Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Proc Natl Acad Sci U S A ; 120(51): e2311276120, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38079547

ABSTRACT

Although the tremendous progress has been made for mRNA delivery based on classical cationic carriers, the excess cationic charge density of lipids was necessary to compress mRNA through electrostatic interaction, and with it comes inevitably adverse events including the highly inflammatory and cytotoxic effects. How to develop the disruptive technologies to overcome cationic nature of lipids remains a major challenge for safe and efficient mRNA delivery. Here, we prepared noncationic thiourea lipids nanoparticles (NC-TNP) to compress mRNA by strong hydrogen bonds interaction between thiourea groups of NC-TNP and the phosphate groups of mRNA, abandoning the hidebound and traditional electrostatic force to construct mRNA-cationic lipids formulation. NC-TNP was a delivery system for mRNA with simple, convenient, and repeatable preparation technology and showed negligible inflammatory and cytotoxicity side effects. Furthermore, we found that NC-TNP could escape the recycling pathway to inhibit the egress of internalized nanoparticles from the intracellular compartment to the extracellular milieu which was a common fact in mRNA-LNP (lipid nanoparticles) formulation. Therefore, NC-TNP-encapsulated mRNA showed higher gene transfection efficiency in vitro and in vivo than mRNA-LNP formulation. Unexpectedly, NC-TNP showed spleen targeting delivery ability with higher accumulation ratio (spleen/liver), compared with traditional LNP. Spleen-targeting NC-TNP with mRNA exhibited high mRNA-encoded antigen expression in spleen and elicited robust immune responses. Overall, our work establishes a proof of concept for the construction of a noncationic system for mRNA delivery with good inflammatory safety profiles, high gene transfection efficiency, and spleen-targeting delivery to induce permanent and robust humoral and cell-mediated immunity for disease treatments.


Subject(s)
Biomimetics , Nanoparticles , RNA, Messenger/metabolism , Lipids/chemistry , Nanoparticles/chemistry , Cations/chemistry , Thiourea , RNA, Small Interfering/genetics
2.
Acc Chem Res ; 57(6): 905-918, 2024 03 19.
Article in English | MEDLINE | ID: mdl-38417027

ABSTRACT

Cancer vaccines have shown tremendous potential in preventing and treating cancer by providing immunogenic antigens to initiate specific tumor immune responses. An in situ vaccine prepared from an autologous tumor can mobilize a patient's own tumor cell lysate as a reservoir of specific antigens, thus triggering a broad immune response and diverse antitumor immunity in an individually tailored manner. Its efficacy is much better than that of conventional vaccines with a limited number of epitopes. Several conventional therapies, including radiotherapy (RT), chemotherapeutics, photodynamic therapy (PDT), and photothermal therapy (PTT) can activate an anticancer in situ vaccine response by inducing immunogenic cell death (ICD), triggering the exposure of tumor-associated antigens (TAAs), cancerous testis antigens, neoantigens, and danger-associated molecular patterns (DAMPs) with low cost. However, the immunogenicity of dying tumor cells is low, making released antigens and DAMPs insufficient to initiate a robust immune response against malignant cancer. Moreover, the immunosuppressive tumor microenvironment (TME) severely hinders the infiltration and sensitization of effector immune cells, causing tolerogenic immunological effects.Herein, we mainly focus on the research in developing nanoplatforms to surmount the major challenges met by ICD-based in situ vaccines. We first summarized a variety of nanotechnologies that enable enhanced immunogenicity of dying cancer cells by enhancing antigenicity and adjuvanticity. The robust antigenicity was obtained via regulating the tumor cells death mode or the dying state to amplify the recognition of tumor debris by professional antigen-presenting cells (APCs). The adjuvanticity was potentiated by raising the level or intensifying the activity of endogenous adjuvants or promoting the intelligent delivery of exogenous immunostimulants to activate immune cell recruitment and promote antigen presentation. Additionally, versatile approaches to reverse immunosuppressive TME to boost the in situ tumor vaccination response are also highlighted in detail. On one hand, by modulating the cell metabolism in TME, the expansion and activity of effector versus immunosuppressive cells can be optimized to improve the efficiency of in situ vaccines. On the other hand, regulating cellular components in TME, such as reversing adverse immune cell phenotypes or inhibiting the activity of interstitial cells, can also significantly enhance the ICD-based antitumor immunotherapy effect. Finally, our viewpoint on the future challenges and opportunities in this hopeful area is presented. We expect that this Account can offer much more insight into the design, planning, and development of cutting-edge in situ tumor vaccine platforms, promoting more attention and academic-industry collaborations, accelerating the advanced progress of in situ tumor vaccine-based immunotherapy in the clinic.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , Cancer Vaccines/therapeutic use , Nanomedicine , Immunogenic Cell Death , Neoplasms/therapy , Vaccination , Adjuvants, Immunologic , Tumor Microenvironment
3.
Acc Chem Res ; 56(13): 1766-1779, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37314368

