Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
ACS Nano ; 18(11): 7923-7936, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38445625

ABSTRACT

Tumor whole cell, carrying a complete set of tumor-associated antigens and tumor-specific antigens, has shown great potential in the construction of tumor vaccines but is hindered by the complex engineering means and limited efficacy to cause immunity. Herein, we provided a strategy for the self-mineralization of autologous tumor cells with palladium ions in microfluidic droplets, which endowed the engineered cells with both immune and catalytic functions, to establish a bioorthogonally catalytic tumor whole-cell vaccine. This vaccine showed strong inhibition both in the occurrence and recurrence of tumor by invoking the immediate antitumor immunity and building a long-term immunity.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , Microfluidics , Immunotherapy , Neoplasms/therapy , Antigens, Neoplasm
2.
ACS Nano ; 18(12): 9031-9042, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38470458

ABSTRACT

Cuproptosis has drawn enormous attention in antitumor material fields; however, the responsive activation of cuproptosis against tumors using nanomaterials with high atom utilization is still challenging. Herein, a copper-based nanoplatform consisting of acid-degradable copper hydride (CuH) nanoparticles was developed via a microfluidic synthesis. After coating with tumor-targeting hyaluronic acid (HA), the nanoplatform denoted as HA-CuH-PVP (HCP) shows conspicuous damage toward tumor cells by generating Cu+ and hydrogen (H2) simultaneously. Cu+ can induce apoptosis by relying on Fenton-like reactions and lead to cuproptosis by causing mitochondrial protein aggregation. Besides, the existence of H2 can enhance both cell death types by causing mitochondrial dysfunction and intracellular redox homeostatic disorders. In vivo experimental results further exhibit the desirable potential of HCP for killing tumor cells and inhibiting lung metastases, which will broaden the horizons of designing copper-based materials triggering apoptosis and cuproptosis for better antitumor efficacy.


Subject(s)
Copper , Nanoparticles , Microfluidics , Apoptosis , Hyaluronic Acid , Hydrogen
3.
Adv Healthc Mater ; 13(14): e2303683, 2024 06.
Article in English | MEDLINE | ID: mdl-38386961

ABSTRACT

Employing tumor whole cells for tumor immunotherapy is a promising tumor therapy proposed in the early stage, but its therapeutic efficacy is weakened by the methods of eliminating pathogenicity and the mass ratio of the effective antigen carried by itself. Here, by adding gold ion to live cancer cells in the microfluidic droplets, this work obtains dead tumor whole cells with NIR-controlled catalytic ability whose pathogenicity is removed while plenary tumor antigens, major structure, and homing ability are reserved. The engineered tumor cell (Cell-Au) with the addition of prodrug provides 1O2 in an O2-free Russell mechanism, which serves better in a hypoxic tumor microenvironment. This tumor whole-cell catalytic vaccine (TWCV) promotes the activation of dendritic cells and the transformation of macrophages into tumor suppressor phenotype. In 4T1 tumor-bearing mice, the Cell-Au-based vaccine supports the polarization of cytotoxicity T cells, resulting in tumor eradication and long-term animal survival. Compared with antigen vaccines or adoptive cell therapy which takes months to obtain, this TWCV can be prepared in just a few days with satisfactory immune activation and tumor therapeutic efficacy, which provides an alternative way for the preparation of personalized tumor vaccines across tumor types and gives immunotherapy a new path.


