Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
2.
Adv Healthc Mater ; 13(2): e2301808, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37602504

ABSTRACT

Implantable biomaterials are widely used in bone tissue engineering, but little is still known about how they initiate early immune recognition and the initial dynamics. Herein, the early immune recognition and subsequent osteoinduction of biphasic calcium phosphate (BCP) after implantation to the protein adsorption behavior is attributed. By liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis, the biomaterial-related molecular patterns (BAMPs) formed after BCP implantation are mapped, dominated by the highly expressed extracellular matrix protein fibronectin (Fn) and the high mobility group box 1 (HMGB1). Molecular dynamics simulations show that Fn has the ability to bind more readily to the BCP surface than HMGB1. The preferential binding of Fn provides a higher adsorption energy for HMGB1. Furthermore, multiple hydrogen bonding sites between HMGB1 and Fn are demonstrated using a molecular docking approach. Ultimately, the formation of BAMPs through HMGB1 antagonist glycyrrhizic acid (GA), resulting in impaired immune recognition of myeloid differentiation factor 88 (MYD88) mediated dendritic cells (DCs) and macrophages (Mφs), as well as failed osteoinduction processes is obstructed. This study introduces a mechanism for early immune recognition of implant materials based on protein adsorption, providing perspectives for future design and application of tissue engineering materials.


Subject(s)
Biocompatible Materials , HMGB1 Protein , Hydroxyapatites , Biocompatible Materials/chemistry , Fibronectins/chemistry , Adsorption , Chromatography, Liquid , Molecular Docking Simulation , Tandem Mass Spectrometry
3.
Article in English | MEDLINE | ID: mdl-37877885

ABSTRACT

Sonodynamic therapy (SDT) has considerable potential in cancer treatment and exhibits high tissue penetration with minimal damage to healthy tissues. The efficiency of SDT is constrained by the complex immunological environment and tumor treatment resistance. Herein, a specific acoustic-actuated tumor-targeted nanomachine is proposed to generate mechanical damage to lysosomes for cancer SDT. The hybrid nanomachine was assembled with gold nanoparticles (GNPs) as the core and encapsulated with macrophage exosomes modified by AS1411 aptamers (GNP@EXO-APs) to optimize the pharmacokinetics and tumor aggregation. GNP@EXO-APs could be specifically transferred to the lysosomes of tumor cells. After induction with ultrasound, GNP@EXO-APs generated strong mechanical stress to produce lysosomal-dependent cell death in cancer cells. Notably, tumor-associated macrophages were reprogrammed in the ultrasound environment to an antitumor phenotype. Enhanced mechanical destruction via GNP@EXO-APs and immunotherapy of cancer cells were verified both in vitro and in vivo under SDT. This study provides a new direction for inside-out killing effects on tumor cells for cancer treatment.

4.
Sci Adv ; 9(38): eadg0300, 2023 Sep 22.
Article in English | MEDLINE | ID: mdl-37729409

ABSTRACT

Femtosecond laser-based technique called two-photon polymerization (TPP) has emerged as a powerful tool for nanofabrication and integrating nanomaterials. However, challenges persist in existing three-dimensional (3D) nanoprinting methods, such as slow layer-by-layer printing and limited material options due to laser-matter interactions. Here, we present an approach to 3D nanoprinting called free-space nanopainting, using an optical force brush (OFB). OFB enables precise spatial writing paths, instantaneous adjustment of linewidths and concentrations, and unrestricted resolution beyond optical limits. OFB allows rapid aggregation and solidification of radicals, resulting in narrower lines at lower polymerization thresholds and enhanced sensitivity to laser energy. This advancement enables high-accuracy free-space painting, analogous to Chinese brush painting on paper. The printing speed is increased substantially compared to layer-by-layer methods, from 100 to 1000 times faster. We successfully printed various bionic muscle models derived from 4D nanostructures with tunable mechanical properties, responsive to electrical signals, and excellent biocompatibility.

