Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
Angew Chem Int Ed Engl ; 60(3): 1403-1410, 2021 01 18.
Article in English | MEDLINE | ID: mdl-33029903

ABSTRACT

Despite widespread applications for cancer treatment, chemotherapy is restricted by several limitations, including low targeting specificity, acquired drug resistance, and concomitant adverse side effects. It remains challenging to overcome these drawbacks. Herein, we report a new bioenergetic approach for treating cancer efficiently. As a proof-of-concept, we construct activatable mitochondria-targeting organoarsenic prodrugs from organoarsenic compounds and traditional chemotherapeutics. These prodrugs could accomplish selective delivery and controlled release of both therapeutic agents to mitochondria, which synergistically promote mitochondrial ROS production and induce mitochondrial DNA damage, finally leading to mitochondria-mediated apoptosis of cancer cells. Our in vitro and in vivo experiments reveal the excellent anticancer efficacy of these prodrugs, underscoring the encouraging outlook of this strategy for effective cancer therapy.


Subject(s)
Energy Metabolism/genetics , Mitochondria/metabolism , Neoplasms/therapy , Prodrugs/chemistry
3.
Autophagy ; 17(6): 1426-1447, 2021 06.
Article in English | MEDLINE | ID: mdl-32397800

ABSTRACT

Dysfunction of macroautophagy/autophagy has been postulated as a major cellular toxicological response to nanomaterials. It has been reported that excessive autophagy activation, induced by silica nanoparticles (SiNPs), contributes to autophagy dysfunction, whereas little is known how SiNPs trigger autophagy activation. Here, we treated normal rat kidney (NRK) cells using 3 different sizes of SiNPs (16, 29, and 51 nm) and observed that 16-nm SiNPs, with a final concentration of 60 µg/mL, dramatically induce autophagy activation without reducing cell viability. We further conducted a transcriptomic, proteomic, and phosphoproteomic profiling, and detected 23 autophagy-related (Atg) genes and 35 autophagy regulators regulated on at least one omic layer. To identify key regulators from the multi-omics data, we developed a new algorithm of computational prediction of master autophagy-regulating kinases (cMAK) to detect 21 candidates and revealed the CDK7-CDK4 cascade to be functional. The silence or inhibition of Cdk7 or Cdk4 significantly attenuated autophagic activation but not influenced autophagic flux blockage induced by 16-nm SiNPs. Further computational modeling indicated that the CDK7-CDK4 signaling axis potentially triggers autophagy activation by phosphorylating RB1 (RB transcriptional corepressor 1), activating two critical transcription factors, E2F1 (E2F transcription factor 1) and FOXO3 (forkhead box O3), and enhancing the transcriptional levels of at least 8 Atg genes and autophagy regulators in response to SiNPs. Our studies not only established a powerful method for predicting regulatory kinases from the multi-omics data but also revealed a potential mechanism of SiNP-triggered autophagy activation through modulating the CDK7-CDK4 cascade.Abbreviations: 3-MA: 3-methyladenine; Atg: autophagy-related; BECN1: beclin 1; CCK-8: cell counting kit-8; CDK4: cyclin dependent kinase 4; CDK7: cyclin dependent kinase 7; cMAK: computational prediction of master autophagy-regulating kinases; CQ: chloroquine; DMEM: Dulbecco's modified Eagle's medium; DMSO: dimethyl sulfoxide; E-ratio: enrichment ratio; E2F1: E2F transcription factor 1; EBSS: Earle's balanced salt solution; ER: endoplasmic reticulum; FOXO3: forkhead box O3; FPKM: fragments per kilobase of exon per million fragments mapped; GO: gene ontology; H2O2: hydrogen peroxide; iGPS: in vivo GPS; KEGG: Kyoto Encyclopedia of Genes and Genomes; LC-MS/MS: liquid chromatography-tandem mass spectrometry; LDH: lactate dehydrogenase; MAP1LC3B/LC3: microtubule associated protein 1 light chain 3 beta; NRK: normal rat kidney; p-site: phosphorylation site; PBS: phosphate-buffered saline; PDI: polydispersity index; PTM: post-translational modification; QKS: quantitative kinase state; RB1: RB transcriptional corepressor 1; RBHs: reciprocal best hits; RNA-Seq: RNA sequencing; ROS: reactive oxygen species; rSiNPs: SiNPs fluorescently labeled with rhodamine B; SEM: scanning electronic microscopy; SiNPs: silica nanoparticles; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; ssKSR: site-specific kinase-substrate relation; TEM: transmission electron microscopy; tfLC3: mRFP-GFP tandem fluorescent-tagged LC3.


