Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Biomater Sci ; 12(8): 1965-1980, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38454904

ABSTRACT

Many studies suggest that tumor microbiome closely relates to the oncogenesis and anti-tumor responses in multiple cancer types (e.g., colorectal cancer (CRC), breast cancer, lung cancer and pancreatic cancer), thereby raising an emerging research area of bacteria-related tumor therapy. Nanomaterials have long been used for both cancer and bacterial infection treatment, holding great potential for bacteria-related tumor therapy. In this review, we summarized recent progress in nanomaterials for bacteria-related tumor therapy. We focus on the types and mechanisms of pathogenic bacteria in the development and promotion of cancers and emphasize how nanomaterials work. We also briefly discuss the design principles and challenges of nanomaterials for bacteria-related tumor therapy. We hope this review can provide some insights into this emerging and rapidly growing research area.


Subject(s)
Lung Neoplasms , Nanostructures , Pancreatic Neoplasms , Humans , Bacteria , Cell Transformation, Neoplastic , Nanostructures/therapeutic use
2.
Adv Mater ; 36(19): e2309927, 2024 May.
Article in English | MEDLINE | ID: mdl-38387609

ABSTRACT

Cytokines are powerful in cancer immunotherapy, however, their therapeutic potential is limited by the severe systemic toxicity. Here a potent strategy to reduce the toxicity of systemic cytokine therapy by delivering its denatured form using a finely designed nanochaperone, is described. It is demonstrated that even if the denatured protein cargos are occasionally released under normal physiological conditions they are still misfolded, while can effectively refold into native states and release to function in tumor microenvironment. Consequently, the systemic toxicity of cytokines is nearly completely overcome. Moreover, an immunogenic cell death (ICD)-inducing chemotherapeutic is further loaded and delivered to tumor using this nanochaperone to trigger the release of tumor-associated antigens (TAAs) that are subsequently captured in situ by nanochaperone and then reflows into lymph nodes (LNs) to promote antigen cross-presentation. This optimized personalized nanochaperone-vaccine demonstrates unprecedented suppressive effects against large, advanced tumors, and in combination with immune checkpoint blockade (ICB) therapy results in a significant abscopal effect and inhibition of postoperative tumor recurrence and metastasis. Hence, this approach provides a simple and universal delivery strategy to reduce the systemic toxicities of cytokines, as well as provides a robust personalized cancer vaccination platform, which may find wide applications in cancer immunotherapy.


Subject(s)
Antigens, Neoplasm , Immunotherapy , Interleukin-12 , Nanostructures , Animals , Humans , Mice , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cancer Vaccines/chemistry , Cell Line, Tumor , Immunogenic Cell Death/drug effects , Immunotherapy/methods , Interleukin-12/chemistry , Interleukin-12/metabolism , Interleukin-12/toxicity , Nanoparticles/chemistry , Nanostructures/chemistry , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/pathology , Protein Folding , Tumor Microenvironment/drug effects
3.
Small Methods ; 8(3): e2301309, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38018349

ABSTRACT

Fusobacterium nucleatum (Fn) existing in the community of colorectal cancer (CRC) promotes CRC progression and causes chemotherapy resistance. Despite great efforts that have been made to overcome Fn-induced chemotherapy resistance by co-delivering antibacterial agents and chemotherapeutic drugs, increasing the drug-loading capacity and enabling controlled release of drugs remain challenging. In this study, a novel supramolecular upconversion nanoparticle (SUNP) is constructed by incorporating a positively charged polymer (PAMAM-LA-CD) with Fn inhibition capacity, a negatively charged platinum (IV) oxaliplatin prodrug (OXA-COOH), upconversion nanoparticle (UCNPs) and polyethylene glycol-azobenzene (PEG-Azo) to enhance drug-loading and enable on-demand drug release for drug-resistant CRC treatment. SUNPs exhibit high drug-loading capacity (30.8%) and good structural stability under normal physiological conditions, while disassembled upon exogenous NIR excitation and endogenous azo reductase in the CRC microenvironment to trigger drug release. In vitro and in vivo studies demonstrate that SUNPs presented good biocompatibility and robust performance to overcome chemoresistance, thereby significantly inhibiting Fn-infected cancer cell proliferation. This study leverages multiple dynamic chemical designs to integrate both advantages of drug loading and release in a single system, which provides a promising candidate for precision therapy of bacterial-related drug-resistant cancers.