5.
Proc Natl Acad Sci U S A ; 120(3): e2213837120, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36626552

ABSTRACT

Implants are widely used in medical applications and yet macrophage-mediated foreign body reactions caused by implants severely impact their therapeutic effects. Although the extensive use of multiple surface modifications has been introduced to provide some mitigation of fibrosis, little is known about how macrophages recognize the stiffness of the implant and thus influence cell behaviors. Here, we demonstrated that macrophage stiffness sensing leads to differential inflammatory activation, resulting in different degrees of fibrosis. The potential mechanism for macrophage stiffness sensing in the early adhesion stages tends to involve cell membrane deformations on substrates with different stiffnesses. Combining theory and experiments, we show that macrophages exert traction stress on the substrate through adhesion and altered membrane curvature, leading to the uneven distribution of the curvature-sensing protein Baiap2, resulting in cytoskeleton remodeling and inflammation inhibition. This study introduces a physical model feedback mechanism for early cellular stiffness sensing based on cell membrane deformation, offering perspectives for future material design and targeted therapies.


Subject(s)
Foreign-Body Reaction , Macrophages , Humans , Macrophages/metabolism , Foreign-Body Reaction/metabolism , Foreign-Body Reaction/pathology , Inflammation/metabolism , Cell Membrane , Fibrosis
6.
J Transl Med ; 20(1): 574, 2022 12 08.
Article in English | MEDLINE | ID: mdl-36482390

ABSTRACT

BACKGROUND: This study aimed to develop a radiogenomic prognostic prediction model for colorectal cancer (CRC) by investigating the biological and clinical relevance of intratumoural heterogeneity. METHODS: This retrospective multi-cohort study was conducted in three steps. First, we identified genomic subclones using unsupervised deconvolution analysis. Second, we established radiogenomic signatures to link radiomic features with prognostic subclone compositions in an independent radiogenomic dataset containing matched imaging and gene expression data. Finally, the prognostic value of the identified radiogenomic signatures was validated using two testing datasets containing imaging and survival information collected from separate medical centres. RESULTS: This multi-institutional retrospective study included 1601 patients (714 females and 887 males; mean age, 65 years ± 14 [standard deviation]) with CRC from 5 datasets. Molecular heterogeneity was identified using unsupervised deconvolution analysis of gene expression data. The relative prevalence of the two subclones associated with cell cycle and extracellular matrix pathways identified patients with significantly different survival outcomes. A radiogenomic signature-based predictive model significantly stratified patients into high- and low-risk groups with disparate disease-free survival (HR = 1.74, P = 0.003). Radiogenomic signatures were revealed as an independent predictive factor for CRC by multivariable analysis (HR = 1.59, 95% CI:1.03-2.45, P = 0.034). Functional analysis demonstrated that the 11 radiogenomic signatures were predominantly associated with extracellular matrix and immune-related pathways. CONCLUSIONS: The identified radiogenomic signatures might be a surrogate for genomic signatures and could complement the current prognostic strategies.


Subject(s)
Colorectal Neoplasms , Genomics , Humans , Aged , Retrospective Studies , Cohort Studies , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/genetics , Tomography, X-Ray Computed
7.
Front Genet ; 13: 880093, 2022.
Article in English | MEDLINE | ID: mdl-35646105

ABSTRACT

Background: Preoperative and postoperative evaluation of colorectal cancer (CRC) patients is crucial for subsequent treatment guidance. Our study aims to provide a timely and rapid assessment of the prognosis of CRC patients with deep learning according to non-invasive preoperative computed tomography (CT) and explore the underlying biological explanations. Methods: A total of 808 CRC patients with preoperative CT (development cohort: n = 426, validation cohort: n = 382) were enrolled in our study. We proposed a novel end-to-end Multi-Size Convolutional Neural Network (MSCNN) to predict the risk of CRC recurrence with CT images (CT signature). The prognostic performance of CT signature was evaluated by Kaplan-Meier curve. An integrated nomogram was constructed to improve the clinical utility of CT signature by combining with other clinicopathologic factors. Further visualization and correlation analysis for CT deep features with paired gene expression profiles were performed to reveal the molecular characteristics of CRC tumors learned by MSCNN in radiographic imaging. Results: The Kaplan-Meier analysis showed that CT signature was a significant prognostic factor for CRC disease-free survival (DFS) prediction [development cohort: hazard ratio (HR): 50.7, 95% CI: 28.4-90.6, p < 0.001; validation cohort: HR: 2.04, 95% CI: 1.44-2.89, p < 0.001]. Multivariable analysis confirmed the independence prognostic value of CT signature (development cohort: HR: 30.7, 95% CI: 19.8-69.3, p < 0.001; validation cohort: HR: 1.83, 95% CI: 1.19-2.83, p = 0.006). Dimension reduction and visualization of CT deep features demonstrated a high correlation with the prognosis of CRC patients. Functional pathway analysis further indicated that CRC patients with high CT signature presented down-regulation of several immunology pathways. Correlation analysis found that CT deep features were mainly associated with activation of metabolic and proliferative pathways. Conclusions: Our deep learning based preoperative CT signature can effectively predict prognosis of CRC patients. Integration analysis of multi-omic data revealed that some molecular characteristics of CRC tumor can be captured by deep learning in CT images.