Subject(s)
Cancer Vaccines , Gold , Immunotherapy , Animals , Gold/chemistry , Immunotherapy/methods , Mice , Cell Line, Tumor , Cancer Vaccines/immunology , Cancer Vaccines/chemistry , Mice, Inbred BALB C , Catalysis , Female , Tumor Microenvironment/immunology , Metal Nanoparticles/chemistry , Dendritic Cells/immunology , Humans , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/pathology
5.
Acta Pharm Sin B ; 14(2): 795-807, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38322334

ABSTRACT

Recent innovations in nanomaterials inspire abundant novel tumor-targeting CRISPR-based gene therapies. However, the therapeutic efficiency of traditional targeted nanotherapeutic strategies is limited by that the biomarkers vary in a spatiotemporal-dependent manner with tumor progression. Here, we propose a self-amplifying logic-gated gene editing strategy for gene/H2O2-mediated/starvation multimodal cancer therapy. In this approach, a hypoxia-degradable covalent-organic framework (COF) is synthesized to coat a-ZIF-8 in which glucose oxidase (GOx) and CRISPR system are packaged. To intensify intracellular redox dyshomeostasis, DNAzymes which can cleave catalase mRNA are loaded as well. When the nanosystem gets into the tumor, the weakly acidic and hypoxic microenvironment degrades the ZIF-8@COF to activate GOx, which amplifies intracellular H+ and hypoxia, accelerating the nanocarrier degradation to guarantee available CRISPR plasmid and GOx release in target cells. These tandem reactions deplete glucose and oxygen, leading to logic-gated-triggered gene editing as well as synergistic gene/H2O2-mediated/starvation therapy. Overall, this approach highlights the biocomputing-based CRISPR delivery and underscores the great potential of precise cancer therapy.

6.
Angew Chem Int Ed Engl ; 63(2): e202316007, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38017308

ABSTRACT

Psoriasis is a chronic skin inflammation characterized by dysregulated crosstalk between immune cells and keratinocytes. Here we show that the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is a key regulator of psoriatic inflammation in a mouse model. Platinum-doped positively charged carbon dots (Pt-CDs) were designed to inhibit the cGAS-STING pathway. By inhibiting the cGAS-STING pathway with Pt-CDs, the secretion of proinflammatory cytokines in macrophages was reduced, and the proinflammatory cytokines-induced breakdown of immunological tolerance and overexpression of chemokines in keratinocytes was restored, which reversed the homeostatic imbalance through breaking these cytokines-mediated intercellular positive feedback loop. Topical Pt-CDs treatment exhibited therapeutic effects in imiquimod-induced psoriasis mice without noticeable toxicity. The reversal of elevated expression of STING, phosphorylated STING, and downstream genes within psoriatic lesions indicates that Pt-CDs effectively inhibit the cGAS-STING pathway. This work suggests a promising strategy for psoriasis treatment by targeting the cGAS-STING pathway with Pt-CDs nanoinhibitor to restore skin homeostatic balance.


Subject(s)
Psoriasis , Signal Transduction , Mice , Animals , Nucleotidyltransferases/metabolism , Inflammation/drug therapy , Cytokines/metabolism , Psoriasis/drug therapy
7.
ACS Nano ; 17(10): 9313-9325, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37155357

ABSTRACT

Type 2 diabetes (T2D) results from the cells' insulin resistance, and to date, insulin therapy and diabetes medications targeting glycemic management have failed to reverse the increase in T2D prevalence. Restoring liver functions to improve hepatic insulin resistance by reducing oxidative stress is a potential strategy for T2D treatment. Herein, the liver-targeted biodegradable silica nanoshells embedded with platinum nanoparticles (Pt-SiO2) are designed as reactive oxygen species (ROS) nanoscavengers and functional hollow nanocarriers. Then, 2,4-dinitrophenol-methyl ether (DNPME, mitochondrial uncoupler) is loaded inside Pt-SiO2, followed by coating a lipid bilayer (D@Pt-SiO2@L) for long-term effective ROS removal (platinum nanoparticles scavenge overproduced ROS, while DNPME inhibits ROS production) in the liver tissue of T2D models. It is found that D@Pt-SiO2@L reverses elevated oxidative stress, insulin resistance, and impaired glucose consumption in vitro, and significantly improves hepatic steatosis and antioxidant capacity in diabetic mice models induced by a high-fat diet and streptozotocin. Moreover, intravenous administration of D@Pt-SiO2@L indicates therapeutic effects on hyperlipidemia, insulin resistance, hyperglycemia, and diabetic nephropathy, which provides a promising approach for T2D treatment by reversing hepatic insulin resistance through long-term ROS scavenging.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Insulin Resistance , Metal Nanoparticles , Mice , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Reactive Oxygen Species/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Silicon Dioxide/metabolism , Platinum/pharmacology , Liver/metabolism , Insulin/metabolism , Oxidative Stress
8.
Med Eng Phys ; 113: 103961, 2023 03.
Article in English | MEDLINE | ID: mdl-36966005