Subject(s)
Colorectal Neoplasms , Fusobacterium nucleatum , Humans , Fusobacterium nucleatum/physiology , Colorectal Neoplasms/drug therapy , Nanomedicine , Tumor Microenvironment
4.
Mater Horiz ; 10(12): 5547-5554, 2023 11 27.
Article in English | MEDLINE | ID: mdl-37843027

ABSTRACT

Regulating protein folding including assisting de novo folding, preventing misfolding and aggregation, and facilitating refolding of proteins are of significant importance for retaining protein's biological activities. Here, we report a mixed shell polymeric micelle (MSPM)-based self-cooperative nanochaperone (self-CO-nChap) with enhanced activity to facilitate protein refolding. This self-CO-nChap was fabricated by introducing Hsp40-mimetic artificial carriers into the traditional nanochaperone to cooperate with the Hsp70-mimetic confined hydrophobic microdomains. The artificial carrier facilitates transfer and immobilization of client proteins into confined hydrophobic microdomains, by which significantly improving self-CO-nChap's capability to inhibit unfolding and aggregation of client proteins, and finally facilitating refolding. Compared to traditional nanochaperones, the self-CO-nChap significantly enhances the thermal stability of horseradish peroxidase (HRP) epicyclically under harsher conditions. Moreover, the self-CO-nChap efficiently protects misfolding-prone proteins, such as immunoglobulin G (IgG) antibody from thermal denaturation, which is hardly achieved using traditional nanochaperones. In addition, a kinetic partitioning mechanism was devised to explain how self-CO-nChap facilitates refolding by regulating the cooperative effect of kinetics between the nanochaperone and client proteins. This work provides a novel strategy for the design of protein folding regulatory materials, including nanochaperones.


Subject(s)
HSP70 Heat-Shock Proteins , Polymers , Humans , Protein Refolding
6.
Article in English | MEDLINE | ID: mdl-36916659

ABSTRACT

Fusobacterium nucleatum (Fn) has long been found to be related to colorectal cancer (CRC), which could promote colorectal tumor progression and cause cancer resistance to chemotherapy. Great efforts have been made to understand the relationship between Fn and CRC, but how to efficiently eliminate intratumoral Fn and overcome chemoresistance remains a critical challenge. Here, an active tumor-targeting acidity-responsive nanomaterial toward eliminating intratumoral Fn is developed for enhancing the treatment of cancer. Lauric acid and phenylboric acid are conjugated to oligomethyleneimine to form OLP followed by interacting with oxaliplatin prodrug-modified polyglycidyl ether (PP) to obtain the OLP/PP nanoassembly. The nanoassembly shows good structural stability under the simulated physiological conditions and has a pH-responsive drug release in an acidic tumor microenvironment. More attractively, the nanoassembly can specifically target the tumor cell, guide cellular uptake, and efficiently eliminate tumor-resident extracellular and intracellular Fn. Through the on-site drug delivery, the nanoassembly can overcome chemoresistance and significantly inhibit tumor growth. Both in vitro and vivo studies show that the prepared nanoassembly presents good biocompatibility. Therefore, this biocompatible nanoassembly possessing efficient antibacterial and antitumor activities provides new promise for the therapy of bacterial infected tumors.

7.
Small Methods ; 7(5): e2201051, 2023 05.
Article in English | MEDLINE | ID: mdl-36228110

ABSTRACT

Despite unprecedented successes of antibody-based cancer immunotherapy, the serious side effects and rapid clearance following systemic administration remain big challenges to realize its full potential. At the same time, combination immunotherapy using multiple antibodies has shown particularly promising in cancer treatment. It is noticed that the working mechanisms of natural holdase and foldase chaperone are desirable to overcome the limitations of therapeutic antibodies. Holdase chaperone stabilizes unfolded client and prevents it from activation and degradation, while foldase chaperone assists unfolded client to its native state to function. Here a holdase/foldase mimetic nanochaperone (H/F-nChap) to co-delivery two types of monoclonal antibodies (mAbs), αCD16 and αPDL1, and resiquimod (R848) is developed, which significantly improves cancer immunotherapy. The H/F-nChap presents holdase activity in blood and normal tissues that hides and protects mAbs from unnecessary targeted activation and degradation, thereby prolonging blood circulation and reducing immunotoxicity in vivo. Furthermore, H/F-nChap switches to foldase activity in the tumor microenvironment that exposes mAbs and releases R848 to enhance the engagement between NK cells and tumor cells and promote immune activation, respectively. The H/F-nChap represents a strategy for safe and spatiotemporal delivery of multiple mAbs, providing a promising platform for improved cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal , Neoplasms , Humans , Antibodies, Monoclonal/therapeutic use , Immunotherapy , Neoplasms/therapy
8.
Mater Horiz ; 10(2): 361-392, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36541078