8.
Adv Healthc Mater ; 11(13): e2200382, 2022 07.
Article in English | MEDLINE | ID: mdl-35543500

ABSTRACT

The entry of implants triggers the secretion of damage associated molecular patterns (DAMPs) that recruit dendritic cells (DCs) and results in subsequent foreign body reaction (FBR). Though several studies have illustrated that the surface accessible area (SAA) of implants plays a key role in the process of DAMPs release and absorption, the effect of SAA on the immune reaction still remains unknown. Here, a series of TiO2 plates with different SAA is fabricated to investigate the relationship between SAA and FBR. Compared with larger SAA surface, the aggregation of DC is significantly inhibited by lower SAA surface. Total internal reflection microscopy (TIRFM) and molecular dynamic (MD) simulation show that although high mobility group box 1 (HMGB1) is adsorbed more on plates with lower SAA, the exposure ratio of cysteine (CYS) residue in HMGB1 is significantly decreased in lower SAA group. The lower exposure of CYS reduces the activation of Toll-like receptors 4 (TLR4), which down-regulates the expression of myeloid differentiation factor (Myd88)-TNF receptor associated factor 6 (TRAF6) to inhibit nuclear factor kappa B (NF-κB) signaling. Generally, this study reveals the mechanism of how SAA, a nanoscale property, affects FBR from perspective of DAMPs, and provides a new direction for designing better biocompatible implants.


Subject(s)
HMGB1 Protein , Foreign-Body Reaction , HMGB1 Protein/metabolism , Humans , NF-kappa B/metabolism , Signal Transduction , Titanium
9.
Adv Healthc Mater ; 11(10): e2101983, 2022 05.
Article in English | MEDLINE | ID: mdl-35104391

ABSTRACT

Mechanistic understanding of fibronectin (FN) adsorption which determines cell adhesion on cell-implant interfaces is significant for improving the osteoconduction and soft-tissue healing of implants. Here, it is shown that the adsorption behavior of FN on the titanium oxide surface (TiO2 ) is highly relative to its Pro-His-Ser-Arg-Asn (PHSRN) peptide. FN lacking PHSRN fails to bind to surfaces, resulting in inhibited cell adhesion and spreading. Molecular dynamics simulation shows higher affinity and greater adsorption energy of PHSRN peptide with TiO2 surface due to the stronger hydrogen bonds formed by the serine and arginine residues with O ion of the substrate. Finally, by increasing O content in TiO2 surfaces through O ion-beam implantation, improving the cell adhesion, cell differentiation, and the subsequent biomineralization on titanium implant is realized. This study reveals the vital role of PHSRN in FN-mediated cell adhesion on implant surfaces, providing a promising new target for further tissue integration and implant success.


Subject(s)
Fibronectins , Titanium , Cell Adhesion , Fibronectins/chemistry , Oxygen , Peptides/chemistry , Surface Properties , Titanium/chemistry , Titanium/pharmacology
10.
Adv Healthc Mater ; 10(20): e2100994, 2021 10.
Article in English | MEDLINE | ID: mdl-34196125

ABSTRACT

Macrophage activation determines the fate of biomaterials implantation. Though researches have shown that fibronectin (FN) is highly involved in integrin-induced macrophage activation on biomaterials, the mechanism of how nanosized structure affects macrophage behavior is still unknown. Here, titanium dioxide nanotube structures with different sizes are fabricated to investigate the effects of nanostructure on macrophage activation. Compared with larger sized nanotubes and smooth surface, 30 nm nanotubes exhibit considerable lesser pro-inflammatory properties on macrophage differentiation. Confocal protein observation and molecular dynamics simulation show that FN displays conformation changes on different nanotubes in a feature of "size-confined," which causes the hiding of Arg-Gly-Asp (RGD) domain on other surfaces. The matching size of nanotube with FN allows the maximum exposure of RGD on 30 nm nanotubes, activating integrin-mediated focal adhesion kinase (FAK)-phosphatidylinositol-3 kinase γ (PI3Kγ) pathway to inhibit nuclear factor kappa B (NF-κB) signaling. In conclusion, this study explains the mechanism of nanostructural-biological signaling transduction in protein and molecular levels, as well as proposes a promising strategy for surface modification to regulate immune responses on bioimplants.