ABSTRACT

BACKGROUND: Exoskeletons have become an important tool to help patients with upper extremity motor dysfunction in rehabilitation training and life assistance. In the study of the upper limb exoskeleton, the human glenohumeral joint will produce accompanying movement during the movement of the shoulder joint. This phenomenon causes a positional deviation between the shoulder joint and the exoskeleton, which affects the accuracy of exoskeleton-assisted human movement and the wearing comfort. Spend. METHOD: Taking the coronal adduction and abduction of the shoulder joint as the research object, the shoulder joint angle and glenohumeral joint bony motion trajectory were fitted by bi-level X-rays, and then the Ultium Motion motion capture system was used to collect the characteristic motion of the shoulder joint surface and establish a model. A back-propagation neural network with shoulder joint motion and shoulder width as input and the coronal position of the glenohumeral joint as output, finally applied the model to the Nimbot exoskeleton upper limb rehabilitation training robot to verify the effectiveness of the algorithm. RESULTS: Real-time prediction of the glenohumeral joint motion trajectory was achieved, and the human-machine coupling compliance during the wearing of the upper limb exoskeleton was improved.


Subject(s)
Robotics , Shoulder Joint , Robotics/instrumentation , Robotics/methods , Biomechanical Phenomena , Humans , Upper Extremity
9.
ACS Nano ; 17(5): 4515-4525, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36847587

ABSTRACT

Targeted construction of therapeutic nanoplatforms in tumor cells with specific activation remains appealing but challenging. Here, we design a cancer-motivated upconversion nanomachine (UCNM) based on porous upconversion nanoparticles (p-UCNPs) for precise phototherapy. The nanosystem is equipped with a telomerase substrate (TS) primer and simultaneously encapsulates 5-aminolevulinic acid (5-ALA) and d-arginine (d-Arg). After coating with hyaluronic acid (HA), it can readily get into tumor cells, where 5-ALA induces efficient accumulation of protoporphyrin IX (PpIX) via the inherent biosynthetic pathway, and the overexpressed telomerase prolonged the TS to form G-quadruplexes (G4) for binding the resulting PpIX as a nanomachine. This nanomachine can respond to near-infrared (NIR) light and promote the active singlet oxygen (1O2) production due to the efficiency of Förster resonance energy transfer (FRET) between p-UCNPs and PpIX. Intriguingly, such oxidative stress can oxidize d-Arg into nitric oxide (NO), which relieves the tumor hypoxia and in turn improves the phototherapy effect. This in situ assembly approach significantly enhances targeting in cancer therapy and might be of considerable clinical value.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Telomerase , Humans , Photochemotherapy/methods , Telomerase/metabolism , Infrared Rays , Phototherapy , Neoplasms/drug therapy , Nanoparticles/therapeutic use , Aminolevulinic Acid/therapeutic use , Reactive Oxygen Species/metabolism , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Cell Line, Tumor
10.
Anal Chem ; 95(2): 1618-1626, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36541937

