Alzheimer's disease (AD). Targeting the highly cytotoxic oligomeric species formed during the early stages of the aggregation process represents a promising therapeutic strategy to reduce the toxicity associated with Aß1-42. Currently, the thioflavin T (ThT) assay is the only established spectrofluorometric method to screen aggregation inhibitors. The success of the ThT assay is that it can detect Aß1-42 aggregates with high ß-sheet content, such as protofibrils or fibrils, which appear in the late aggregation steps. Unfortunately, by using the ThT assay, the detection of inhibitors of early soluble oligomers that present a low ß-sheet character is challenging. Herein, a new, facile, and robust boron-dipyrromethene (BODIPY) real-time assay suitable for 96-well plate format, which allows screening of compounds as selective inhibitors of the formation of Aß1-42 oligomers, is reported. These inhibitors decrease the cellular toxicity of Aß1-42, although they fail in the ThT assay. The findings have been confirmed and validated by structural analysis and cell viability assays under comparable experimental conditions. It is demonstrated that the BODIPY assay is a convenient method to screen and discover new candidate compounds that slow down or stop the pathological early oligomerization process and are active in the cellular assay. Therefore, it is a suitable complementary screening method of the current ThT assay.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Boron Compounds/metabolism , Drug Monitoring/methods , High-Throughput Screening Assays/methods , Neuroprotective Agents/pharmacology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Small Molecule Libraries/pharmacology , Humans
20.
PLoS Pathog ; 14(2): e1006850, 2018 02.
Article in English | MEDLINE | ID: mdl-29425238

ABSTRACT

Recent development of benzoxaborole-based chemistry gave rise to a collection of compounds with great potential in targeting diverse infectious diseases, including human African Trypanosomiasis (HAT), a devastating neglected tropical disease. However, further medicinal development is largely restricted by a lack of insight into mechanism of action (MoA) in pathogenic kinetoplastids. We adopted a multidisciplinary approach, combining a high-throughput forward genetic screen with functional group focused chemical biological, structural biology and biochemical analyses, to tackle the complex MoAs of benzoxaboroles in Trypanosoma brucei. We describe an oxidative enzymatic pathway composed of host semicarbazide-sensitive amine oxidase and a trypanosomal aldehyde dehydrogenase TbALDH3. Two sequential reactions through this pathway serve as the key underlying mechanism for activating a series of 4-aminomethylphenoxy-benzoxaboroles as potent trypanocides; the methylamine parental compounds as pro-drugs are transformed first into intermediate aldehyde metabolites, and further into the carboxylate metabolites as effective forms. Moreover, comparative biochemical and crystallographic analyses elucidated the catalytic specificity of TbALDH3 towards the benzaldehyde benzoxaborole metabolites as xenogeneic substrates. Overall, this work proposes a novel drug activation mechanism dependent on both host and parasite metabolism of primary amine containing molecules, which contributes a new perspective to our understanding of the benzoxaborole MoA, and could be further exploited to improve the therapeutic index of antimicrobial compounds.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Amine Oxidase (Copper-Containing)/metabolism , Boron Compounds/metabolism , Models, Biological , Prodrugs/metabolism , Trypanocidal Agents/metabolism , Trypanosoma brucei brucei/enzymology , Activation, Metabolic , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/chemistry , Aldehyde Dehydrogenase/genetics , Aldehyde Oxidoreductases/antagonists & inhibitors , Aldehyde Oxidoreductases/chemistry , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Amine Oxidase (Copper-Containing)/chemistry , Amine Oxidase (Copper-Containing)/genetics , Amino Acid Substitution , Animals , Boron Compounds/chemistry , Boron Compounds/pharmacology , Drug Resistance , High-Throughput Screening Assays , Humans , Molecular Structure , Mutation , Phylogeny , Prodrugs/chemistry , Prodrugs/pharmacology , Protein Interaction Domains and Motifs , RNA Interference , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/physiology
SELECTION OF CITATIONS
SEARCH DETAIL