ABSTRACT

Therapeutic cancer vaccines, which are designed to amplify tumor-specific T cell responses, have been envisioned as one of the most powerful tools for effective cancer immunotherapy. However, increasing the potency, quality and durability of the vaccine response remains a big challenge. In recent years, materials-based delivery systems focusing on the co-delivery of antigens and adjuvants to enhance cancer vaccination therapy have attracted increasing interest. Among various materials, polymeric nanoparticles (NPs) with different physicochemical properties which can incorporate multiple immunological cues are of great interest. In this review, the recent progress in the design and construction of both ex vivo subunit and in situ cancer vaccines using polymeric NPs is summarized. Especially, we will focus on how these NPs improve the adjuvanticity of vaccines. The design principles of polymeric NPs for ex vivo subunit cancer vaccines and in situ cancer vaccination are also discussed. Finally, we want to briefly discuss molecular chaperones in cancer immunity and the applications of our unique self-assembly mixed shell polymeric micelle-based nanochaperones for cancer vaccines.


Subject(s)
Cancer Vaccines , Nanoparticles , Neoplasms , Humans , Cancer Vaccines/therapeutic use , Immunotherapy , Vaccination , Vaccines, Subunit , Nanoparticles/chemistry , Neoplasms/therapy
9.
Small ; 18(32): e2203100, 2022 08.
Article in English | MEDLINE | ID: mdl-35843873

ABSTRACT

Personalized cancer vaccination using nanomaterials holds great potential for cancer immunotherapy. Here, a nanochaperone (PBA-nChap) is tailored for in situ capture of tumor-associated antigens (TAAs) to improve cancer immunotherapy. The PBA-nChap is capable of i) efficiently capturing TAAs in situ; ii) protecting TAAs from degradation; iii) transporting TAAs to antigen-presenting cells and promoting cross-presentation. Intratumor injection of PBA-nChap in combination with pretreatment with photodynamic therapy (PDT) significantly enhances immune response and exhibits excellent antitumor efficacy. Moreover, nanovaccine prepared by simply co-culturing PBA-nChap with tumor cell fragments from surgery resected primary tumor in vitro synergized with immune checkpoint blockade (ICB) therapy can effectively inhibit tumor recurrence and metastasis after an operation. This work provides a promising platform for personalized cancer vaccination.


Subject(s)
Cancer Vaccines , Neoplasms , Photochemotherapy , Animals , Antigens, Neoplasm , Humans , Immunotherapy , Mice , Mice, Inbred C57BL , Neoplasms/therapy
10.
Angew Chem Int Ed Engl ; 61(19): e202200192, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35229425

ABSTRACT

Protein misassembly leads to the formation of dysfunctional and toxic molecular species relating to neurodegeneration in Parkinson's disease and Alzheimer's disease. Here, we tailored a nanochaperone (αS-nChap) for α-synuclein to regulate its assembly. The αS-nChap is capable of i) specifically recognizing α-synuclein; ii) dynamically capturing and stabilizing monomeric α-synuclein and retarding oligomerization; iii) tightly capturing oligomeric α-synuclein to prevent fibrillization; and iv) transporting α-synuclein oligomers to the lysosomal degradation system. The regulation of α-synuclein assembly by αS-nChap was studied in vitro. Moreover, the role of αS-nChap preventing α-synuclein pathology in cells and protecting neurons from apoptosis was investigated. The strategy of tailoring a nanochaperone to regulate aberrant assembly of pathogenic proteins provides important insights into protein misfolding diseases. We foresee that αS-nChap has therapeutic value for Parkinson's disease.


Subject(s)
Alzheimer Disease , Parkinson Disease , Alzheimer Disease/metabolism , Humans , Neurons/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism
11.
ChemMedChem ; 17(6): e202100678, 2022 03 18.
Article in English | MEDLINE | ID: mdl-34890114

ABSTRACT

Many intracellular signaling events remain poorly characterized due to a general lack of tools to interfere with "undruggable" targets. Antibodies have the potential to elucidate intracellular mechanisms via targeted disruption of cell signaling cascades because of their ability to bind to a target with high specificity and affinity. However, due to their size and chemical composition, antibodies cannot innately cross the cell membrane, and thus access to the cytosol with these macromolecules has been limited. Herein we describe strategies for accessing the intracellular space with recombinant antibodies mediated by cationic lipid nanoparticles to selectively disrupt intracellular signaling events. Together, our results demonstrate the use of recombinantly produced antibodies, delivered at concentrations of 10 nM, to selectively interfere with signaling driven by a single posttranslational modification. Efficient intracellular delivery of engineered antibodies opens up possibilities for modulation of previously "undruggable" targets, including for potential therapeutic applications.