Subject(s)
Fibronectins , Nanostructures , Cell Adhesion , Humans , Inflammation , Macrophages , Titanium
11.
Theranostics ; 11(5): 2349-2363, 2021.
Article in English | MEDLINE | ID: mdl-33500729

ABSTRACT

The cell membrane-coated nanoparticles (MNPs) showed great potential in treating infectious disease due to their superior biofunctions in improving biocompatibility of nanoparticles and neutralization of pathogen or toxins. However, bone infection is accompanied with severe inflammation and bone loss, which also requires anti-inflammatory and osteoconductive treatment. The conventional membrane coating method has to undergo ultrasonication and extrusion procedures, which reduces the functionality of cell membrane and limits the choice of nanoparticles. In this study, we proposed an electroporation-based membrane coating strategy to facilitate the synthesis of MNPs to tackle those problems. Methods: Magnetic composite nanoparticles with osteoconductive Ca3(PO4)2 and bactericidal TiO2 were assembled into macrophages through phagocytosis and then collected to expose in electric field for obtaining macrophage membrane-coating nanoparticles. By using molecular dynamics simulation and materials characterizations, the cell membrane coating efficiency was confirmed. The in vitro anti-bacterial and anti-inflammatory abilities were tested by bacteria culturing and immune cells activation. Then drug-resistant bacteria induced bone infection model was established to verify its in vivo therapeutic effects. Results: The coated membrane prepared through electroporation reserved the integrality of membrane structure and right-sidedness, with more functional proteins. Those led to the superior properties of recognition and adsorption with bacteria, toxins and inflammatory cytokines. Owing to the benefits of electroporation, the MNPs exhibited significant better antibacterial and anti-inflammatory abilities for enhancing the tissue repair process. Conclusion: This study provides a novel self-assembly cell membrane coating strategy by electroporation to construct multifunctional membrane-coating nanoparticles for bone infection treatment. This strategy not only improves the functions of coated membrane, but is also proved to be universal for varies nanoparticles or cells, indicating a great potential for future applications in the bioengineering field.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cell Membrane/chemistry , Coated Materials, Biocompatible/pharmacology , Electroporation/methods , Nanoparticles/administration & dosage , Osteomyelitis/prevention & control , Staphylococcal Infections/drug therapy , Animals , Female , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Mice , Nanoparticles/chemistry , Osteomyelitis/immunology , Osteomyelitis/microbiology , Staphylococcal Infections/complications , Staphylococcal Infections/microbiology
12.
Adv Healthc Mater ; 8(21): e1901030, 2019 11.
Article in English | MEDLINE | ID: mdl-31583846

ABSTRACT

Magnesium (Mg2+ ), as a main component of bone, is widely applied to promote bone growth and regeneration. However, Mg2+ can chemically inhibit the crystallization of amorphous calcium phosphate into hydroxyapatite (HA). The underlying mechanisms by which Mg2+ improves bone biomineralization remain elusive. Here, it is demonstrated that Mg2+ plays dual roles in bone biomineralization from a developmental perspective. During embryonic development, the Mg2+ concentration is enriched in the early stage from embryonic day 13.5 (E13.5) to E15.5, but gradually decreases to a stable state in the late phase, after E15.5. Appropriate concentrations of Mg2+ can promote the mineralization of bone marrow mesenchymal stem cells, while excessive Mg2+ impairs their osteogenesis. The earlier the Mg2+ is added, the stronger the observed inhibition of mineralization. In particular, less Mg2+ is present in fully mineralized collagen than in poorly mineralized collagen. Furthermore, a high concentration of Mg2+ changes the crystalline morphology of HA and inhibits collagen calcification. Functionally, a high-Mg2+ diet inhibits bone biomineralization in mouse offspring. Taken together, the results suggest that appropriate regulation of Mg2+ concentration over time is vital for normal biomineralization. This study is significant for the future design of bone substitutes and implants associated with Mg2+ content.