ABSTRACT

CRISPR/Cas12a shows excellent potential in disease diagnostics. However, insensitive signal conversion strategies hindered its application in detecting protein biomarkers. Here, we report a metal-organic framework (MOF)-based DNA bio-barcode integrated with the CRISPR/Cas12a system for ultrasensitive detection of protein biomarkers. In this work, zirconium-based MOF nanoparticles were comodified with antibodies and bio-barcode phosphorylated DNA as an efficient signal converter, which not only recognized the protein biomarker to form the sandwich complex but also released the bio-barcode DNA activators after MOF dissociation to activate the trans-cleavage activity of Cas12a. Due to the obvious advantages, including numerous loaded oligonucleotides, a convenient release process, and the nontoxic release reagent, this MOF-DNA bio-barcode strategy could amplify the CRISPR/Cas12a system to achieve simple and highly sensitive detection of tumor protein biomarkers with detection limits of 0.03 pg/mL (PSA) and 0.1 pg/mL (CEA), respectively. Furthermore, this platform could detect PSA directly in clinical serum samples, offering a powerful tool for early disease diagnosis.


Subject(s)
Biosensing Techniques , Metal-Organic Frameworks , CRISPR-Cas Systems/genetics , Biomarkers, Tumor/genetics , DNA , Antibodies
11.
Nano Lett ; 22(23): 9714-9722, 2022 12 14.
Article in English | MEDLINE | ID: mdl-36412588

ABSTRACT

CRISPR/Cas12a has shown great potential in molecular diagnostics, but its application in sensing of microRNAs (miRNAs) was limited by sensitivity and complexity. Here, we have sensitively and conveniently detected microRNAs by reasonably integrating metal-organic frameworks (MOFs) based biobarcodes with CRISPR/Cas12a assay (designated as MBCA). In this work, DNA-functionalized Zr-MOFs were designed as the converter to convert and amplify each miRNA target into activators that can initiate the trans-cleavage activity of CRISPR/Cas12a to further amplify the signal. Such integration provides a universal strategy for sensitive detection of miRNAs. By tuning the complementary sequences modified on nanoprobes, this assay achieves subattomolar sensitivity for different miRNAs and was selective to single-based mismatches. With the proposed method, the expression of miR-21 in different cancer cells can be assessed, and breast cancer patients and healthy individuals can be differentiated by analyzing the target miRNAs extracted from serum samples, holding great potential in clinical diagnosis.


Subject(s)
Biosensing Techniques , Breast Neoplasms , Metal-Organic Frameworks , MicroRNAs , Humans , Female , MicroRNAs/genetics , CRISPR-Cas Systems/genetics , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Cell Differentiation
12.
Biomaterials ; 291: 121871, 2022 12.
Article in English | MEDLINE | ID: mdl-36323073

ABSTRACT

Effective activation of cGAS-STING pathway combined with immune checkpoint blockade (ICB) within the immunosuppressive tumor microenvironment to induce stronger immune responsiveness yet remains challenging. CRISPR-Cas9 gene editing technology, which offers the benefits of permanence and irreversibility, could recognize the target genome sequence with sgRNA (Guide RNA) and guide the Cas9 protease to knock down the target gene. Herein, a nanoplatform (HMnMPH) for dual activation of cGAS-STING pathway in combination with CRISPR-Cas9 gene editing to silence programmed death ligand 1 (PD-L1) to trigger long-term immunotherapy was reported. The HMnMPH consists of hollow manganese dioxide (HMn) loaded with STING agonist (MSA-2) and CRISPR-Cas9/sg-PD-L1 plasmid with further modification of hyaluronic acid (HA). In acidic and GSH overexpressed tumor environment, HMnPMH was degraded to release large amounts of Mn ions and STING agonists, strongly and persistently activating the cGAS-STING pathway to promote the release of type I interferon and pro-inflammatory factors. Meanwhile, the released CRISPR-Cas9 plasmid could knockdown the PD-L1 immune checkpoint and restart immunosuppressive T cells to differentiate into cytotoxic T lymphocytes significantly, which reduced the activity of primary and distal tumors and demonstrated a long-term immune memory effect on distal tumors.


