Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
ACS Sens ; 9(6): 3150-3157, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38717584

ABSTRACT

Tracking trace protein analytes in precision diagnostics is an ongoing challenge. Here, we developed an ultrasensitive detection method for the detection of SARS-CoV-2 nucleocapsid (N) protein by combining enzyme-linked immunosorbent assay (ELISA) with the clustered regularly interspaced short palindromic repeat/CRISPR-associated protein (CRISPR/Cas) system. First, the SARS-CoV-2 N protein bound by the capture antibody adsorbed on the well plate was sequentially coupled with the primary antibody, biotinylated secondary antibody, and streptavidin (SA), followed by biotin primer binding to SA. Subsequently, rolling circle amplification was initiated to generate ssDNA strands, which were targeted by CRISPR/Cas12a to cleave the FAM-ssDNA-BHQ1 probe in trans to generate fluorescence signals. We observed a linear relationship between fluorescence intensity and the logarithm of N protein concentration ranging from 3 fg/mL to 3 × 107 fg/mL. The limit of detection (LOD) was 1 fg/mL, with approximately nine molecules in 1 µL of the sample. This detection sensitivity was 4 orders magnitude higher than that of commercially available ELISA kits (LOD: 5.7 × 104 fg/mL). This method was highly specific and sensitive and could accurately detect SARS-CoV-2 pseudovirus and clinical samples, providing a new approach for ultrasensitive immunoassay of protein biomarkers.


Subject(s)
Coronavirus Nucleocapsid Proteins , Limit of Detection , SARS-CoV-2 , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Humans , Coronavirus Nucleocapsid Proteins/immunology , Coronavirus Nucleocapsid Proteins/analysis , Enzyme-Linked Immunosorbent Assay/methods , Immunoassay/methods , COVID-19/diagnosis , COVID-19/virology , CRISPR-Cas Systems/genetics , Phosphoproteins/immunology , Phosphoproteins/chemistry , CRISPR-Associated Proteins/chemistry , Endodeoxyribonucleases/chemistry , Nucleocapsid Proteins/immunology , Bacterial Proteins
2.
Opt Lett ; 49(5): 1193-1196, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38426971

ABSTRACT

We present a light sheet fluorescence microscopy (LSFM) with active optical manipulation by using linear optical tweezers (LOTs). In this method, two coaxially transmitting laser beams of different wavelengths are shaped using cylindrical lenses to form a linear optical trapping perpendicular to the optical axis and an excitation light sheet (LS) parallel to the optical axis, respectively. Multiple large-sized polystyrene fluorescent microspheres are stably captured by LOTs, and their rotation angles around specific rotation axes are precisely controlled. During a sample rotation, the stationary excitation LS scans the sample to obtain fluorescence sectioning images of the sample at different angles.

3.
Biomed Opt Express ; 15(3): 1585-1594, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38495715

ABSTRACT

We present a dynamic speckle illumination wide-field fluorescence microscopy (DSIWFM) combined with a line optical tweezers (LOTs) for rotational fluorescence sectioning imaging. In this method, large polystyrene fluorescent microspheres are stably trapped with LOTs, and precisely manipulated to rotate around a specific rotation axis. During the rotation process, multiple raw fluorescence images of trapped microspheres are obtained with dynamic speckle illumination. The root-mean-square (RMS) algorithm is used to extract the drastically changing fluorescent signals in the focal plane to obtain the fluorescence sectioning images of the samples at various angles. The influence of speckle granularity on the image quality of fluorescence sectioning images is experimentally analyzed. The rotational fluorescence sectioning images obtained by DSIWFM with LOTs could provide an alternative technique for applications of biomedical imaging.