ABSTRACT

Development of multifunctional nanoparticles (NPs) with desired properties is a significant topic in the field of nanotechnology and has been anticipated to revolutionize cancer diagnosis and treatment modalities. The surface character is one of the most important parameters of NPs that can directly affect their in vivo fate, bioavailability, and final theranostic outcomes and thus should be carefully tuned to maximize the diagnosis and treatment effects while minimizing unwanted side effects. Surface engineered NPs have utilized various surface functionality types and approaches to meet the requirements of cancer therapy and imaging. Despite the various strategies, these surface modifications generally serve similar purposes, namely, introducing therapeutic/imaging modules, improving stability and circulation, enhancing targeting ability, and achieving controlled functions. These surface engineered NPs hence could be applied in various cancer diagnosis and treatment scenarios and continuously contribute to the clinical translation of the next-generation NP-based platforms toward cancer theranostics.In this Account, we present recent advances and research efforts on the development of NP surface engineering toward cancer theranostics. First, we summarize the general strategies for NP surface engineering. Various types of surface functionalities have been applied including inorganic material-based functionality, organic material-based functionality like small molecules, polymers, nucleic acids, peptides, proteins, carbohydrates, antibodies, etc., and biomembrane-based functionality. These surface modifications can be realized by prefabrication or postfabrication functionalization, driven by covalent conjugations or noncovalent interactions. Second, we highlight the general aims of these different NPs surface functionalities. Different therapeutic and diagnostic modules, such as nanozymes, antibodies, and imaging contrast agents, have been modified on the surface of NPs to achieve theranostic function. Surface modification also can improve stability and circulation of NPs by protecting the NPs from immune recognition and clearance. In addition, to achieve targeted therapy and imaging, various targeting moieties have been attached on the NP surface to enhance active targeting ability to tissues or cells of interest. Furthermore, the NP surfaces can be tailored to achieve controlled functions which only respond to specific internal (e.g., pH, thermal, redox, enzyme, hypoxia) or external (e.g., light, ultrasound) triggers at the precise action sites. Finally, we present our perspective on the remaining challenges and future developments in this significant and rapidly evolving field. We hope this Account can offer an insightful overlook on the recent progress and an illuminating prospect on the advanced strategies, promoting more attention in this area and adoption by more scientists in various research fields, accelerating the development of NP surface engineering with a solid foundation and broad cancer theranostics applications.


Subject(s)
Nanoparticles , Neoplasms , Humans , Precision Medicine , Neoplasms/diagnostic imaging , Neoplasms/therapy , Nanoparticles/therapeutic use , Nanoparticles/chemistry , Engineering
4.
Ecotoxicol Environ Saf ; 255: 114774, 2023 Apr 15.
Article in English | MEDLINE | ID: mdl-36931087

ABSTRACT

Although numerous investigations on the adverse impact of Cr and Pb have been performed, studies on intestinal homeostasis in amphibians are limited. Here, single and combined effects of Cr (104 µg/L) and Pb (50 µg/L) on morphological and histological features, bacterial community, digestive enzymes activities, as well as transcriptomic profile of intestines in Rana chensinensis tadpoles were assessed. Significant decrease in the relative intestine length (intestine length/snout-to-vent length, IL/SVL) was observed after exposure to Pb and Cr/Pb mixture. Intestinal histology and digestive enzymes activities were altered in metal treatment groups. In addition, treatment groups showed significantly increased bacterial richness and diversity. Tadpoles in treatment groups were observed to have differential gut bacterial composition from controls, especially for the abundance of phylum Proteobacteria, Firmicutes, Verrucomicrobia, Actinobacteria, and Fusobacteria as well as genus Citrobacter, Anaerotruncus, Akkermansia, and Alpinimonas. Moreover, transcriptomic analysis showed that the transcript expression profiles of GPx and SOD isoforms responded differently to Cr and/or Pb exposure. Besides, transcriptional activation of pro-apoptotic and glycolysis-related genes, such as Bax, Apaf 1, Caspase 3, PK, PGK, TPI, and GPI were detected in all treatment groups but downregulation of Bcl2 in Pb and Cr/Pb mixture groups. Collectively, these results suggested that Cr and Pb exposure at environmental relevant concentration, alone and in combination, could disrupt intestinal homeostasis of R. chensinensis tadpoles.