Subject(s)
Autophagy/physiology , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinases/metabolism , Nanoparticles , Humans , Lysosomes/metabolism , Nanoparticles/metabolism , Proteomics/methods , Signal Transduction/physiology , Silicon Dioxide/metabolism , TOR Serine-Threonine Kinases/metabolism , Tandem Mass Spectrometry/methods , Cyclin-Dependent Kinase-Activating Kinase
4.
Anal Chem ; 92(24): 16293-16300, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33252213

ABSTRACT

Magnetic resonance imaging (MRI) is one of the most popular imaging techniques, which offers an ionization-free noninvasive means for imaging deep tissues with high resolution. Conventional 1H MRI is well versed in providing detailed anatomical information but suffers from low contrast for tracking biomarkers because of the abundance of water in living bodies. 19F MRI with negligible endogenous background interference enables highly sensitive detection of biomolecular targets and has drawn extensive attention from the biomedical research community recently. However, this imaging technique only acquires the "hot spot" signals of exogenous 19F nucleus-containing imaging probes. 1H/19F MRI dual-modal imaging is expected to compensate for the limitations of either single-modal imaging and accomplish synergistic morphological and physiological imaging. Herein, we report a highly biocompatible nanoconjugate composed of pH-responsive 19F nucleus-bearing Gd3+ chelates, which enables significant contrast enhancement for T1-weighted 1H MRI and permits pH-responsive activation of 19F signals for 19F MRI, providing both clear anatomical details of living bodies and the biorelevant molecular information with low background interference. This nanoconjugate facilitates sensitive and accurate detection of tumors with contrast-enhanced T1-weighted 1H and pH-activatable 19F dual-modal imaging on a single MRI scanner.


Subject(s)
Chelating Agents/chemistry , Gadolinium/chemistry , Halogenation , Magnetic Resonance Imaging/methods , Nanoconjugates/chemistry , Hydrogen-Ion Concentration
5.
Chem Commun (Camb) ; 56(29): 4106-4109, 2020 Apr 09.
Article in English | MEDLINE | ID: mdl-32163087

ABSTRACT

Herein we report a pair of redox-responsive manganese complexes Mn(iii)/(ii)-N,N'-bis(2-hydroxy-4-trifluoromethylbenzyl)ethylenediamine-N,N'-diacetate (HTFBED, L1), which are water soluble and biologically interconvertible, as reversible redox-responsive probes in 1H/19F MRI for detecting and imaging biological redox species, offering a means to access valuable redox information associated with various diseases.


Subject(s)
Coordination Complexes , Edetic Acid , Manganese , Molecular Probes , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Coordination Complexes/administration & dosage , Coordination Complexes/chemistry , Edetic Acid/administration & dosage , Edetic Acid/analogs & derivatives , Edetic Acid/chemistry , Hep G2 Cells , Humans , Magnetic Resonance Imaging , Manganese/administration & dosage , Manganese/chemistry , Molecular Probes/administration & dosage , Molecular Probes/chemistry , Oxidation-Reduction , Pyocyanine/pharmacology
6.
J Am Chem Soc ; 142(1): 198-206, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31823608

ABSTRACT

Multinuclear complexes as metallo-agents for clinical use have caught extensive attention. In this paper, using 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) as both a functioning unit and a constructing junction, we build a series of DOTA-branched organic frameworks with multiple chelating holes by organizing DOTA layer by layer. These giant chelators are well characterized, which reveals their nanosized and soft structures. Further experiments demonstrate that they could efficiently hold abundant metal ions with much higher kinetic stabilities than the conventional small DOTA chelator. Their corresponding polynuclear complexes containing Gd3+, Tb3+, or both show superior imaging properties, excellent feasibility for peripheral modification, and unusual kinetic stability. This work can be easily extended to the fabrication of diverse homomultinuclear complexes and core/shell heteromultinuclear complexes with multifunctional properties. We expect that this new type of giant molecules and the ligand-branching strategy would open up a new avenue for the design and construction of next-generation polymetallic agents with high performance and stabilities for biomedical applications.

7.
Nano Lett ; 20(1): 363-371, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31838855

ABSTRACT

Molecular probes featuring promising capabilities including specific targeting, high signal-to-noise ratio, and in situ visualization of deep tissues are in great demand for tumor diagnosis and therapy. 19F magnetic resonance imaging (MRI) techniques incorporating stimuli-responsive probes are anticipated to be highly beneficial for specific detection and imaging of tumors because of negligible background and deep tissue penetration. Herein, we report a cascaded multiresponsive self-assembled nanoprobe, which enables sequential redox-triggered and near-infrared (NIR) irradiation-induced 19F MR signal activation/amplification for sensing and imaging. Specifically, we designed and synthesized a cascaded multiresponsive 19F-bearing nanoprobe based on the self-assembly of amphiphilic redox-responsive 19F-containing polymers and NIR-absorbing indocyanine green (ICG) molecules. It could realize the activation of 19F signals in the reducing tumor microenvironment and subsequent signal amplification via the photothermal process. This stepwise two-stage activation/amplification of 19F signals was validated by 19F NMR and MRI both in vitro and in vivo. The multiresponsive 19F nanoprobes capable of cascaded 19F signal activation/amplification and photothermal effect exertion can provide accurate sensing and imaging of tumors.