4.
J Mater Chem B ; 11(17): 3951-3957, 2023 05 03.
Article in English | MEDLINE | ID: mdl-37067569

ABSTRACT

Mitochondria play a vital role in maintaining cellular homeostasis. In recent years, studies have found that mitochondria have an important role in the occurrence and development of tumors, and targeting mitochondria has become a new strategy for tumor treatment. Lonidamine (LND), as a hexokinase inhibitor, can block the energy supply and destroy mitochondria. However, poor water solubility and low mitochondrial selectivity limit its clinical application. To overcome these obstacles, we report redox-activated self-assembled carrier-free nanoparticles (Cy-TK-LND NPs) based on a small molecule prodrug, in which photosensitizer IR780 (Cy) which targets mitochondria is conjugated to LND via a sensitive thioketal (TK) linker. Intracellular oxidative stress induced by laser radiation leads to the responsive cleavage of Cy-TK-LND NPs, facilitating the release of free LND into mitochondria. Subsequently, LND damages mitochondria, triggering the apoptosis pathway. The results show the effective killing effect of Cy-TK-LND NPs on cancer cells in vitro and in vivo. The IC50 value of irradiated Cy-TK-LND NPs is 5-fold lower than that of free LND. Moreover, tumor tissue section staining results demonstrate that irradiated Cy-TK-LND NPs induce necrosis and apoptosis of tumor cells, upregulate cytochrome C and pro-apoptotic Bax, and downregulate anti-apoptotic Bcl-2. Generally, Cy-TK-LND NPs exhibit efficient mitochondria-targeted delivery to improve the medicinal availability of LND. Accordingly, such a carrier-free prodrug-based nanomedicine holds promise as an effective cancer chemotherapy strategy.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Prodrugs , Humans , Antineoplastic Agents/therapeutic use , Prodrugs/metabolism , Drug Liberation , Mitochondria/metabolism , Neoplasms/pathology , Nanoparticles/therapeutic use , Oxidation-Reduction
5.
ACS Nano ; 17(7): 6410-6422, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36988613

ABSTRACT

Micro-/nanorobots have attracted great interest in the field of drug delivery and treatment, while preparations for biocompatible robots are extremely challenging. Here, a self-driving yeast micro-/nanorobot (Cur@CaY-robot) is designed via dual biomineralization and acid catalysis of calcium carbonate (CaCO3). Inner nano-CaCO3 inside yeast cells (CaY) is biomineralized through cell respiration and provides nanoscaffolds for highly encapsulating curcumin (Cur). Meanwhile, the CaCO3 crystals outside yeast cells (outer-CaCO3) through uniaxial growth offer an asymmetric power source for self-propelled motility. The Cur@CaY-robot displays an efficient motion in gastric acid, with the potential for deep penetration to the thick gastric mucus, which significantly improves the accumulation of drug agents in the stomach wall tissue for robust gastritis therapy. More importantly, Ca2+ cations released from the Cur@CaY-robot also synergistically repair the gastric motility of gastritis mice. Such yeast micro-/nanorobots exhibit desirable biocompatibility and biodegradability with a good loading capacity for drugs. This work provides an idea for the design of micro-/nanorobots through an environmentally friendly biosynthesis strategy for active drug delivery and precise therapy.


Subject(s)
Curcumin , Gastritis , Nanoparticles , Mice , Animals , Saccharomyces cerevisiae , Drug Delivery Systems , Curcumin/chemistry , Gastritis/drug therapy , Nanoparticles/chemistry
6.
ACS Nano ; 17(1): 421-436, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36573683

ABSTRACT

Glioblastoma (GBM) is the most devastating brain tumor and highly resistant to conventional chemotherapy. Herein, we introduce biomimetic nanosonosensitizer systems (MDNPs) combined with noninvasive ultrasound (US) actuation for orthotopic GBM-targeted delivery and sonodynamic-enhanced chemotherapy. MDNPs were fabricated with biodegradable and pH-sensitive polyglutamic acid (PGA) and the chemotherapeutic agent and sonosensitizer doxorubicin (DOX), camouflaged with human GBM U87 cell membranes. MDNPs presented homologous targeting accumulation and in vivo long-term circulation ability. They effectively passed through the blood-brain barrier (BBB) under US assistance and reached the orthotopic GBM site. MDNPs exhibited controllable US-elicited sonodynamic effect by generation of reactive oxygen species (ROS). ROS not only induced cancer cell apoptosis but also downregulated drug-resistance-related factors to disrupt chemoresistance and increase sensitivity to chemotherapy. The in vivo study of orthotopic GBM treatments further proved that MDNPs exhibited US-augmented synergistic antitumor efficacy and strongly prolonged the survival rate of mice. The use of low-dose DOX and the safety of US enabled repeated treatment (4 times) without obvious cardiotoxicity. This effective and safe US-enhanced chemotherapy strategy with the advantages of noninvasive brain delivery and high drug sensitivity holds great promise for deep-seated and drug-resistant tumors.