Subject(s)
Gastrointestinal Microbiome , Intestines , Animals , Larva , Lead/toxicity , Ranidae/genetics , Verrucomicrobia
5.
Nano Lett ; 20(3): 1928-1933, 2020 03 11.
Article in English | MEDLINE | ID: mdl-32073871

ABSTRACT

Immunogenic cell death (ICD) elicited by photodynamic therapy (PDT) is mediated through generation of reactive oxygen species (ROS) that induce endoplasmic reticulum (ER) stress. However, the half-life of ROS is very short and the intracellular diffusion depth is limited, which impairs ER localization and thus limits ER stress induction. To solve the problem, we synthesized reduction-sensitive Ds-sP NPs (PEG-s-s-1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000] nanoparticles) loaded with an efficient ER-targeting photosensitizer TCPP-TER (4,4',4″,4'″-(porphyrin-5,10,15,20-tetrayl)tetrakis(N-(2-((4-methylphenyl)sulfonamido)ethyl)benzamide). The resulting Ds-sP/TCPP-TER NPs could selectively accumulate in the ER and locally generate ROS under near-infrared (NIR) laser irradiation, which induced ER stress, amplified ICD, and activated immune cells, leading to augmented immunotherapy effect. This study presents a novel ICD amplifying, ER-targeting PDT strategy that can effectively eradicate primary tumors under NIR exposure, as well as distant tumors through an abscopal effect.


Subject(s)
Drug Delivery Systems , Endoplasmic Reticulum Stress , Endoplasmic Reticulum , Immunotherapy , Infrared Rays , Neoplasms, Experimental , Animals , Cell Death/drug effects , Cell Death/immunology , Cell Line, Tumor , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Endoplasmic Reticulum Stress/radiation effects , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Reactive Oxygen Species/immunology
6.
J Am Chem Soc ; 142(36): 15320-15330, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32820914

ABSTRACT

Current chemodynamic therapy (CDT) primarily relies on the delivery of transition metal ions with Fenton activity to trigger hydroxyl radical production from hydrogen peroxide. However, administration of an excess amount of exogenous Fenton-type heavy metals may cause potential adverse effects to human health, including acute and chronic damages. Here, we present a new CDT strategy that uses intracellular labile iron pool (LIP) as the endogenous source of Fenton-reactive metals for eliciting free radical generation, and the discovery of hydroperoxides (R'OOH) as an optimal LIP-mediated chemodynamic agent against cancer. By simulating the metabolic fates of peroxo compounds within cells, R'OOH was found to have excellent free radical-producing ability in the presence of labile iron(II) and to suffer only moderate elimination by glutathione/glutathione peroxidase, which contributes to its superior chemodynamic efficacy. The LIP-initiated nontoxic-to-toxic transition of R'OOH, together with increased LIP levels in tumor cells, enabled efficient and specific CDT of cancer. Moreover, pH/labile iron(II) cascade-responsive nanomedicines comprising encapsulated methyl linoleate hydroperoxide and LIP-increasing agent in pH-sensitive polymer particles were fabricated to realize enhanced CDT. This work not only paves the way to using endogenous Fenton-type metals for cancer therapy but also offers a paradigm for the exploration of high-performance chemodynamic agents activated by intracellular LIP.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/therapy , Ferrous Compounds/metabolism , Free Radicals/metabolism , Glioblastoma/therapy , Peroxides/pharmacology , Photothermal Therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Ferrous Compounds/chemistry , Free Radicals/chemistry , Glioblastoma/pathology , Humans , Optical Imaging , Peroxides/chemistry , Peroxides/metabolism
7.
Small ; 16(42): e2004016, 2020 10.
Article in English | MEDLINE | ID: mdl-32985099

ABSTRACT

Chemical transformation in cellular environment is critical for regulating biological processes and metabolic pathways. Harnessing biocatalytic cascades to produce chemicals of interest has become a research focus to benefit industrial and pharmaceutic areas. Nanoreactors, which can act as artificial cell-like devices to organize cascade reactions, have been recently proposed for potential therapeutic applications for life-threatening illnesses. Among various types of nanomaterials, there is a growing interest in 2D metal-organic frameworks (MOFs). By virtue of the ultralarge specific surface area, high porosity, and structural diversity, 2D MOF nanosheets hold great promise for a broad spectrum of biomedical use. Herein, a unique planar MOF-based hybrid architecture (GMOF-LA) is introduced by incorporating ultrasmall gold nanoparticles (Au NPs) as nanozyme and l-Arginine (l-Arg) as nitric oxide (NO) donor. The prepared Au NPs enable oxidation of glucose into hydrogen peroxide, which drives biocatalytic cascades to covert l-Arg into NO. Interestingly, the well-designed nanosheets not only possess excellent catalytical activity for NO generation, resulting in gas therapeutic effect, but also serve as a desired photosensitizer for photodynamic therapy. This study establishes a good example of exploring bioinspired nanoreactors for cooperative anticancer effect, which may pave the path for future "bench-to-bedside" design of nanomedicine.