Subject(s)
Nanoparticles , Antibodies , Liposomes , Nanoparticles/chemistry , Signal Transduction
12.
Angew Chem Int Ed Engl ; 60(19): 10865-10870, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33595165

ABSTRACT

Nanochaperones have been designed and used for regulating the (re)folding of proteins, treating protein misfolding-related diseases, and, more recently, in drug delivery. Despite various successes, a complete understanding of the working mechanisms remains elusive, which represents a challenge for the realization of their full potential. Here, we thoroughly investigated the functioning of differently charged nanochaperones that regulate the refolding of thermally denatured lysozyme. We found that the balance between the capture and release of lysozyme clients that are controlled by nanochaperones plays a key role in regulating refolding. More importantly, the findings could be applied to other enzymes with various physicochemical properties. On the basis of these results, we could recover the activity of enzymes with high efficiency either after 20 days of storage at 40 °C or heating at high temperatures for 30-60 min. Our results provide important new design strategies for nanochaperone systems to improve their properties and expand their applications.


Subject(s)
Molecular Chaperones/chemistry , Muramidase/chemistry , Temperature , Muramidase/metabolism , Particle Size , Protein Denaturation , Protein Folding
13.
J Control Release ; 330: 1244-1249, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33234362

ABSTRACT

Proteolysis-targeting chimaera (PROTAC) technology is an emerging approach for achieving targeted degradation of a protein of interest (POI) intracellularly. However, the cell permeability of PROTACs is limited by their high molecular weight and total polar surface area. Moreover, the activation of the proteasome-mediated degradation by PROTAC requires the formation of a ternary (three-component) complex, composed of the PROTAC, the POIs, and E3-ligases related proteins (E3Ps). Simplifying the three-component system to two-component system could theoretically increase the efficiency of the formation of ternary complex and enhance the protein degradation efficiency. Herein, we demonstrate that pre-fusion of PROTACs with E3Ps (called "pre-fused PROTACs") before administration could transform the original PROTAC system to two-component system. After delivery by lipid nanoparticles, the degradation of POI by pre-fused PROTACs was dramatically increased and accelerated compared with standard PROTACs. Moreover, we demonstrated that this approach could be generalized to another hydrophobic tag (HyT) degrader by demonstrating the improved targeted protein degradation after pre-fusion the HyT degrader with heat shock protein 70 (HSP70).


Subject(s)
Lipids , Nanoparticles , Proteolysis
14.
Sci Adv ; 6(30): eabb4429, 2020 07.
Article in English | MEDLINE | ID: mdl-32832671

ABSTRACT

Safe and efficient delivery of blood-brain barrier (BBB)-impermeable cargos into the brain through intravenous injection remains a challenge. Here, we developed a previously unknown class of neurotransmitter-derived lipidoids (NT-lipidoids) as simple and effective carriers for enhanced brain delivery of several BBB-impermeable cargos. Doping the NT-lipidoids into BBB-impermeable lipid nanoparticles (LNPs) gave the LNPs the ability to cross the BBB. Using this brain delivery platform, we successfully delivered amphotericin B (AmB), antisense oligonucleotides (ASOs) against tau, and genome-editing fusion protein (-27)GFP-Cre recombinase into the mouse brain via systemic intravenous administration. We demonstrated that the NT-lipidoid formulation not only facilitates cargo crossing of the BBB, but also delivery of the cargo into neuronal cells for functional gene silencing or gene recombination. This class of brain delivery lipid formulations holds great potential in the treatment of central nervous system diseases or as a tool to study the brain function.


Subject(s)
Lipids , Nanoparticles , Animals , Brain , Injections, Intravenous , Liposomes , Mice , Neurotransmitter Agents
15.
Mol Ther Nucleic Acids ; 19: 1357-1367, 2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32160706

ABSTRACT

The efficient delivery of antisense oligonucleotides (ASOs) to the targeted cells and organs remains a challenge, in particular, in vivo. Here, we investigated the ability of a library of biodegradable lipid nanoparticles (LNPs) in delivering ASO to both cultured human cells and animal models. We first identified three top-performing lipids through in vitro screening using GFP-expressing HEK293 cells. Next, we explored these three candidates for delivering ASO to target proprotein convertase subtilisin/kexin type 9 (PCSK9) mRNA in mice. We found that lipid 306-O12B-3 showed efficiency with the median effective dose (ED50) as low as 0.034 mg·kg-1, which is a notable improvement over the efficiency reported in the literature. No liver or kidney toxicity was observed with a dose up to 5 mg·kg-1 of this ASO/LNP formulation. The biodegradable LNPs are efficient and safe in the delivery of ASO and pave the way for clinical translation.