Subject(s)
Glioblastoma , Nanoparticles , Humans , Mice , Animals , Glioblastoma/drug therapy , Glioblastoma/metabolism , Reactive Oxygen Species/metabolism , Biomimetics , Ultrasonography , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Resistance , Cell Line, Tumor
7.
Acta Biomater ; 142: 264-273, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35101580

ABSTRACT

Malignant melanoma remains the life-threatening form of skin cancer with high mortality and poor prognosis. Thus, an ideal melanoma therapeutic strategy is of immediate importance which can remove the primary tumor, as well as inhibit the metastasis and recurrence. Here, we report the fabrication of adjuvant monophosphoryl lipid A (MPLA) lipid bilayer-enveloped and photosensitizer indocyanine green (ICG)-loaded gold nanocages (MLI-AuNCs) for immunogenic phototherapy of aggressive melanoma. Hollow porous AuNCs are used as carriers to deliver MPLA and ICG, and protect ICG from photodegradation. Both AuNCs and ICG absorb near infrared (NIR) light and can be applied in controllable NIR-triggered photothermal and photodynamic combination therapy (PTT/PDT) of melanoma. MLI-AuNCs coated by thermosensitive lipid bilayer exhibit uniform size, good biocompatibility and bioavailability with prominent tumor accumulation, which further improve the PTT/PDT efficacy. MLI-AuNCs under NIR irradiation not only destroy the primary tumor by PTT/PDT, but also elicit robust antitumor immune response with melanoma associated antigens and MPLA released in situ. The released antigens and MPLA subsequently enhance the recruitment and maturation of dendritic cells, which further activate the effector T cells to inhibit metastases and recurrence of melanoma. This immunomodulatory-boosted PTT/PDT nanoplatform provides a new opportunity for highly aggressive melanoma treatment. STATEMENT OF SIGNIFICANCE: An ideal tumor therapeutic strategy not only can remove the primary tumor, but also inhibit metastasis and recurrence. Here, we introduced a versatile nanoplatform MLI-AuNCs for immunogenic phototherapy of aggressive melanoma. Adjuvant MPLA and photosensitizer ICG can be protected and co-delivered to the tumors by thermosensitive lipid-enveloped AuNCs. MLI-AuNCs exhibited prominent tumor accumulation ability and produced the potent PTT/PDT effect to destroy the primary tumors with a single dose of NIR irradiation, as well as elicited the strong antitumor immunity to inhibit the metastasis and relapse. This study may provide a potential therapeutic vaccination strategy against advanced melanoma and other difficult-to-treat cancers.


Subject(s)
Melanoma , Nanoparticles , Photochemotherapy , Cell Line, Tumor , Gold/pharmacology , Humans , Indocyanine Green/pharmacology , Lipid Bilayers , Melanoma/therapy , Photosensitizing Agents/pharmacology , Phototherapy
8.
Biomaterials ; 269: 120670, 2021 02.
Article in English | MEDLINE | ID: mdl-33485214

ABSTRACT

Immunotherapy is one of the most promising approaches to inhibit tumor growth and metastasis by activating host immune functions. However, the arising problems such as low immune response caused by complex tumor microenvironment and extremely systemic immune storm still limit the clinical applications of immunotherapy. Here, we construct Poly I: C-encapsulated poly (lactic-co-glycolic acid) nanoparticles (PLP NPs) with a slow release profile. A biomimetic system (MPLP), which loads PLP NPs on the surface of bone marrow-derived macrophage (BMDM) via the maleimide-thiol conjugation, is synthesized to effectively deliver PLP, control drug release and activate the tumor-specific immune response in situ. The results show that PLP NPs loading does not affect the activity and function of BMDM. Then, BMDM acts as a living cell drug vehicle and promotes the accumulation of PLP NPs in tumors, where Poly I: C is released from PLP NPs and reprograms BMDM into tumoricidal M1 macrophage. Furthermore, MPLP triggers potent antitumor immune responses in vivo and effectively inhibits local and metastatic tumors without causing adverse pathological immune reactions. This study offers an inspiration to facilitate clinical translation through the delivery of drugs by living immune cells for future anticancer therapy.