Subject(s)
Metal Nanoparticles , Metal-Organic Frameworks , Neoplasms , Catalysis , Gold , Humans , Neoplasms/drug therapy
8.
J Am Chem Soc ; 141(25): 9937-9945, 2019 06 26.
Article in English | MEDLINE | ID: mdl-31199131

ABSTRACT

Chemodynamic therapy (CDT) employs Fenton catalysts to kill cancer cells by converting intracellular H2O2 into hydroxyl radical (•OH), but endogenous H2O2 is insufficient to achieve satisfactory anticancer efficacy. Despite tremendous efforts, engineering CDT agents with specific and efficient H2O2 self-supplying ability remains a great challenge. Here, we report the fabrication of copper peroxide (CP) nanodot, which is the first example of a Fenton-type metal peroxide nanomaterial, and its use as an activatable agent for enhanced CDT by self-supplying H2O2. The CP nanodots were prepared through coordination of H2O2 to Cu2+ with the aid of hydroxide ion, which could be reversed by acid treatment. After endocytosis into tumor cells, acidic environment of endo/lysosomes accelerated the dissociation of CP nanodots, allowing simultaneous release of Fenton catalytic Cu2+ and H2O2 accompanied by a Fenton-type reaction between them. The resulting •OH induced lysosomal membrane permeabilization through lipid peroxidation and thus caused cell death via a lysosome-associated pathway. In addition to pH-dependent •OH generation property, CP nanodots with small particle size showed high tumor accumulation after intravenous administration, which enabled effective tumor growth inhibition with minimal side effects in vivo. Our work not only provides the first paradigm for fabricating Fenton-type metal peroxide nanomaterials, but also presents a new strategy to improve CDT efficacy.


Subject(s)
Antineoplastic Agents/therapeutic use , Copper/chemistry , Hydrogen Peroxide/metabolism , Neoplasms/drug therapy , Quantum Dots/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Hydroxyl Radical/metabolism , Lysosomes/drug effects , Mice , Quantum Dots/chemistry , Xenograft Model Antitumor Assays
9.
Bioconjug Chem ; 30(6): 1745-1753, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31181890

ABSTRACT

As highly expressed in insulinomas, the glucagon-like peptide-1 receptor (GLP-1R) is believed to be an attractive target for diagnosis, localization, and treatment with radiolabeled exendin 4. However, the high and persistent radioactivity accumulation of exendin 4 in the kidneys limits accurate diagnosis and safe, as well as effective, radiotherapy in insulinomas. In this study, we intend to reduce the renal accumulation of radiolabeled exendin 4 through degradation mediated by brush border membrane enzymes. A new exendin 4 ligand NOTA-MVK-Cys40-Leu14-Exendin 4 containing Met-Val-Lys (MVK) linker between the peptide and 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) chelator was synthesized and labeled with 68Ga. The in vitro mouse serum stability and cell binding affinity of the tracer were evaluated. Initial in vitro cleavage of the linker was determined by incubation of a model compound Boc-MVK-Dde with brush border membrane vesicles (BBMVs) with and without the inhibitor of neutral endopeptidase (NEP). Further cleavage studies were performed with the full structure of NOTA-MVK-Cys40-Leu14-Exendin 4. Kidney and urine samples were collected in the in vivo metabolism study after intravenous injection of 68Ga-NOTA-MVK-Cys40-Leu14-Exendin 4. The microPET images were acquired in INS-1 tumor model at different time points; the radioactivity uptake of 68Ga-NOTA-MVK-Cys40-Leu14-Exendin 4 in tumor and kidneys were determined and compared with the control radiotracer without MVK linker. 68Ga-NOTA-MVK-Cys40-Leu14-Exendin 4 was stable in mouse serum. The MVK modification did not affect the affinity of NOTA-MVK-Cys40-Leu14-Exendin 4 toward GLP-1R. The in vitro cleavage study and in vivo metabolism study confirmed that the MVK sequence can be recognized by BBM enzymes and cleaved at the amide bond between Met and Val, thus releasing the small fragment containing Met. MicroPET images showed that the tumor uptake of 68Ga-NOTA-MVK-Cys40-Leu14-Exendin 4 was comparable to that of the control, while the kidney uptake was significantly reduced. As a result, more favorable tumor to kidney ratios were achieved. In this study, a novel exendin 4 analogue, NOTA-MVK-Cys40-Leu14-Exendin 4, was successfully synthesized and labeled with 68Ga. With the cleavable MVK sequence, this ligand could be cleaved by the enzymes on kidneys, and releasing the fragment of 68Ga-NOTA-Met-OH, which will rapidly excrete from urine. As the high and consistent renal radioactivity accumulation could be significantly reduced, NOTA-MVK-Cys40-Leu14-Exendin 4 shows great potential in the diagnosis and radiotherapy for insulinoma.