Subject(s)
Gene Editing , Neoplasms , Humans , B7-H1 Antigen/metabolism , CRISPR-Cas Systems/genetics , Immunotherapy , Neoplasms/genetics , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Tumor Microenvironment
13.
Acta Pharm Sin B ; 12(11): 4224-4234, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36386466

ABSTRACT

Near-infrared (NIR)-light-triggered nanomedicine, including photodynamic therapy (PDT) and photothermal therapy (PTT), is growing an attractive approach for cancer therapy due to its high spatiotemporal controllability and minimal invasion, but the tumor eradication is limited by the intrinsic anti-stress response of tumor cells. Herein, we fabricate a tumor-microenvironment responsive CRISPR nanoplatform based on oxygen-deficient titania (TiO2-x ) for mild NIR-phototherapy. In tumor microenvironment, the overexpressed hyaluronidase (HAase) and glutathione (GSH) can readily destroy hyaluronic acid (HA) and disulfide bond and releases the Cas9/sgRNA from TiO2-x to target the stress alleviating regulators, i.e., nuclear factor E2-related factor 2 (NRF2) and heat shock protein 90α (HSP90α), thereby reducing the stress tolerance of tumor cells. Under subsequent NIR light illumination, the TiO2-x demonstrates a higher anticancer effect both in vitro and in vivo. This strategy not only provides a promising modality to kills cancer cells in a minimal side-effects manner by interrupting anti-stress pathways but also proposes a general approach to achieve controllable gene editing in tumor region without unwanted genetic mutation in normal environments.

14.
Small ; 18(40): e2204244, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36055775

ABSTRACT

As a promising therapeutic modality targeting cancer, gas therapy still faces critical challenges, especially in enhancing therapeutic efficacy and avoiding gas poisoning risks. Here, a pH/glutathione (GSH) dual stimuli-responsive CRISPR/Cas9 gene-editing nanoplatform combined with calcium-enhanced CO gas therapy for precise anticancer therapy, is established. In the tumor microenvironment (TME), the fast biodegradation of the CaCO3 layer via pH-induced hydrolyzation allows glucose oxidase (GOx) to catalyze glucose for H2 O2 production, which further reacts with manganese carbonyl (MnCO) and achieves the precise release of CO gas. Simultaneously, in situ Ca2+ overload from CaCO3 degradation disturbs mitochondrial Ca2+ homeostasis, resulting in Ca2+ -driven reactive oxygen species (ROS) formation and subsequent mitochondrial apoptosis signaling pathway activation. Subsequently, by GSH-induced cleavage of a disulfide bond, the released Cas9/sgRNA (RNP) can achieve nuclear factor E2-related factor 2 (Nrf2) gene ablation to sensitize gas therapy by interfering with ROS signaling. This therapeutic modality endows codelivery of CRISPR, ions, and gas with smart control features, which demonstrates great potential for future clinical applications in precise nanomedicine.


Subject(s)
Nanoparticles , Neoplasms , Calcium , Carbon Monoxide/therapeutic use , Cell Line, Tumor , Disulfides , Gene Editing/methods , Glucose , Glucose Oxidase , Glutathione , Humans , Ions , Manganese , NF-E2-Related Factor 2/therapeutic use , Nanoparticles/chemistry , Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Tumor Microenvironment
15.
Biosens Bioelectron ; 209: 114239, 2022 Aug 01.
Article in English | MEDLINE | ID: mdl-35429769

ABSTRACT

In addition to applications in genome editing, clustered regularly interspaced short palindromic repeats (CRISPR) have recently been engineered for medical diagnostics based on their trans-cleavage activity owing to their high base resolution and isothermal signal amplification. However, trans-cleavage activity is too fragile to be applied in vivo. Herein, we introduce a hollow covalent organic framework (COF)-sheltering CRISPR/aptamer-based sensor (h-CCS) for ATP imaging in living animals. The CRISPR/aptamer-based complex is comprised of the CRISPR-Cas12a system, fluorophore quencher-labeled single-stranded DNA substrate (ssDNA-FQ), and a DNA activator that pre-hybridizes with ATP aptamer to prevent the trans-cleavage activity of the Cas12a system in the absence of ATP. After being encapsulated in a hollow COF, the constructed nanoreactor is highly robust and can be lit up by ATP for in vivo imaging. Considering the unique properties of h-CCS, this strategy offers great potential to broaden applications of not only CRISPR-Cas systems but also other proteins in porous matrixes for clinical diagnostics, medical research, and biomimetic nanodevices.