Subject(s)
Nanoparticles , Pharmaceutical Preparations , Cell Line, Tumor , Immunotherapy , Macrophages , Poly I-C , Polylactic Acid-Polyglycolic Acid Copolymer
9.
Biomaterials ; 269: 120639, 2021 02.
Article in English | MEDLINE | ID: mdl-33434714

ABSTRACT

Sonodynamic therapy (SDT) is a promising approach for tumor treatment because of the noninvasion, and future would be perfect while it activates systemic immune responses through deep penetration to effectively avoid tumor recurrence. Here, a multifunctional nanosonosensitizer system (FA-MnPs) is designed by encapsulating manganese-protoporphyrin (MnP) into folate-liposomes. The nanoparticles of FA-MnPs not only exhibit excellent depth-responsive SDT but also simultaneously activate SDT-mediated immune response. Under US irradiation, FA-MnPs show the high acoustic intensity in mimic tissue up to 8 cm depth and generate amount of singlet oxygen (1O2). Density functional theory (DFT) calculations reveal that metal coordination in MnP has enhanced the US response ability. The good depth-responsed SDT of FA-MnPs efficiently suppresses the growth of not only the superficial tumors but also the deep lesion in the triple-negative breast cancer (TNBC) mice model. Importantly, FA-MnPs-induced SDT further re-polarizes immunosuppressive M2 macrophages to antitumor M1 macrophages, and elicits immunogenic cell death (ICD) to activate dendritic cells, T lymphocytes, and natural killercells (NK), which consequently trigger the antitumor immune, contributing to the tumor growth inhibition. This study put forward an idea for curing deep-seated and metastatic tumors through noninvasively depth-irradiated immunogenic SDT by reasonably designing multifunctional sonosensitizers.


Subject(s)
Triple Negative Breast Neoplasms , Ultrasonic Therapy , Animals , Cell Line, Tumor , Humans , Liposomes , Mice , Neoplasm Recurrence, Local , Protoporphyrins , Triple Negative Breast Neoplasms/drug therapy
10.
Bioact Mater ; 6(4): 951-962, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33102938

ABSTRACT

The adoptive transfer of chimeric antigen receptor-T (CAR-T) cells has shown remarkable clinical responses in hematologic malignancies. However, unsatisfactory curative results and side effects for tumor treatment are still unsolved problems. Herein we develop a click CAR-T cell engineering strategy via cell glycometabolic labeling for robustly boosting their antitumor effects and safety in vivo. Briefly, paired chemical groups (N3/BCN) are separately incorporated into CAR-T cell and tumor via nondestructive intrinsic glycometabolism of exogenous Ac4GalNAz and Ac4ManNBCN, serving as an artificial ligand-receptor. Functional groups anchored on cell surface strengthen the interaction of CAR-T cell and tumor via bioorthogonal click chemistry, further enhancing specific recognition, migration and selective antitumor effects of CAR-T cells. In vivo, click CAR-T cell completely removes lymphoma cells and minimizes off-target toxicity via selective and efficient bioorthogonal targeting in blood cancer. Surprisingly, compared to unlabeled cells, artificial bioorthogonal targeting significantly promotes the accumulation, deep penetration and homing of CAR-T cells into tumor tissues, ultimately improving its curative effect for solid tumor. Click CAR-T cell engineering robustly boosts selective recognition and antitumor capabilities of CAR T cells in vitro and in vivo, thereby holding a great potential for effective clinical cell immunotherapy with avoiding adverse events in patients.

11.
Kaohsiung J Med Sci ; 37(2): 136-144, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33128488

ABSTRACT

Skimmin, a natural coumarin derivate, has been showed to be protective against experimental diabetic nephropathy; however, its protective effect on diabetic cardiomyopathy (DCM) is not clarified. By using in vitro and in vivo models, we investigated skimmin's protective effect on impaired heart tissues in DCM. DCM was induced by streptozotocin (STZ, 60 mg/kg) using Sprague Dawley rats, and diabetic rats were treated with either skimmin (15 or 30 mg/kg) or the vehicle for 16 weeks, and normal rats were used as a control. Hematoxylin and eosin and Masson's trichrome staining were performed to evaluate the cardiac histopathology, and the oxidative stress and proinflammation cytokines in heart tissues were measured. The protein levels of key mediators in fibrosis, pyroptosis, and autophagy in heart tissues were investigated using western blotting. In vitro, primary neonatal cardiomyocytes were treated with skimmin (2 and 10 µM) under stimulation by high glucose (30 mM) and low glucose (5 mM) respectively, and the molecular mechanisms on pyroptosis and autophagy were studied. Compared to the vehicle-treated DCM group, skimmin treatment significantly improved the ejection fraction and fractional shortening of the left ventricle and reduced the oxidative stress by increasing the glutathione level and activity of superoxide dismutase and catalase. Skimmin also reduced cardiac fibrosis, and decreased proinflammation cytokines in cardiac tissues. Mechanism studies showed skimmin may enhance the autophagy and ameliorate NLRP3 inflammasome activation to play a protective role in DCM. This study, for the first time, indicates that skimmin might be a promising lead compound for DCM.