Subject(s)
Exenatide/pharmacokinetics , Gallium Radioisotopes/pharmacokinetics , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Insulinoma/diagnostic imaging , Animals , Exenatide/chemistry , Exenatide/therapeutic use , Female , Gallium Radioisotopes/chemistry , Gallium Radioisotopes/therapeutic use , Glucagon-Like Peptide-1 Receptor/analysis , HEK293 Cells , Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 1-Ring/therapeutic use , Humans , Insulinoma/radiotherapy , Mice , Positron-Emission Tomography , Theranostic Nanomedicine
10.
Angew Chem Int Ed Engl ; 58(41): 14758-14763, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31429173

ABSTRACT

Reactive oxygen species (ROS) can be used not only as a therapeutic agent for chemodynamic therapy (CDT), but also as a stimulus to activate release of antitumor drugs, achieving enhanced efficacy through the combination of CDT and chemotherapy. Here we report a pH/ROS dual-responsive nanomedicine consisting of ß-lapachone (Lap), a pH-responsive polymer, and a ROS-responsive polyprodrug. In the intracellular acidic environment, the nanomedicine can realize pH-triggered disassembly. The released Lap can efficiently generate hydrogen peroxide, which will be further converted into highly toxic hydroxyl radicals via the Fenton reaction. Subsequently, through ROS-induced cleavage of thioketal linker, doxorubicin is released from the polyprodrug. In vivo results indicate that the cascade of ROS generation and antitumor-drug release can effectively inhibit tumor growth. This design of nanomedicine with cascade reactions offers a promising strategy to enhance antitumor efficacy.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Doxorubicin/chemistry , Naphthoquinones/chemistry , Prodrugs , A549 Cells , Animals , Drug Liberation , Humans , Mice , Nanoparticles , Neoplasms, Experimental/drug therapy , Reactive Oxygen Species
11.
Ecotoxicol Environ Saf ; 166: 242-250, 2018 Dec 30.
Article in English | MEDLINE | ID: mdl-30273847

ABSTRACT

The aim of the current study was to determine the potential developmental and metabolic abnormalities caused by Cr (VI) exposure on Bufo gargarizans (B. gargarizans) embryos. B. gargarizans embryos were treated with different concentrations of Cr (VI) (13, 52, 104, 208, and 416 µg Cr6+ L-1) for 6 days. Morphological abnormalities, total length, weight and developmental stage were monitored. Malformations of embryos were also examined using scanning electron microscopy (SEM). In addition, the transcript levels of several genes associated with lipid metabolism, oxidative stress, and thyroid hormones signaling pathways were also determined. Our results showed a time-dependent inhibitory effect of Cr (VI) on the growth and development of B. gargarizans embryos. On day 4, total length, weight, and developmental stage were significantly lower at 416 µg Cr6+ L-1 relative to control embryos. On day 6, significant reductions in total length, weight, and developmental stage were observed at 104, 208, and 416 µg Cr6+ L-1. Malformed embryos were found in all Cr (VI) treatments, which were characterized by axial flexures, yolk sac edema and rupture, surface tissue hyperplasia, stunted growth, wavy fin and fin flexure. RT-qPCR results showed that exposure to Cr (VI) down-regulated TRß and Dio2 mRNA expression and up-regulated Dio3 mRNA level at 416 µg Cr6+ L-1. The transcript levels of SOD and GPx were upregulated at 52, 208, and 416 µg Cr6+ L-1, while the transcript level of HSP90 was downregulated at 52, 208, and 416 µg Cr6+ L-1. Also, mRNA expression of lipid synthesis-related genes (FAE and ACC) were significantly downregulated in embryos treated with 208 and 416 µg Cr6+ L-1, but mRNA expression of fatty acid ß-oxidation-related genes (ACOX, CPT, and SCP) was significantly upregulated at 416 µg Cr6+ L-1. Therefore, our results suggested that Cr (VI) could disrupt thyroid endocrine pathways and lipid synthesis, leading to the inhibition of growth and development in B. gargarizans embryos. Furthermore, the decreased ability of scavenging ROS induced by Cr (VI) might be responsible for the teratogenic effects of Cr (VI).


Subject(s)
Bufonidae/embryology , Chromium/toxicity , Lipid Metabolism/drug effects , Oxidative Stress/drug effects , Thyroid Gland/drug effects , Animals , Bufonidae/growth & development , Endocrine Disruptors/metabolism , Gene Expression Regulation, Developmental/drug effects , RNA, Messenger/metabolism , Superoxide Dismutase/metabolism , Thyroid Hormones/metabolism
12.
Ecotoxicol Environ Saf ; 126: 129-137, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26745004

ABSTRACT

The present study examined the adverse effects of fluoride exposure on embryos and larvae of Rana chensinensis. Survival, morphological abnormalities, growth and development, time to metamorphosis and size at metamorphic climax of R. chensinensis were examined. Our results showed that embryos malformation occurred in all fluoride treatments. Morphological abnormalities of embryos are characterized by axial flexures, the extrusion of fin axis, edema, and ruffled dorsal and ventral fin. Additionally, 4.1mg F(-)/L and above could significantly inhibit embryos growth and development. On day 15, total length and weight of tadpole were significantly lower in 19.6 and 42.4 mg F(-)/L treatments compared to control. However, significant reductions in total length and weight were observed only at 42.4 mg F(-)/L on day 30. Moreover, significant metamorphic delay and decrease in the size at metamorphic climax were found in larvae exposed to 42.4 mg F(-)/L. Taken together, embryos of R. chensinensis are more vulnerable to fluoride exposure than their tadpoles. Our results suggested that the presence of high concentrations fluoride might increase mortality risk and a reduction in juvenile recruitment in the field by increasing embryos malformation, delaying metamorphosis and decreasing size at metamorphosis.


