Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Mater Chem B ; 12(17): 4248-4261, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38602387

ABSTRACT

Prolonged use of very commonly prescribed non-steroidal anti-inflammatory drugs (NSAIDs) is often associated with undesired side effects, including gastrointestinal ulcers due to the non-selective inhibition of cyclooxygenases. We describe the development of an inflammatory-stimuli-responsive turn-on fluorogenic theranostic prodrug DCF-HS for adjuvant drug delivery. Upon activation by reactive oxygen species (ROS), the prodrug releases diclofenac DCF (active drug) and the NIR fluorophore DCI-NH2 along with carbonyl sulfide (COS). The second activation of COS by the enzyme carbonic anhydrase (CA) generates hydrogen sulfide (H2S). The prodrug was conveniently synthesized using multi-step organic synthesis. The UV-Vis and fluorescence studies revealed the selective reactivity of DCF-HS towards ROS such as H2O2 in the aqueous phase and the desired uncaging of the drug DCF with turn-on NIR fluorescent reporter under physiological conditions. Furthermore, the release of fluorophore DCI-NH2 and drug DCF was confirmed using the reverse phase HPLC method. Compatibility of prodrug activation was studied next in the cellular medium. The prodrug DCF-HS was non-toxic in a representative cancer cell line (HeLa) and a macrophage cell line (RAW 264.7) up to 100 µM concentration, indicating its biocompatibility. The intracellular ROS-mediated activation of the prodrug with the release of NIR dye DCI-NH2 and H2S was investigated in HeLa cells using the H2S-selective probe WSP2. The anti-inflammatory activity of the active drug DCF from the prodrug DCF-HS was studied in the lipopolysaccharide (LPS)-induced macrophage cell line and compared to that of the parent drug DCF using western blot analysis and it was found that the active drug resulted in pronounced inhibition of COX-2 in a dose-dependent manner. Finally, the anti-inflammatory potential of the prodrug and the turn-on fluorescence were validated in the inflammation-induced Wister rat models.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Diclofenac , Hydrogen Sulfide , Prodrugs , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/chemical synthesis , Hydrogen Sulfide/metabolism , Animals , Humans , Diclofenac/pharmacology , HeLa Cells , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Rats , Theranostic Nanomedicine , Inflammation/drug therapy , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Fluorescent Dyes/chemical synthesis , Mice , RAW 264.7 Cells , Drug Delivery Systems , Edema/drug therapy , Edema/chemically induced
2.
Chemistry ; 29(66): e202302197, 2023 Nov 24.
Article in English | MEDLINE | ID: mdl-37665099

ABSTRACT

Stimuli-responsive fluorogenic prodrugs are advantageous for the targeted drug delivery enabling real-time non-invasive monitoring with turn-on fluorescence. We report herein the dual-stimuli (ROS and CA)-responsive thiocarbamate-based prodrug (AM-TCB) for the turn-on fluorogenic delivery of the naphthalimide-based anticancer agent amonafide along with the gasotransmitter hydrogen sulfide (H2 S). A carbamate-based prodrug AM-CB was also designed, capable of releasing the anticancer agent amonafide without any H2 S. The prodrugs were synthesized using multi-step organic synthesis. UV-Vis and fluorescence spectroscopic studies revealed selective reactivity of the boronate ester group of prodrugs towards ROS (primarily H2 O2 ) with the release of amonafide and COS/CO2 via self-immolative processes. Hydrolysis of the generated COS by carbonic anhydrase (CA) produces H2 S. While the prodrug AM-TCB retained the anticancer activity of free amonafide in cancer cells (MDA-MB-231 and HeLa), unlike amonafide, it enhanced the cellular viability of the non-malignant cells (HEK-293). Fluorescence imaging in HeLa cells revealed the simultaneous delivery of the anticancer agent and H2 S from AM-TCB with turn-on fluorescence. Western blot studies further revealed the cytoprotective effects of the released H2 S from AM-TCB. The present adjuvant strategy therefore would be helpful in future for ameliorating the anticancer drug-induced side-effects.


Subject(s)
Antineoplastic Agents , Hydrogen Sulfide , Prodrugs , Humans , Prodrugs/pharmacology , Prodrugs/chemistry , Hydrogen Sulfide/chemistry , HeLa Cells , Fluorescence , Reactive Oxygen Species , HEK293 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry
3.
Chem Commun (Camb) ; 59(32): 4802-4805, 2023 Apr 18.
Article in English | MEDLINE | ID: mdl-37000594

ABSTRACT

L-Cysteine (Cys)-responsive turn-on fluorogenic prodrug AM-ITC was developed for the adjuvant delivery of the anti-cancer drug amonafide and the gasotransmitter hydrogen sulfide (H2S) in aqueous and cellular media. Considering the cytoprotective roles of H2S, the present adjuvant strategy would be helpful in minimizing the anti-cancer drug-induced side-effects.