Subject(s)
Cardiotonic Agents/therapeutic use , Coumarins/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/prevention & control , Animals , Autophagy/drug effects , Body Weight/drug effects , Cardiotonic Agents/pharmacology , Caspase 1/metabolism , Chronic Disease , Coumarins/pharmacology , Cytokines/metabolism , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/physiopathology , Glucose/toxicity , Heart/drug effects , Heart/physiopathology , Heart Rate/drug effects , Inflammation/complications , Inflammation/pathology , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/drug effects , Pyroptosis/drug effects , Rats, Sprague-Dawley , Streptozocin
12.
J Control Release ; 323: 387-397, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32330573

ABSTRACT

Chemotherapy is an important modality available for cancer treatment. However, the present chemotherapy is still far from being satisfactory mainly owing to the severe side effects of the chemotherapeutic agents and drug resistance of cancer cells. Thus, reversing drug resistance by constructing an ideal chemotherapeutic strategy with the least side effects and the best efficacy is greatly needed. Here, we designed a smart nanosystem of thermo-sensitive liposome coated gold nanocages with doxorubicin (DOX) loading (LAD) for near-infrared (NIR)-triggered drug release and chemo-photothermal combination therapy. The biocompatible liposomes coating facilitated the cellular uptake of LAD and meanwhile avoided drug leakage during the circulation. More importantly, LAD exhibited controllable photothermal conversion property and produced mild heat under NIR irradiation, which not only triggered DOX release and transferred DOX from lysosome to nucleus, but also elicited the mild heat cell killing effect to improve the curative efficiency. Further mechanism study revealed that mild heat could reverse drug resistance by down-regulation of the chemoresistance-related markers (e.g., HSF-1, p53, P-gp), and inhibited DOX export and increased drug sensitiveness, thereby prominently increased the anticancer efficiency. This versatile nanoplatform with enhanced curative efficacy and lower side effect is promising to apply in the field of drug controlled release and combination tumor therapy.


Subject(s)
Gold , Hyperthermia, Induced , Cell Line, Tumor , Doxorubicin , Drug Delivery Systems , Drug Liberation , Drug Resistance, Neoplasm , Hot Temperature , Phototherapy
13.
ACS Appl Mater Interfaces ; 12(11): 12573-12583, 2020 Mar 18.
Article in English | MEDLINE | ID: mdl-32119518

ABSTRACT

Reactive oxygen species (ROS)-mediated cell apoptosis has been a significant strategy for tumor oxidative damage, while tumor hypoxia is a major bottleneck for efficiency. Here, a novel TiO-porphyrin nanosystem (FA-TiOPs) is designed by encapsulating TiO-porphyrin (TiOP) in folate-liposome. The nanosysytem can photocatalyze H2O and tumor-overexpressed H2O2, in situ generating sufficient ROS. TiOP can photosplit water to produce ·OH radical, H2O2, and O2. Generated O2 not only conquers the hypoxia of tumor environment but also can be further excited by TiOP to 1O2 for killing tumor cells. Density functional theory calculations indicate that high energy in excited state (S1) of TiOP and narrow gap energy between S1 and the triplet excited state (Tn) might contribute to the efficient photocatalytic action. Moreover, the generated and overexpressed H2O2 in tumors can also be photocatalyzed to generate 1O2 especially in acid condition, helpful to specific anticancer effect while harmless to normal tissues. This research might pave a new way to bypass the hypoxia-triggered problem for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Nanoparticles/chemistry , Oxidative Stress , Tumor Hypoxia , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Photolysis , Porphyrins/chemistry , Porphyrins/pharmacology , Reactive Oxygen Species/metabolism , Titanium/chemistry , Titanium/pharmacology , Tumor Hypoxia/drug effects , Tumor Hypoxia/radiation effects
14.
Acta Biomater ; 104: 188-197, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31945508