Subject(s)
Fluorides/toxicity , Ranidae/physiology , Water Pollutants, Chemical/toxicity , Animals , Body Size/drug effects , Embryonic Development/drug effects , Larva/drug effects , Metamorphosis, Biological/drug effects , Ranidae/growth & development
13.
Ecotoxicology ; 25(8): 1600-1608, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27599820

ABSTRACT

Triclosan (TCS) is commonly used worldwide in a range of personal care and sanitizing products. The aim of this study was to evaluate potential effects of TCS exposure on embryonic development of Bufo gargarizans, an endemic frog species in China. Standard Gosner stage 3 B. gargarizans embryos were exposed to 10 ~ 150 µg/L TCS during embryogenesis. Survival, total length, weight, developmental stage, duration of different embryo stages, malformation, and type II and III deiodinase (D2 and D3) expression were measured. Inhibitory effects on embryo developmental stage, total length and weight were found at 30 ~ 150 µg/L TCS. Moreover, the duration of embryonic development was increased at gastrula, neural, circulation, and operculum development stage in TCS-treated groups. In addition, TCS exposure induced morphological malformations in B. gargarizans embryos, which are characterized by hyperplasia, abdominal edema, and axial flexures. Furthermore, our results showed that the expression of D2 in embryos was probably down-regulated at 60 and 150 µg/L TCS, but its spatial expression patterns was not affected by TCS. In summary, our study suggested that TCS exposure not only resulted in delayed growth and development but also caused teratogenic effects in B. gargarizans embryos, and the developmental effects of TCS at high concentrations may be associated with disruption of THs homeostasis. Although further studies are necessary, the present findings could provide a basis for understanding on harmful effects and the potential mechanisms of TCS in amphibian embryos.


Subject(s)
Anti-Infective Agents, Local/toxicity , Bufonidae/physiology , Embryonic Development/drug effects , Toxicity Tests, Chronic , Triclosan/toxicity , Animals , China , Iodide Peroxidase/metabolism , Metamorphosis, Biological , Water Pollutants, Chemical/toxicity
14.
Biomacromolecules ; 15(11): 4281-92, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25325531

ABSTRACT

The application of PEG-b-PCL micelles was dampened by their inherent low drug-loading capability and relatively poor cell uptake efficiency. In this study, a series of novel PEG-b-PCL copolymers methoxy poly(ethylene glycol)-b-poly(ε-caprolactone-co-γ-dimethyl maleamidic acid -ε-caprolactone) (mPEG-b-P(CL-co-DCL)) bearing different amounts of acid-labile ß-carboxylic amides on the polyester moiety were synthesized. The chain structure and chemical composition of copolymers were characterized by (1)H NMR, Fourier transform infrared spectroscopy (FT-IR), and gel permeation chromatography (GPC). mPEG-b-P(CL-co-DCL) with critical micellar concentrations (CMCs) of 3.2-6.3 µg/mL could self-assemble into stable micelles in water with diameters of 100 to 150 nm. Doxorubicin (DOX), a cationic hydrophobic drug, was successfully encapsulated into the polymer micelles, achieving a very high loading content due to electrostatic interaction. Then the stability, charge-conversional behavior, loading and release profiles, cellular uptake and in vitro cytotoxicity of free drug and drug-loaded micelles were evaluated. The ß-carboxylic amides functionalized polymer micelles are negatively charged and stable in neutral solution but quickly become positively charged at pH 6.0, due to the hydrolysis of ß-carboxylic amides in acidic conditions. The pH-triggered negative-to-positive charge reversal not only resulted in a very fast drug release in acidic conditions, but also effectively enhanced the cellular uptake by electrostatic absorptive endocytosis. The MTT assay demonstrated that mPEG-b-P(CL-co-DCL) micelles were biocompatible to HepG2 cells while DOX-loaded micelles showed significant cytotoxicity. In sum, the introduction of acid-labile ß-carboxylic amides on the polyester block in mPEG-b-P(CL-co-DCL) exhibited great potentials for the modifications in the stability in blood circulation, drug solubilization, and release properties, as well as cell internalization and intracellular drug release.