16.
ACS Biomater Sci Eng ; 6(2): 1064-1073, 2020 02 10.
Article in English | MEDLINE | ID: mdl-33464849

ABSTRACT

Invasive fungal infections are well-known causes of morbidity and mortality in immunocompromised patients. Amphotericin B (AmB) is a polyene fungicidal agent with excellent properties of the broad antifungal spectrum, high activity, and relatively rare drug resistance. However, significant toxicities limit the clinical application of AmB and its conventional formulation AmB deoxycholate (Fungizone). Here we investigated nanoparticle formulations of AmB using synthetic biodegradable lipidoids and evaluated their stability, in vitro antifungal efficacy, and in vivo toxicity and pharmacokinetics. We found that the AmB formulated using a mixture of quaternized lipidoid (Q78-O14B) and DSPE-PEG2000 has the size around 70-100 nm and is stable during storage. The formulation showed no hemotoxicity to red blood cells (RBCs) in vitro. It also possesses the highest antifungal activity (in vitro) and lowest toxicity (both in vitro and in vivo). These metrics are significantly superior to the commercial antifungal product Fungizone. Meanwhile, AmB/Q78-O14B-P exhibited prolonged blood circulation in comparison to Fungizone in vivo. In AmB/Q78-O14B-P formulation, AmB was still detectable in the liver, spleen, and lung tissues with a concentration above the minimum inhibitory concentrations 72 h after low-dose intravenous injection. Based on these results, AmB in lipidoid nanoparticle formulation may produce sustained antifungal activity against blood-borne and systemic organ infections. Moreover, the new AmB formulation showed low nephrotoxicity and hepatotoxicity in rats even at high doses, allowing a dramatically wider and safer therapeutic window than Fungizone. This method provides a means to develop much needed antifungal agents that will be more therapeutically efficacious, more affordable (than AmBisome), and less toxic (than Fungizone) for the treatment of systemic fungal infections.


Subject(s)
Mycoses , Nanoparticles , Amphotericin B/pharmacology , Animals , Antifungal Agents , Humans , Microbial Sensitivity Tests , Rats
17.
Adv Mater ; 32(3): e1805945, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31045287

ABSTRACT

Folding and unfolding are essential ways for a protein to regulate its biological activity. The misfolding of proteins usually reduces or completely compromises their biological functions, which eventually causes a wide range of diseases including neurodegeneration diseases, type II diabetes, and cancers. Therefore, materials that can regulate protein folding and maintain proteostasis are of significant biological and medical importance. In living organisms, molecular chaperones are a family of proteins that maintain proteostasis by interacting with, stabilizing, and repairing various non-native proteins. In the past few decades, efforts have been made to create artificial systems to mimic the structure and biological functions of nature chaperonins. Herein, recent progress in the design and construction of materials that mimic different kinds of natural molecular chaperones is summarized. The fabrication methods, construction rules, and working mechanisms of these artificial chaperone systems are described. The application of these materials in enhancing the thermal stability of proteins, assisting de novo folding of proteins, and preventing formation of toxic protein aggregates is also highlighted and explored. Finally, the challenges and potential in the field of chaperone-mimetic materials are discussed.


Subject(s)
Biocompatible Materials/chemistry , Molecular Chaperones/metabolism , Proteins/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Biocompatible Materials/metabolism , Humans , Hydrogels/chemistry , Micelles , Molecular Chaperones/chemistry , Nanoparticles/chemistry , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Folding , Proteins/chemistry
18.
Nano Lett ; 19(2): 674-683, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30444372