Subject(s)
Fluorine-19 Magnetic Resonance Imaging , Infrared Rays , Liver Neoplasms, Experimental/diagnostic imaging , Nanoparticles , Tumor Microenvironment/drug effects , Animals , Female , Hep G2 Cells , Humans , Mice , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/therapeutic use
8.
Chem Commun (Camb) ; 55(83): 12455-12458, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31565704

ABSTRACT

Hypochlorous acid (HClO) is one of the most important reactive oxygen species (ROS) and plays a vital role in many physiological and pathological processes. The comprehensive exploration of mechanistic details and the potential clinical translation necessitate the development of reliable probes for prompt and accurate detection of HClO in complex biological environments. Herein we report a fluorinated bihydrazide conjugate as a 19F NMR/MRI probe with a "turn-on" character for the detection of HClO. This probe could selectively respond to HClO, leading to a significant recovery of 19F signals for 19F NMR/MRI. Activatable sensing and imaging of HClO were achieved with SMMC-7721 cells and nude mice, which demonstrates that this small molecular conjugate could serve as a selective probe for real-time sensing and imaging of HClO in biological systems.


Subject(s)
Fluorescent Dyes/chemistry , Hydrazines/chemistry , Hypochlorous Acid/analysis , Magnetic Resonance Imaging , Animals , Cell Line, Tumor , Fluorine Radioisotopes , Halogenation , Humans , Magnetic Resonance Spectroscopy , Mice , Mice, Nude , Reactive Oxygen Species
9.
Nano Lett ; 19(8): 5394-5402, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31286778

ABSTRACT

Multifunctional nanoplatforms featuring promising properties including excellent loading efficiency, real-time monitoring, and improved cargo bioavailability and bioselectivity are in great demand by the biomedical research community. During the development of such nanoplatforms, stimuli-responsive nanoparticles (NPs) as a smart nanoplatform have recently received extensive attention. Herein, we report small-sized octapod-shaped hollow porous manganese(II) oxide (HPMO) NPs as a stimuli-responsive T1-activatable nanoplatform for tumor-specific cargo delivery and real-time monitoring. The HPMO NPs functionalized by zwitterionic dopamine sulfonate (ZDS) can act as a versatile platform to load organic dyes or chemotherapeutic drugs with high loading efficiency. The obtained Cargo@HPMO would decompose into paramagnetic Mn2+ ions and subsequently release cargoes in mild acidic conditions, especially in tumor microenvironment and lysosome. The released Mn2+ can enhance T1 magnetic resonance signal for real-time monitoring of the cargo delivery in vivo. This octapod-shaped Cargo@HPMO can act as a smart and versatile nanoplatform with pH-responsive multimodal imaging and site-specific drug delivery for the development of accurate diagnosis and effective therapy for cancer.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Delayed-Action Preparations/chemistry , Dopamine/analogs & derivatives , Doxorubicin/administration & dosage , Manganese Compounds/chemistry , Oxides/chemistry , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/therapeutic use , Cell Line, Tumor , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Drug Liberation , Hydrogen-Ion Concentration , Mice, Inbred BALB C , Neoplasms/drug therapy , Porosity
10.
Nanotechnology ; 30(17): 175101, 2019 Apr 26.
Article in English | MEDLINE | ID: mdl-30654348