Subject(s)
Doxorubicin/chemistry , Drug Delivery Systems/methods , Intracellular Fluid/drug effects , Lactones/chemistry , Micelles , Polyethylene Glycols/chemistry , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Doxorubicin/administration & dosage , Doxorubicin/metabolism , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Intracellular Fluid/metabolism , Lactones/administration & dosage , Lactones/metabolism , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/metabolism , Surface Properties/drug effects
15.
Adv Mater ; 36(13): e2307822, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37929780

ABSTRACT

Messenger RNA (mRNA)-based therapy has emerged as a powerful, safe, and rapidly scalable therapeutic approach that involves technologies for both mRNA itself and the delivery vehicle. Although there are some unique challenges for different applications of mRNA therapy, a common challenge for all mRNA therapeutics is the transport of mRNA into the target cell cytoplasm for sufficient protein expression. This review is focused on the behaviors at the cellular level of nanotechnology-mediated mRNA delivery systems, which have not been comprehensively reviewed yet. First, the four main therapeutic applications of mRNA are introduced, including immunotherapy, protein replacement therapy, genome editing, and cellular reprogramming. Second, common types of mRNA cargos and mRNA delivery systems are summarized. Third, strategies to enhance mRNA delivery efficiency during the cellular trafficking process are highlighted, including accumulation to the cell, internalization into the cell, endosomal escape, release of mRNA from the nanocarrier, and translation of mRNA into protein. Finally, the challenges and opportunities for the development of nanotechnology-mediated mRNA delivery systems are presented. This review can provide new insights into the future fabrication of mRNA nanocarriers with desirable cellular trafficking performance.


Subject(s)
Nanoparticles , RNA, Messenger/metabolism , Nanoparticles/metabolism , Nanotechnology , Endosomes/metabolism , Proteins , Drug Delivery Systems
16.
J Control Release ; 367: 697-707, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38331001

ABSTRACT

Precise targeting is a major prerequisite for effective cancer therapy because it ensures a sufficient therapeutic dosage in tumors while minimizing off-target side effects. Herein, we report a live-macrophage-based therapeutic system for high-efficiency tumor therapy. As a proof of concept, anti-human epidermal growth factor receptor-2 (HER2) affibodies were genetically engineered onto the extracellular membrane of macrophages (AE-Mφ), which further internalized doxorubicin (DOX)-loaded poly(lactic-co-glycolic acid) nanoparticles (NPs) to produce a macrophage-based therapeutic system armed with anti-HER2 affibodies. NPs(DOX)@AE-Mφ were able to target HER2+ cancer cells and specifically elicit affibody-mediated cell therapy. Most importantly, the superior HER2 + -targeting capability of NPs(DOX)@AE-Mφ greatly guaranteed high accumulation at the tumor site for improved chemotherapy, which acted synergistically with cell therapy to significantly enhance anti-tumor efficacy. This study suggests that NPs(DOX)@AE-Mφ could be utilized as an innovative 'living targeted drug' platform for combining both macrophage-mediated cell therapy and targeted chemotherapy for the individualized treatment of solid tumors.


Subject(s)
Nanoparticles , Neoplasms , Humans , Drug Carriers , Neoplasms/drug therapy , Drug Delivery Systems , Doxorubicin/therapeutic use , Macrophages , Cell Line, Tumor
17.
ACS Nano ; 17(20): 19550-19580, 2023 10 24.
Article in English | MEDLINE | ID: mdl-37819640

ABSTRACT

In late 2020, the U.S. Food and Drug Administration (FDA) approved a lipid-based mRNA vaccine for the prevention of COVID-19, which has pushed this field to be more closely studied and motivated researchers to delve deeper into mRNA therapeutics. To date, the research on mRNA cancer vaccines has been developed rapidly, and substantial hopeful therapeutic results have been achieved against various solid tumors in clinical trials. In this review, we first introduce three main components of mRNA cancer vaccines, including mRNA antigens, adjuvants, and delivery vectors. Engineering these components can optimize the therapeutic effects of mRNA cancer vaccines. For instance, appropriate modification of mRNA structure can alleviate the poor stability and innate immunogenicity of mRNA, and the use of mRNA delivery vectors can address the issues of low delivery efficiency in vivo. Second, we emphatically discuss some strategies to further improve the efficacy of mRNA cancer vaccines, namely modulating the immunosuppressive tumor environment, optimizing administration routes, achieving targeting delivery to intended tissues or organs, and employing combination therapy. These strategies can strengthen the tumor inhibitory ability of mRNA cancer vaccines and increase the possibility of tumor elimination. Finally, we point out some challenges in the clinical practice of mRNA cancer vaccines and offer our perspectives on future developments in this rapidly evolving field. It is anticipated that mRNA cancer vaccines will be rapidly developed for clinical cancer therapy in the near future.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Neoplasms/drug therapy , Antigens, Neoplasm , Immunotherapy/methods , RNA, Messenger/genetics
18.
Sci Total Environ ; 863: 160849, 2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36521604