Subject(s)
Bone Regeneration/physiology , Bone and Bones/metabolism , Magnesium/metabolism , Animals , Biomineralization , Bone and Bones/ultrastructure , Calcification, Physiologic/physiology , Female , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Electron, Scanning , Osteogenesis/physiology , Stress, Mechanical
13.
Clin Implant Dent Relat Res ; 21(5): 977-986, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31373150

ABSTRACT

BACKGROUND: In the past few decades, very little research has been carried out to modify implant surfaces to improve osteointegration through the regulation of immune cells. PURPOSE: The aim of this study is to investigate whether the poly(dopamine) (pDA)-assisted immobilization of IL4 on titanium surfaces could modulate the inflammatory profile of macrophages in vitro and search for the possibility of enhancing implant integration in this way. MATERIAL AND METHODS: The surface composition, topography, and roughness of SLA, SLA-pDA, and SLA-pDA-IL4 discs were examined by scanning electron microscope (SEM) and energy dispersive spectrometer (EDS). Then the releasing profile of the SLA-pDA-IL4 implants was recorded for 1 week and the bioactivity of released IL4 was investigated by ELISA. Then macrophage polarization was investigated via three methods including: (a) surface marker via immunofluorescence; (b) mRNA levels of M1 and M2 polarization markers via real-time PCR, and (c) cytokine release via ELISA. RESULTS: SEM and EDS revealed that pDA and IL4 were coated successfully on SLA surfaces. The ELISA results showed that IL4 remained its bioactivity on SLA surface and were immobilized on the SLA surface. The immobilization of IL4 through pDA has no significant influence on the attachment, morphology, and proliferation of macrophages, while it increased the M2/M1 proportion in human macrophages revealed by immunofluorescence. The real-time PCR and ELISA results demonstrated that SLA-pDA-IL4 surface reduced the pro-inflammatory profile compared with SLA-pDA and SLA surfaces. CONCLUSIONS: The SLA-pDA-IL4 surfaces described here is able to activate adherent macrophages into M2 phenotype and reduce the release of pro-inflammatory cytokines. Immobilization of IL4 via pDA is convenient and effective, thus providing an applicable way to control macrophage behavior upon implant insertion and is anticipated to accelerating further bone integration.


Subject(s)
Dental Implants , Indoles/metabolism , Interleukin-4/metabolism , Polymers/metabolism , Titanium , Dopamine , Humans , Macrophages , Surface Properties
14.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 26(3): 900-904, 2018 Jun.
Article in Chinese | MEDLINE | ID: mdl-29950240

ABSTRACT

OBJECTIVE: To study the curative effect and safety of menchymal stem cell infusion in treatment of children with refractory late-onset hemorrhagic cystitis(LOHC) after allogeneic HSCT. METHODS: Thirty cases of children with refractory LOHC after allo-HSCT in our department between December 2010 and July 2016 were analyzed retrospectively, out of 30 cases 7 received MSC treatment. The used MSC of all were four-to-five generation MSC from bone marrows of third party donors, and were infused into patients with (1.87±0.456)×106/kg MSCs once a week (1-4 times in total) until the hematuria and odynuria symptoms being improved. To observe whether unfavorable reactions occurred after MSC treatment, the patients accepted daily physical examination and regular assistant examination. The cytokine levels were also measured and dynamically detected in 2 cases before and after MSC treatment. RESULTS: In 30 children with refractory LOHC, the hematuria difficultly reached the remission after routine hydration, alkalizing and antiviral therapy, Among 25 cases who were received methylprednisolone, MTX and CTX therapy, 7 cases received MSC infusion for 1-4 times with dose of (1.87±0.456)×106/(kg·time) as a result, 7 cases of LOHC were cured. The TNF-α and IL-2R levels in 2 cases progressively decreased after MSC infusion, no occurence of fever, rash, embolism and so on were found in 7 cases received MSC infusion; the BKV detection showed that the viral load did not increase; the leukemia relapse or secondary cancer did not occure. CONCLUSION: The MSC treatment is safe and effective for refractory LOHC after allo-HSCT.


Subject(s)
Cystitis , Child , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Humans , Mesenchymal Stem Cell Transplantation , Retrospective Studies , Transplantation, Homologous , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...