ABSTRACT

Arsenic trioxide (ATO), an FDA-approved drug for acute promyelocytic leukemia, also has great potential for treatment of solid tumors. Drug delivery powered by recent advances in nanotechnology has boosted the efficacy of many drugs, which is enlightening for applications of ATO in treating solid tumors. Herein, we reported arsenite-loaded multifunctional nanoparticles that are capable of pH-responsive ATO release for treating hepatocellular carcinoma (HCC) and real-time monitoring via magnetic resonance imaging. We fabricated these nanoparticles (designated as magnetic large-pore mesoporous silica nanoparticle (M-LPMSN)-NiAsO x ) by loading nanoparticulate ATO prodrugs (NiAsO x ) into the pores of large-pore mesoporous silica nanoparticles (LPMSNs) that contain magnetic iron oxide nanoparticles in the center. The surface of these nanodrugs was modified with a targeting ligand folic acid (FA) to further enhance the drug efficacy. Releasing profiles manifest the responsive discharging of arsenite in acidic environment. In vitro experiments with SMMC-7721 cells reveal that M-LPMSN-NiAsO x -FA nanodrugs have significantly higher cytotoxicity than traditional free ATO and induce more cell apoptosis. In vivo experiments with mice bearing H22 tumors further confirm the superior antitumor efficacy of M-LPMSN-NiAsO x -FA over traditional free ATO and demonstrate the outstanding imaging ability of M-LPMSN-NiAsO x -FA for real-time tumor monitoring. These targeted arsenite-loaded magnetic mesoporous silica nanoparticles integrating imaging and therapy hold great promise for treatment of HCC, indicating the auspicious potential of LPMSN-based nanoplatforms.


Subject(s)
Antineoplastic Agents/administration & dosage , Arsenites/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Drug Carriers , Liver Neoplasms/drug therapy , Magnetite Nanoparticles/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Arsenites/chemistry , Arsenites/pharmacokinetics , Cell Line, Tumor , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Female , Humans , Magnetite Nanoparticles/administration & dosage , Mice, Inbred BALB C
11.
Biomater Sci ; 7(1): 262-271, 2018 Dec 18.
Article in English | MEDLINE | ID: mdl-30465673

ABSTRACT

Cisplatin (CDDP) and arsenic trioxide (ATO), two representative inorganic anticancer drugs, have been successful in the treatment against several kinds of malignancies. However, combination therapy with these two drugs in clinical application suffers from poor pharmacokinetics, serious side effects, and drug resistance of the tumor. Herein, we report a carrier-free aquo-cisplatin arsenite multidrug nanocomposite loaded with cisplatin and arsenic trioxide prodrugs simultaneously. This nanocomposite achieves a high loading capacity and pH-dependent controlled release of the drugs. Because of these features, this nanocomposite shows better in vitro toxicity against various carcinoma cell lines than either the single drug or free drug combination, promotes the synergistic effect of cisplatin and arsenic trioxide, and significantly inhibits the growth of tumors in vivo. Furthermore, cisplatin and arsenic trioxide in this nanocomposite can realize a coordination of both enhanced DNA damage and DNA repair interference within cisplatin-resistant cells, which results in overcoming the drug resistance effectively. Gene expression profiles demonstrate the reduced expression of proto-oncogenes and DNA damage repair related genes MYC, MET, and MSH2, along with the increase of tumor suppressor genes PTEN, VHL, and FAS after the nanocomposite treatment. This type of multidrug nanocomposite offers an alternative and promising strategy for combination therapy and overcoming drug resistance.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Arsenic Trioxide/analogs & derivatives , Arsenic Trioxide/pharmacology , Cisplatin/analogs & derivatives , Cisplatin/pharmacology , Nanocomposites/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Arsenic Trioxide/chemical synthesis , Arsenic Trioxide/therapeutic use , Arsenites/chemical synthesis , Arsenites/chemistry , Arsenites/pharmacology , Cell Line, Tumor , Cisplatin/chemical synthesis , Cisplatin/therapeutic use , DNA Damage/drug effects , Drug Resistance, Neoplasm , Humans , Male , Mice, Inbred BALB C , Nanocomposites/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Transcriptome/drug effects
12.
J Mater Chem B ; 6(48): 8127-8136, 2018 Dec 28.
Article in English | MEDLINE | ID: mdl-32254932

ABSTRACT

The physicochemical properties of nanoparticles have been tuned via various synthetic methods to improve their diagnostic or curative capability. However, systematic understanding of the relationship between their physicochemical properties and biological effects is still not well established. Particularly, the latent ability of nanomaterials to regulate autophagy has already drawn more attention. In this report, by comparing cellular interactions, uptakes, and autophagic effects of gold nanoparticles with different shapes, we reveal that gold nanoparticles could modulate autophagy in a shape-dependent manner. Western blot assays and confocal images confirm that nanospheres cause more autophagosome accumulation than nanorods, which are highly correlated with the difference in cellular uptakes. With biological TEM, we observe remarkable lysosome swelling and clearly identify the engulfed gold nanoparticles together with undegraded organelles in autolysosomes. Additionally, monitoring of the lysosomal activity and p62 degradation indicates an autophagy flux decrease induced by the impairment of lysosomes after treatment with nanoparticles. Our study not only reveals the effects of nanostructure morphology on autophagy, but also provides an alternative strategy to modulate autophagy, which would contribute to the guidelines for further biomedical applications of various nanomaterials.

SELECTION OF CITATIONS
SEARCH DETAIL
...