ABSTRACT

Lead (Pb) and copper (Cu) are ubiquitous metal contaminants and can pose a threat to ecosystem and human health. Bile acids have recently received considerable attention for their role in the maintenance of health. However, there were few studies on whether Pb and Cu affect bile acid metabolism in amphibians. In this study, a combination approach of histological analysis, targeted metabolomics, 16S rDNA sequencing and qPCR was used to explore the impacts of Pb, Cu and their mixture (Mix) on bile acid in Bufo gargarizans tadpoles. The results showed that Pb, Cu, and Mix resulted in intestinal damage and altered the bile acid profiles. Specifically, Pb and Mix exposure decreased total bile acid concentrations while increased toxic bile acid levels; in contrast, Cu exposure increased total bile acid levels. And hydrophilic bile acids were reduced in all treated tadpoles. Moreover, Pb and/or Cu changed the composition of intestinal microbiota, especially Clostridia, Bacteroides and Eubacterium involved in bile acid biotransformation. qPCR revealed that the decreased total bile acid concentrations in Pb- and Mix-treated tadpoles were most likely attributed to the activation of intestinal farnesoid X receptor (Fxr), which suppressed bile acid synthesis and reabsorption. While activated fxr in the Cu treatment group may be a regulatory mechanism in response to increased bile excretion, which is a detoxification route of tadpoles under Cu stress. Collectively, Pb, Cu and Mix changed bile acid profiles by affecting intestinal microbial composition and activating Fxr signaling. This study provided insight into the impacts of Pb and Cu on bile acid metabolism and contributed to the assessment of the potential ecotoxicity of heavy metals on amphibians.


Subject(s)
Copper , Gastrointestinal Microbiome , Humans , Animals , Copper/toxicity , Lead/toxicity , Ecosystem , Bufonidae , Larva , Bile Acids and Salts
19.
Sci Total Environ ; 855: 159031, 2023 Jan 10.
Article in English | MEDLINE | ID: mdl-36170915

ABSTRACT

Bile acids, as metabolic regulators and signaling molecules, play key roles in the regulation of host metabolism and immune responses. Heavy metals such as lead (Pb) and copper (Cu) are widespread environmental pollutants that threaten public health. However, the effects of heavy metals on bile acid metabolism and the underlying molecular mechanisms remain unclear, particularly for ecologically important amphibian species. In the present research, the effects of exposure to environmentally-relevant concentrations of Pb (250 µg/L), Cu (50 µg/L), and a mixture of both (Mix) on bile acid metabolism and the underlying molecular mechanisms in the intestines of Bufo gargarizans larvae were comprehensively investigated using histopathology, metabolomics and transcriptomics analysis. Our results suggested that Pb and/or Cu caused histopathological damage to the intestine and liver, such as decreased intestinal epithelial cell height and dilated hepatic sinusoid. The total bile acid level was decreased in the Pb and Mix exposure groups but elevated in the Cu treatment. A significant decrease in the ratio of conjugated to unconjugated bile acids was present in all treatment groups. Also, the level of GCA was increased while TCA and TCDCA were decreased in all exposure groups. In addition, exposure to Pb and Cu altered the expression levels of genes related to intestinal absorption. For example, mrp2, mrp3 and aqp4 had higher expression in the Pb and Mix treatment groups, and aqp1 and mrp4 were increased in the Cu treatment group. Overall, we speculated that the dysregulation of bile acid homeostasis induced by Pb and Cu exposure may be due to impaired intestinal absorption. These findings raise further concerns about the hazards of Pb and/or Cu in influencing bile acid metabolism that might lead to the development of metabolic diseases and inflammatory disorders.


Subject(s)
Copper , Metals, Heavy , Animals , Larva , Copper/toxicity , Bile Acids and Salts , Lead/toxicity , Transcriptome , Bufonidae , Homeostasis , Metabolomics , Intestinal Absorption
20.
J Control Release ; 360: 564-577, 2023 08.
Article in English | MEDLINE | ID: mdl-37442200

ABSTRACT

Phototherapies, mainly including photodynamic and photothermal therapy, have made considerable strides in the field of cancer treatment. With the aid of phototherapeutic agents, reactive oxygen species (ROS) or heat are generated under light irradiation to selectively damage cancer cells. However, sole-modality phototherapy faces certain drawbacks, such as limited penetration of phototherapeutic agents into tumor tissues, inefficient ROS generation due to hypoxia, treatment-induced inflammation and resistance of tumor to treatment (e.g., high levels of antioxidants, expression of heat shock protein). Gas therapy, an emerging therapy approach that damages cancer cells by improving the level of certain gas at the tumor site, shows potential to overcome the challenges associated with phototherapies. In addition, with the rapid development of nanotechnology, gas-assisted phototherapy based on nanomedicines has emerged as a promising strategy to enhance the treatment efficacy. This review summarizes recent advances in gas-assisted phototherapy and discusses the prospects and challenges of this strategy in cancer phototherapy.


Subject(s)
Neoplasms , Photochemotherapy , Humans , Reactive Oxygen Species/metabolism , Phototherapy , Neoplasms/therapy , Neoplasms/pathology , Nanotechnology , Photosensitizing Agents
SELECTION OF CITATIONS
SEARCH DETAIL