ABSTRACT

Advanced colorectal cancer has a high mortality rate since conventional treatments have limited therapeutic effects and poor prognosis with high risks of metastasis and recurrence. Photodynamic therapy (PDT) is a promising treatment modality for the eradication of colorectal cancer, but its curative efficacy is severely affected by tumor hypoxia. Herein, we developed a core-shell gold nanocage coated with manganese dioxide and hyaluronic acid (AMH) for targeted delivery to colorectal tumors and oxygenation-boosted immunogenic phototherapy in situ. The AMH nanoparticles can generate abundant oxygen from mild acidic/H2O2 medium, which can further enhance the PDT efficacy of AMH itself under near infrared (NIR) irradiation. Meanwhile, AMH-based PDT induced immunogenic cell death (ICD) of tumor cells with damage-associated molecular patterns (DAMPs) release and facilitated the dendritic cells (DCs) maturation to further potentiate the systematic antitumor immunity against advanced tumors. In vivo experiment results exhibited that AMH nanoparticles not only had the ability of targeting tumor but also in situ produced sufficient oxygen to relieve the tumor hypoxia. Furthermore, AMH-mediated oxygen-boosted immunogenic PDT effectively inhibited the tumor growth and recurrence. Thus, this work provides a potent targeted delivery nanoplatform for enhanced immunogenic PDT against advanced cancers. STATEMENT OF SIGNIFICANCE: Local hypoxic tumor microenvironment not only greatly limits the photodynamic therapy (PDT) efficacy, but also has an association with tumor invasiveness and metastasis. This study provides an AMH nanoparticle for targeted delivery to colorectal tumors and oxygenation-boosted immunogenic PDT in situ. AMH nanoparticle exhibits a good tumor-targeted ability to in situ produce abundant oxygen to relieve the tumor hypoxia, and initiates the potent oxygen-boosted immunogenic PDT effect under NIR irradiation to effectively inhibit the growth and recurrence of colorectal tumor. This oxygen-boosted immunogenic PDT nanosystem can be a promising candidate for advanced tumor treatment.


Subject(s)
Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Nanoparticles/chemistry , Oxygen/pharmacology , Phototherapy , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Dendritic Cells/metabolism , Female , Gold/chemistry , Hyaluronic Acid/chemistry , Manganese Compounds/chemistry , Mice, Inbred BALB C , Nanoparticles/ultrastructure , Oxides/chemistry , Photochemotherapy , Tissue Distribution
15.
Adv Sci (Weinh) ; 6(15): 1900251, 2019 Aug 07.
Article in English | MEDLINE | ID: mdl-31406665

ABSTRACT

Due to specific immune recognition receptors on the surface of T cells, their membranes are promising mimic nanocarriers for delivering drugs to tumor lesions. However, this single targeting strategy potentially compromises therapy efficacy for tumor targeting due to inter- and intra-heterogeneity of tumors. Azide (N3) or bicyclo [6.1.0] nonyne (BCN) modified unnatural sugars can be successfully incorporated into surface glycans of various tumor cells as artificial receptors, which is expected to overcome the insufficiency of single targeting. Based on this artificial tumor targeting strategy, indocyanine green nanoparticles (INPs) coated with N3-labeled T cell membrane (N3-TINPs) are constructed, which can specifically target the natural antigen and BCN artificial receptors on tumors through immune recognition and bioorthogonal chemistry, respectively. The results show that the fluorescence intensity in the tumors of mice treated with N3-TINPs is 1.5 fold compared with that of the mice treated with unlabeled TINPs. The accumulated N3-TINPs in the tumor significantly increase the photothermal therapeutic effect without adverse effect. Therefore, this T cell membrane mimicking nanoparticles based bioorthogonal chemistry may provide an alternative artificial targeting strategy for further tumor targeting photothermal therapy.

16.
Int J Mol Sci ; 20(15)2019 Jul 25.
Article in English | MEDLINE | ID: mdl-31349643

ABSTRACT

Minimizing exposure of the fetus to medication and reducing adverse off-target effects in the mother are the primary challenges in developing novel drugs to treat pregnancy complications. Nanomedicine has introduced opportunities for the development of novel platforms enabling targeted delivery of drugs in pregnancy. This review sets out to discuss the advances and potential of surface-functionalized nanoparticles in the targeted therapy of pregnancy complications. We first describe the human placental anatomy, which is fundamental for developing placenta-targeted therapy, and then we review current knowledge of nanoparticle transplacental transport mechanisms. Meanwhile, recent surface-functionalized nanoparticles for targeting the uterus and placenta are examined. Indeed, surface-functionalized nanoparticles could help prevent transplacental passage and promote placental-specific drug delivery, thereby enhancing efficacy and improving safety. We have achieved promising results in targeting the placenta via placental chondroitin sulfate A (plCSA), which is exclusively expressed in the placenta, using plCSA binding peptide (plCSA-BP)-decorated nanoparticles. Others have also focused on using placenta- and uterus-enriched molecules as targets to deliver therapeutics via surface-functionalized nanoparticles. Additionally, we propose that placenta-specific exosomes and surface-modified exosomes might be potential tools in the targeted therapy of pregnancy complications. Altogether, surface-functionalized nanoparticles have great potential value as clinical tools in the targeted therapy of pregnancy complications.