Subject(s)
Antineoplastic Agents , Hydrogen Sulfide , Prodrugs , Hydrogen Sulfide/pharmacology , Cysteine , Prodrugs/pharmacology , Antineoplastic Agents/pharmacology , Adjuvants, Immunologic
4.
Chem Commun (Camb) ; 58(56): 7833-7836, 2022 Jul 12.
Article in English | MEDLINE | ID: mdl-35748501

ABSTRACT

Reactive oxygen species (ROS)-responsive near infrared (NIR) fluorogenic prodrug DCI-ROS is developed for the self-immolative release of diclofenac (DCF) with turn-on fluorescence. The non-toxic prodrug exhibited turn-on red fluorescence with endogenous ROS in cancer cells and inhibited COX-2 expression in the inflammation-induced macrophage cells. The prodrug strategy thus would be helpful for the controlled fluorogenic delivery of DCF for inflammatory diseases.


Subject(s)
Prodrugs , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Diclofenac/pharmacology , Drug Liberation , Fluorescence , Prodrugs/pharmacology , Reactive Oxygen Species/metabolism
5.
J Mater Chem B ; 10(13): 2183-2193, 2022 03 30.
Article in English | MEDLINE | ID: mdl-35266940

ABSTRACT

We describe herein the rational development of an organopolysulfide-based fluorogenic donor of hydrogen sulfide (H2S) DCI-PS, which can be activated by the antioxidant selenoenzyme thioredoxin reductase (TrxR) with concomitant release of the dicyanoisophorone-based near-infrared (NIR) fluorophore. Along with the polysulfide probe DCI-PS capable of releasing the NIR fluorophore and H2S, the corresponding disulfide-probe DCI-DS was also rationally designed and synthesized, which releases the fluorophore without donating H2S. Detailed spectroscopic and kinetic studies in an aqueous medium revealed significantly higher reactivity of the probes towards DTT (for TrxR activity) over the well-known cellular abundant biothiol GSH. Mechanistically, the nucleophilic attack at the disulfide/polysulfide linkage by the thiol/selenol group of the bio-analytes leads to the self-immolative cyclization process with the release of the turn-on fluorophore with/without H2S. Considering the overexpression of mammalian TrxR in cancer cells, the turn-on fluorogenic H2S donation process from the cellular non-toxic DCI-PS was validated in a representative breast cancer cell line (MDA-MB-231) for the sustained donation of H2S with concomitant release of the red-emitting NIR fluorophore. The TrxR-triggered fluorescence turn-on process in DCI-PS was further supported by the significant inhibition of the fluorogenic process in the presence of TrxR-selective small-molecule inhibitors and by the significant binding affinity predicted by the protein-ligand docking study. Results with the antioxidant enzyme-triggered intracellular sustained donation of H2S with concomitant fluorescence turn-on will certainly find wider biomedical applications in the near future, particularly in H2S-mediated therapeutics in disease states.


Subject(s)
Hydrogen Sulfide , Animals , Fluorescence , Fluorescent Dyes/chemistry , Hydrogen Sulfide/chemistry , Kinetics , Mammals/metabolism , Thioredoxin-Disulfide Reductase/metabolism
6.
Inorg Chem ; 58(8): 4741-4752, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30920210

ABSTRACT

Mycobacterium tuberculosis ( Mtb) expresses heme binding protein nanocages, bacterioferritin A (BfrA), along with nonheme bacterioferritin B (BfrB). BfrA is unique to bacteria and, like BfrB, carries out ferroxidase activity to synthesize iron oxide biominerals. The expression of BfrA, in the presence of BfrB, indicates that Mtb may utilize it for some additional purpose apart from its natural iron storage activity. However, the mechanism of ferroxidase activity (iron biomineralization) in Mtb BfrA still remains unexplored. H2O2 is secreted by the host during host-pathogen interaction. In some bacteria, heme containing Bfr and/or Dps (DNA binding protein during starvation) detoxify H2O2 by utilizing it during their ferroxidase activity. Interestingly, Mtb lacks the gene for Dps which protects DNA from H2O2-induced oxidative cleavage. Therefore, the current work investigates the kinetics of O2/H2O2-dependent ferroxidase activity, DNA protection, and catalase-like activity of recombinant Mtb BfrA. Ferroxidase activity by Mtb BfrA was found to proceed via the formation of a transient intermediate and its initial rate exhibited sigmoidal behavior, with increasing Fe2+ concentration. Moreover, Mtb BfrA exhibited catalase-like activity by evolving O2 upon reaction with H2O2, which gets inhibited in the presence of catalase inhibitors (NaN3 and NaCN). In addition, Mtb BfrA protected plasmid DNA from Fenton reagents (Fe2+ and H2O2), similar to Dps, by forming BfrA-DNA complexes. Thereby, Mtb BfrA executes multiple functions (ferroxidase, catalase, and Dps-like activities) in order to cope with the host generated oxidative stress and to promote pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Catalase/metabolism , Cytochrome b Group/metabolism , Ferritins/metabolism , Iron/metabolism , Mycobacterium tuberculosis/metabolism , DNA/metabolism , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Iron/chemistry , Microscopy, Electron, Transmission , Molecular Mimicry , Oxidation-Reduction , Oxygen Consumption , Plasmids , Spectrophotometry, Ultraviolet
SELECTION OF CITATIONS
SEARCH DETAIL
...