Subject(s)
Biosensing Techniques , Metal-Organic Frameworks , Adenosine Triphosphate , Animals , Biosensing Techniques/methods , CRISPR-Cas Systems/genetics , DNA, Single-Stranded , Gene Editing/methods , Oligonucleotides
16.
Angew Chem Int Ed Engl ; 61(14): e202114239, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35080112

ABSTRACT

Cancer has become a leading cause of morbidity and mortality, and there is an increasing need for versatile tools to enable sensitive, simple and early cancer monitoring. Here, we report platinum supernanoparticles as an exogenous nanosensor which can dissociate into ultrasmall platinum nanoclusters (PtNCs) under tumor-specific hypoxia conditions. The resulting PtNCs can be filtered through the kidney as urinary reporters to be quantified by a companion volumetric bar-chart chip (V-Chip) for point-of-care analysis. The V-Chip signals of triple-negative breast cancer and its lung metastasis mouse model showed a significant increase compared to healthy mice. Our nanosensor can also noninvasively monitor the course of treatment, which is significant for screening tumor recurrence and individualized evaluation of pharmacological and follow-up efficacy. Importantly, this strategy could be adapted for various diseases to form a common diagnostic platform by changing responsive linkers.


Subject(s)
Lung Neoplasms , Platinum , Animals , Hypoxia , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/drug therapy , Mice , Microfluidics , Point-of-Care Systems
17.
Angew Chem Int Ed Engl ; 60(39): 21200-21204, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34297462

ABSTRACT

Near-infrared (NIR)-light-triggered photothermal therapy (PTT) is usually associated with undesirable damage to healthy organs nearby due to the high temperatures (>50 °C) available for tumor ablation. Low-temperature PTT would therefore have tremendous value for clinical application. Here, we construct a hypoxia-responsive gold nanorods (AuNRs)-based nanocomposite of CRISPR-Cas9 for mild-photothermal therapy via tumor-targeted gene editing. AuNRs are modified with azobenzene-4,4'-dicarboxylic acid (p-AZO) to achieve on-demand release of CRISPR-Cas9 using hypoxia-responsive azo bonds. In the hypoxic tumor microenvironment, the azo groups of the hypoxia-activated CRISPR-Cas9 nanosystem based on gold nanorods (APACPs) are selectively reduced by the overexpression of reductases, leading to the release of Cas9 and subsequent gene editing. Owing to the knockout of HSP90α for reducing the thermal resistance of cancer cells, highly effective tumor ablation both in vitro and in vivo was achieved with APACPs under mild PTT.


Subject(s)
Antineoplastic Agents/pharmacology , Azo Compounds/pharmacology , CRISPR-Cas Systems/drug effects , Cell Hypoxia/drug effects , Dicarboxylic Acids/pharmacology , Gold/pharmacology , Photothermal Therapy , A549 Cells , Antineoplastic Agents/chemistry , Azo Compounds/chemistry , CRISPR-Cas Systems/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Dicarboxylic Acids/chemistry , Drug Screening Assays, Antitumor , Gene Editing , Gold/chemistry , Humans , Infrared Rays , Metal Nanoparticles/chemistry , Particle Size
18.
Biosens Bioelectron ; 190: 113450, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34197999

ABSTRACT

Apart from the great potential in genome editing, the clustered regularly interspaced short palindromic repeat (CRISPR)/Cas system has recently been widely used in biosensing. However, due to the complex and inefficient signal conversion strategies, most of the works focused on nucleic acid analysis rather than protein biomarkers. Herein, by employing DNA-AuNPs (gold nanoparticles) nanotechnology to activate trans-cleavage activity of CRISPR/Cas12a, a universal signal transduction strategy was established between trans-cleavage of CRISPR/Cas12a and protein analytes. As a result, a sensitive platform was developed for sensing carcinoembryonic antigen (CEA) and prostate specific-antigen (PSA) biomarkers, which was designated as Nano-CLISA (Nano-immunosorbent assay based on Cas12a/crRNA). Nano-CLISA was directly employed to test PSA in clinical samples, indicating its great potential in practical detection. This platform has been used to quantitatively analyze protein at attomolar levels, which was 1000-fold more sensitive than traditional ELISA, and the detection range is 15 times wider than that of traditional ELISA.


Subject(s)
Biosensing Techniques , Metal Nanoparticles , CRISPR-Cas Systems , Gold , Immunosorbents
19.
J Mater Chem B ; 9(35): 7076-7099, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34124735

ABSTRACT

Light has been present throughout the history of mankind and even the universe. It is of great significance to human life, contributing to energy, agriculture, communication, and much more. In the biomedical field, light has been developed as a switch to control medical processes with minimal invasion and high spatiotemporal selectivity. During the past three years, near-infrared (NIR) light as long-wavelength light has been applied to more than 3000 achievements in biological applications due to its deep penetration depth and low phototoxicity. Remotely controlled cancer therapy usually involves the conversion of biologically inert NIR light. Thus, various materials, especially nanomaterials that can generate reactive oxygen species (ROS), ultraviolet (UV)/visual light, or thermal energy and so on under NIR illumination achieve great potential for the research of nanomedicine. Here, we offered an overview of recent advances in NIR light-activated nanomedicine for cancer therapeutic applications. NIR-light-conversion nanotechnologies for both directly triggering nanodrugs and smart drug delivery toward tumor therapy were discussed emphatically. The challenges and future trends of the use of NIR light in biomedical applications were also provided as a conclusion. We expect that this review will spark inspiration for biologists, materials scientists, pharmacologists, and chemists to fight against diseases and boost the future clinical-translational applications of NIR technology-based precision nanomedicine.


Subject(s)
Antineoplastic Agents/therapeutic use , Biocompatible Materials/therapeutic use , Nanomedicine , Neoplasms/drug therapy , Precision Medicine , Antineoplastic Agents/chemistry , Biocompatible Materials/chemistry , Humans , Infrared Rays , Materials Testing
20.
Adv Healthc Mater ; 10(3): e2000973, 2021 02.
Article in English | MEDLINE | ID: mdl-33369176

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic relapsing autoimmune disease that is characterized by segmental intestinal inflammation. There is an urgent need for more efficient inflammation-targeting strategies to improve therapeutic effect and reduce systemic drug exposure. Herein, an oxidation-responsive metal-organic framework material (Ce-MOF@PSS) is reported that preferentially adheres to inflamed intestine via enema. The overproduced reactive oxygen species (ROS) at inflammatory sites induces transformation of Ce-MOF@PSS from mesopore to macropore with local drug release. In experimental colitis, the Ce-MOF@PSS delivery system exhibits excellent inflammation-targeting efficacy and superior therapeutic effect over free drug on suppressing inflammation and repairing intestinal barrier function. Accordingly, by targeting intestinal inflammation, increasing local drug concentrations, scavenging ROS, reducing systemic exposure, and exhibiting excellent safety profiles, it is considered that the Ce-MOF drug delivery platform can be intensively developed as a translational nanomedicine for the management of IBD and other inflammatory diseases.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Metal-Organic Frameworks , Pharmaceutical Preparations , Drug Delivery Systems , Humans , Inflammatory Bowel Diseases/drug therapy , Metal-Organic Frameworks/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...