Subject(s)
Molecular Targeted Therapy , Nanoparticles , Pregnancy Complications/drug therapy , Theranostic Nanomedicine , Biological Transport , Drug Carriers/chemistry , Exosomes/metabolism , Female , Humans , Maternal-Fetal Exchange , Nanoparticles/chemistry , Placenta/anatomy & histology , Placenta/drug effects , Placenta/metabolism , Pregnancy , Pregnancy Complications/etiology , Pregnancy Complications/metabolism , Surface Properties , Theranostic Nanomedicine/methods , Uterus/drug effects , Uterus/metabolism
17.
Biomaterials ; 214: 119226, 2019 09.
Article in English | MEDLINE | ID: mdl-31174068

ABSTRACT

Bacteria-driven drug-delivery systems have attracted great attention for their enhanced therapeutic specificity and efficacy in cancer treatment. YB1, a particularly attractive genetically modified safe Salmonella Typhimurium strain, is known to penetrate hypoxic tumor cores with its self-driven properties while remarkably avoiding damage to normal tissues. Herein, nanophotosensitizers (indocyanine green (ICG)-loaded nanoparticles, INPs) were covalently attached to the surface of YB1 with amide bonds to develop a biotic/abiotic cross-linked system (YB1-INPs) for tumor precision therapy. YB1 microswimmer retained its viability after efficiently linking with INPs. This YB1-INPs treatment strategy demonstrated specific hypoxia targeting to solid tumors, perfect photothermal conversion, and efficient fluorescence (FL) imaging properties. Benefited from the combined contribution of tumor tissue destruction and the bacteria-attracting nutrients generation after photothermal treatment, the bioaccumulation of YB1-INPs was significantly improved 14-fold compared to no photothermal intervention. Furthermore, YB1-INPs pervaded throughout the large solid tumor (≥500 mm3). Under near-infrared (NIR) laser irradiation, YB1-INPs exhibited a dependable and highly efficient photothermal killing ability for eradicating the large solid tumor without relapse. This strategy of bacteria-driven hypoxia-targeting delivery has a great value for large solid tumors therapy with low toxicity and high efficiency.


Subject(s)
Salmonella/physiology , Urinary Bladder Neoplasms/therapy , Animals , Bioaccumulation , Female , Fluorescent Antibody Technique , Hyperthermia, Induced/methods , Immunohistochemistry , Indocyanine Green/chemistry , Mice , Mice, Inbred C57BL , Microbial Viability , Nanoparticles/chemistry , Phototherapy/methods , Salmonella/metabolism , Salmonella typhimurium/metabolism , Salmonella typhimurium/physiology , Temperature , Urinary Bladder Neoplasms/microbiology
18.
Small ; 15(5): e1804028, 2019 02.
Article in English | MEDLINE | ID: mdl-30589210

ABSTRACT

Metal complexes are widely used as anticancer drugs, while the severe side effects of traditional chemotherapy require new therapeutic modalities. Sonodynamic therapy (SDT) provides a significantly noninvasive ultrasound (US) treatment approach by activating sonosensitizers and initiating reactive oxygen species (ROS) to damage malignant tissues. In this work, three metal 4-methylphenylporphyrin (TTP) complexes (MnTTP, ZnTTP, and TiOTTP) are synthesized and encapsulated with human serum albumin (HSA) to form novel nanosonosensitizers. These nanosonosensitizers generate abundant singlet oxygen (1 O2 ) under US irradiation, and importantly show excellent US-activatable abilities with deep-tissue depths up to 11 cm. Compared to ZnTTP-HSA and TiOTTP-HSA, MnTTP-HSA exhibits the strongest ROS-activatable behavior due to the lowest highest occupied molecular orbital-lowest unoccupied molecular orbital gap energy by density functional theory. It is also effective for deep-tissue photoacoustic/magnetic resonance dual-modal imaging to trace the accumulation of nanoparticles in tumors. Moreover, MnTTP-HSA intriguingly achieves high SDT efficiency for simultaneously suppressing the growth of bilateral tumors away from ultrasound source in mice. This work develops a deep-tissue imaging-guided SDT strategy through well-defined metalloporphyrin nanocomplexes and paves a new way for highly efficient noninvasive SDT treatments of malignant tumors.