ABSTRACT

Alzheimer's disease (AD) is a progressive and irreversible brain disorder. Recent studies revealed the pivotal role of ß-amyloid (Aß) in AD. However, there is no conclusive indication that the existing therapeutic strategies exerted any effect on the mitigation of Aß-induced neurotoxicity and the elimination of Aß aggregates simultaneously in vivo. Herein, we developed a novel nanocomposite that can eliminate toxic Aß aggregates and mitigate Aß-induced neurotoxicity in AD mice. This nanocomposite was designed to be a small-sized particle (14 ± 4 nm) with Aß-binding peptides (KLVFF) integrated on the surface. The nanocomposite was prepared by wrapping a protein molecule with a cross-linked KLVFF-containing polymer layer synthesized by in situ polymerization. The presence of the nanocomposite remarkably changed the morphology of Aß aggregates, which led to the formation of Aß/nanocomposite coassembled nanoclusters instead of Aß oligomers. With the reduction of the pathological Aß oligomers, the nanocomposites attenuated the Aß-induced neuron damages, regained endocranial microglia's capability to phagocytose Aß, and eventually protected hippocampal neurons against apoptosis. Thus, we anticipate that the small-sized nanocomposite will potentially offer a feasible strategy in the development of novel AD treatments.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Nanocomposites/therapeutic use , Nanomedicine/methods , Peptides/therapeutic use , Protein Aggregation, Pathological/therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid beta-Peptides/isolation & purification , Animals , Disease Models, Animal , Mice , Models, Molecular , Nanocomposites/chemistry , Nanocomposites/ultrastructure , Peptides/chemistry , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology
19.
ACS Nano ; 11(10): 10549-10557, 2017 10 24.
Article in English | MEDLINE | ID: mdl-28968070

ABSTRACT

The folding process of a protein is inherently error-prone, owing to the large number of possible conformations that a protein chain can adopt. Partially folded or misfolded proteins typically expose hydrophobic surfaces and tend to form dysfunctional protein aggregates. Therefore, materials that can stabilize unfolded proteins and then efficiently assist them refolding to its bioactive form are of significant interest. Inspired by natural chaperonins, we have synthesized a series of polymeric nanochaperones that can facilitate the refolding of denatured proteins with a high recovery efficiency (up to 97%). Such nanochaperones possess phase-separated structure with hydrophobic microdomains on the surface. This structure allows nanochaperones to stabilize denatured proteins by binding them to the hydrophobic microdomains. We have also investigated the mechanism by which nanochaperones assist the protein refolding and established the design principles of nanochaperones in order to achieve effective recovery of a certain protein from their denatured forms. With a carefully designed composition of the microdomains according to the surface properties of the client proteins, the binding affinity between the hydrophobic microdomain and the denatured protein molecules can be tuned precisely, which enables the self-sorting of the polypeptides and the refolding of the proteins into their bioactive states. This work provides a feasible and effective strategy to recover inclusion bodies to their bioactive forms, which has potential to reduce the cost of the manufacture of recombinant proteins significantly.


Subject(s)
Molecular Chaperones/chemistry , Muramidase/chemistry , Nanoparticles/chemistry , Protein Refolding , Molecular Chaperones/chemical synthesis , Molecular Chaperones/metabolism , Muramidase/chemical synthesis , Muramidase/metabolism , Particle Size , Protein Denaturation , Surface Properties , Temperature
20.
Chemistry ; 23(4): 926-934, 2017 Jan 18.
Article in English | MEDLINE | ID: mdl-27809357

ABSTRACT

Quantitative analysis of multisite interactions between a protein and its binding partner at atomic resolution is complicated because locating the binding sites is difficult and differentiating the flexibility of each binding site is even more elusive. Introduction of a paramagnetic metal center close to the binding pocket greatly attenuates the signals in the NMR spectrum upon binding. Herein, the multisite binding of hen egg white lysozyme (HEWL) with lanthanide complexes [Ln(DPA)3 ]3- (DPA=dipicolinic acid) was analyzed with sensitive paramagnetic NMR spectroscopy. Paramagnetic relaxation enhancement (PRE) revealed that HEWL interacts with [Ln(DPA)3 ]3- at four major binding sites in aqueous solution, which is in contrast to a previous X-ray structural analysis. The varied binding affinities for the ligands and different flexibilities at each binding site were in good agreement with atomistic molecular dynamics (MD) simulations. The present work demonstrates that a combination of paramagnetic NMR spectroscopy and MD simulations is a powerful tool to delineate the multisite interactions of a protein with its binding partner at atomic resolution, in terms of both affinity and flexibility.


Subject(s)
Muramidase/chemistry , Animals , Binding Sites , Chickens , Coordination Complexes/chemistry , Coordination Complexes/metabolism , Lanthanoid Series Elements/chemistry , Ligands , Magnetics , Molecular Dynamics Simulation , Muramidase/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Tertiary , Water/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL