Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Appl Mater Interfaces ; 15(47): 54346-54352, 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-37967322

ABSTRACT

Hydrogen-bonded organic frameworks (HOFs) are a novel class of porous nanomaterials that show great potential for intracellular delivery of protein therapeutics. However, the inherent challenges in interfacing protein with HOFs, and the need for spatiotemporally controlling the release of protein within cells, have constrained their therapeutic potential. In this study, we report novel biodegradable hydrogen-bonded organic frameworks, termed DS-HOFs, specially designed for the cytosolic delivery of protein therapeutics in cancer cells. The synthesis of DS-HOFs involves the self-assembly of 4-[tris(4-carbamimidoylphenyl) methyl] benzenecarboximidamide (TAM) and 4,4'-dithiobisbenzoic acid (DTBA), governed by intermolecular hydrogen-bonding interactions. DS-HOFs exhibit high efficiency in encapsulating a diverse range of protein cargos, underpinned by the hydrogen-bonding interactions between the protein residue and DS-HOF subcomponents. Notably, DS-HOFs are selectively degraded in cancer cells triggered by the distinct intracellular reductive microenvironments, enabling an enhanced and selective release of protein inside cancer cells. Additionally, we demonstrate that the efficient delivery of bacterial effector protein DUF5 using DS-HOFs depletes the mutant RAS in cancer cells to prohibit tumor cell growth both in vitro and in vivo. The design of biodegradable HOFs for cytosolic protein delivery provides a powerful and promising strategy to expand the therapeutic potential of proteins for cancer therapy.


Subject(s)
Bacterial Proteins , Hydrogen , Cytosol , Cell Cycle , Cell Proliferation
2.
Biomacromolecules ; 24(11): 5132-5141, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37859395

ABSTRACT

Biomineralization of metal-organic frameworks (MOFs) provides a powerful approach for intracellular protein delivery, enabling the study of biological function and therapeutic potential of proteins. However, the potency of this approach is largely challenged by the low efficiency of current strategies for interfacing proteins with MOFs for biomineralization and intracellular delivery. Here, we report a versatile and convenient biomineralization strategy for the rapid encapsulation and enhanced delivery of proteins using MOFs, accelerated by histidine-rich proteins. We demonstrate that the histidine-rich green fluorescent protein (H39GFP) can accelerate the biomineralization of MOFs by promoting the coordination between proteins and metal ions, leading to enhanced protein delivery efficiency up to 15-fold. Moreover, we show that the delivery of H39GFP-fused cytotoxic ribonuclease and bacterial-derived RAS protease can effectively inhibit tumor cell growth. The strategy of promoting the biomineralization of MOFs via histidine-rich proteins for enhanced intracellular delivery could be expanded to other biomacromolecules, advancing their therapeutic potential and the biomedical scope of MOFs.


Subject(s)
Metal-Organic Frameworks , Neoplasms , Zeolites , Humans , Histidine , Zeolites/pharmacology , Zeolites/therapeutic use , Biomineralization , Metal-Organic Frameworks/pharmacology , Neoplasms/drug therapy , Green Fluorescent Proteins
3.
Biomater Sci ; 11(9): 3172-3179, 2023 May 02.
Article in English | MEDLINE | ID: mdl-36919841

ABSTRACT

Bacterial effector proteins are virulence factors that are secreted and mediate orthogonal post-translational modifications of proteins that are not found naturally in mammalian systems. They hold great promise for developing biotherapeutics by regulating malignant cell signaling in a specific and targeted manner. However, delivering bacterial effectors into disease cells poses a significant challenge to their therapeutic potential. In this study, we report on the design of a combinatorial library of bioreducible lipid nanoparticles containing disulfide bonds for highly efficient bacterial effector delivery and potential cancer therapy. A leading lipid, PPPDA-O16B, identified from the library, can encapsulate and deliver DNA plasmids into cells. The gene cargo is released in response to the reductive cellular environment that is upregulated in cancer cells, leading to enhanced gene delivery and protein expression efficiency. Furthermore, we demonstrate that PPPDA-O16B can deliver the bacterial effector protein, DUF5, to degrade mutant RAS and inactivate downstream MAPK signaling cascades to suppress cancer cell growth in vitro and in tumor-bearing mouse xenografts. This strategy of delivering bacterial effectors using biodegradable lipid nanoparticles can be expanded for cancer cell signaling regulation and antitumor studies.


Subject(s)
Nanoparticles , Neoplasms , Mice , Humans , Animals , Gene Transfer Techniques , Nanoparticles/chemistry , Genetic Therapy , Bacterial Proteins , Neoplasms/drug therapy , Mammals
4.
Angew Chem Int Ed Engl ; 60(51): 26740-26746, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34622541

ABSTRACT

The delivery of protein into mammalian cells enables the dissection and manipulation of biological processes; however, this potency is challenged by the lack of an efficient protein delivery tool and a means to monitor its intracellular trafficking. Herein, we report that the hierarchical self-assembly of tetraphenylethylene (TPE)-featured metal-organic cages (MOCs) and ß-cyclodextrin-conjugated polyethylenimine can generate fluorescent supramolecular nanoparticles (FSNPs) to deliver protein into neural cells, a cell line that is hard to transfect using conventional strategy. Further, the aggregation-induced emission (AIE) of TPE enabled the fluorescent monitoring of cytosolic protein release. It is found that FSNPs can deliver and release protein into cytosol for subcellular targeting as fast as 18 h post-delivery. Moreover, the delivery of molecular chaperone DJ-1 using FSNPs activates MAPK/ERK signaling of neural cells to protect cells from oxidative stress.


Subject(s)
Fluorescent Dyes/pharmacology , Nanoparticles/chemistry , Neural Stem Cells/drug effects , Stilbenes/pharmacology , Cell Line, Tumor , Fluorescent Dyes/chemistry , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/pharmacology , Neural Stem Cells/metabolism , Oxidative Stress/drug effects , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacology , Stilbenes/chemistry , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/pharmacology
5.
Angew Chem Int Ed Engl ; 60(41): 22315-22321, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34382314

ABSTRACT

Hydrogen-bonded organic frameworks (HOFs) are porous materials with great potential for biological applications. The self-assembly of HOFs and biomacromolecules, however, is challenging. We report herein the self-assembly of nanoscale HOFs (nHOFs) to encapsulate protein for intracellular biocatalysis. The self-assembly of tetrakis(4-amidiniumphenyl)methane and azobenzenedicarboxylate can encapsulate protein in situ to form protein@nHOFs under mild conditions. This strategy is applicable to proteins with different surface charge and molecular weight, showing a high protein encapsulation efficiency and minimal effect on protein activity. A cellular delivery study shows that the protein@TA-HOFs can efficiently enter cells and retain enzyme activity for biochemical catalysis in living cells for neuroprotection. Our strategy paves new avenues for interfacing nHOFs with biological settings and sheds light on expanding nHOFs as a platform for biomacromolecule delivery and disease treatment.


Subject(s)
Azo Compounds/metabolism , Carboxylic Acids/metabolism , Methane/metabolism , Proteins/metabolism , Azo Compounds/chemistry , Biocatalysis , Carboxylic Acids/chemistry , Cell Line, Tumor , Cell Survival , Humans , Hydrogen Bonding , Methane/analogs & derivatives , Methane/chemistry , Molecular Structure , Particle Size , Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...