Subject(s)
Metalloporphyrins/chemistry , Nanoparticles/chemistry , Neoplasms/diagnosis , Neoplasms/therapy , Theranostic Nanomedicine , Ultrasonic Therapy , Animals , Humans , MCF-7 Cells , Magnetic Resonance Imaging , Mice, Nude , Nanoparticles/ultrastructure , Neoplasms/diagnostic imaging , Photoacoustic Techniques , Serum Albumin, Human/chemistry , Superoxides/metabolism
19.
ACS Nano ; 12(8): 8633-8645, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30005164

ABSTRACT

An ideal cancer therapeutic strategy is expected to possess potent ability to not only ablate primary tumors but also prevent distance metastasis and relapse. In this study, human serum albumin was hybridized with hemoglobin by intermolecular disulfide bonds to develop a hybrid protein oxygen nanocarrier with chlorine e6 encapsulated (C@HPOC) for oxygen self-sufficient photodynamic therapy (PDT). C@HPOC realized the tumor-targeted co-delivery of photosensitizer and oxygen, which remarkably relieved tumor hypoxia. C@HPOC was favorable for more efficient PDT and enhanced infiltration of CD8+ T cells in tumors. Moreover, oxygen-boosted PDT of C@HPOC induced immunogenic cell death, with the release of danger-associated molecular patterns to activate dendritic cells, T lymphocytes, and natural killer cells in vivo. Notably, C@HPOC-mediated immunogenic PDT could destroy primary tumors and effectively suppress distant tumors and lung metastasis in a metastatic triple-negative breast cancer model by evoking systemic anti-tumor immunity. This study provides a paradigm of oxygen-augmented immunogenic PDT for metastatic cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Nanoparticles/chemistry , Oxygen/chemistry , Photochemotherapy , Photosensitizing Agents/pharmacology , Serum Albumin, Human/chemistry , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Chlorophyllides , Drug Screening Assays, Antitumor , Female , Humans , Mice , Mice, Inbred BALB C , Photosensitizing Agents/chemistry , Porphyrins/chemistry , Tumor Cells, Cultured
20.
Theranostics ; 8(13): 3584-3596, 2018.
Article in English | MEDLINE | ID: mdl-30026868

ABSTRACT

Hypoxia is a characteristic feature of solid tumors and an important causation of resistance to chemotherapy and photodynamic therapy (PDT). It is challenging to develop efficient functional nanomaterials for tumor oxygenation and therapeutic applications. Methods: Through disulfide reconfiguration to hybridize hemoglobin and albumin, tumor-targeted hybrid protein oxygen carriers (HPOCs) were fabricated, serving as nanomedicines for precise tumor oxygenation and simultaneous enhancement of hypoxia-dampened chemotherapy and photodynamic therapy. Based on encapsulation of doxorubicin (DOX) and chlorin e6 (Ce6) into HPOCs to form ODC-HPOCs, the mechanism and therapeutic efficacy of oxygen-enhanced chemo-PDT was investigated in vitro and in vivo. Results: The precise oxygen preservation and release of the HPOC guaranteed sufficient tumor oxygenation, which is able to break hypoxia-induced chemoresistance by downregulating the expressions of hypoxia-inducible factor-1α (HIF-1α), multidrug resistance 1 (MDR1) and P-glycoprotein (P-gp), resulting in minimized cellular efflux of chemodrug. Moreover, the oxygen supply is fully exploited for upgrading the generation of reactive oxygen species (ROS) during the photodynamic process. As a result, only a single-dose treatment of the HPOCs-based chemo-PDT exhibited superior tumor suppression. The combination therapy was guided by in vivo fluorescence/photoacoustic imaging with nanoparticle tracking and oxygen monitoring. Conclusion: This well-defined HPOC as a versatile nanosystem is expected to pave a new way for breaking multiple hypoxia-induced therapeutic resistances to achieve highly effective treatment of solid tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Hemoglobins/administration & dosage , Neoplasms/drug therapy , Oxygen/metabolism , Porphyrins/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Animals , Carrier Proteins/administration & dosage , Cell Line, Tumor , Chlorophyllides , Disease Models, Animal , Drug Therapy/methods , Humans , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Nanomedicine/methods , Photochemotherapy/methods , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...