Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-39007694

ABSTRACT

High levels of glutathione (GSH) are an important characteristic of malignant tumors and a significant cause of ineffective treatment and multidrug resistance. Although reactive oxygen species (ROS) therapy has been shown to induce tumor cell death, the strong clearance effect of GSH on ROS significantly reduces its therapeutic efficacy. Therefore, there is a need to develop new strategies for targeting GSH. In this study, novel carbon quantum dots derived from gentamycin (GM-CQDs) were designed and synthesized. On the basis of the results obtained, GM-CQDs contain sp2 and sp3 carbon atoms as well as nitrogen oxygen groups, which decrease the intracellular levels of GSH by downregulating SLC7A11, thereby disrupting redox balance, mediating lipid peroxidation, and inducing ferroptosis. Transcriptome analysis demonstrated that GM-CQDs downregulated the expression of molecules related to GSH metabolism while significantly increasing the expression of molecules related to ferroptosis. The in vivo results showed that the GM-CQDs exhibited excellent antitumor activity and immune activation ability. Furthermore, because of their ideal biological safety, GM-CQDs are highly promising for application as drugs targeting GSH in the treatment of malignant tumors.

2.
J Transl Med ; 22(1): 477, 2024 May 19.
Article in English | MEDLINE | ID: mdl-38764038

ABSTRACT

BACKGROUND: Oral squamous cell carcinoma (OSCC) is a malignant tumor with a poor prognosis. Traditional treatments have limited effectiveness. Regulation of the immune response represents a promising new approach for OSCC treatment. B cells are among the most abundant immune cells in OSCC. However, the role of B cells in OSCC treatment has not been fully elucidated. METHODS: Single-cell RNA sequencing analysis of 13 tissues and 8 adjacent normal tissues from OSCC patients was performed to explore differences in B-cell gene expression between OSCC tissues and normal tissues. We further investigated the relationship between differentially expressed genes and the immune response to OSCC. We utilized tissue microarray data for 146 OSCC clinical samples and RNA sequencing data of 359 OSCC samples from The Cancer Genome Atlas (TCGA) to investigate the role of T-cell leukemia 1 A (TCL1A) in OSCC prognosis. Multiplex immunohistochemistry (mIHC) was employed to investigate the spatial distribution of TCL1A in OSCC tissues. We then investigated the effect of TCL1A on B-cell proliferation and trogocytosis. Finally, lentiviral transduction was performed to induce TCL1A overexpression in B lymphoblastoid cell lines (BLCLs) to verify the function of TCL1A. RESULTS: Our findings revealed that TCL1A was predominantly expressed in B cells and was associated with a better prognosis in OSCC patients. Additionally, we found that TCL1A-expressing B cells are located at the periphery of lymphatic follicles and are associated with tertiary lymphoid structures (TLS) formation in OSCC. Mechanistically, upregulation of TCL1A promoted the trogocytosis of B cells on dendritic cells by mediating the upregulation of CR2, thereby improving antigen-presenting ability. Moreover, the upregulation of TCL1A expression promoted the proliferation of B cells. CONCLUSION: This study revealed the role of B-cell TCL1A expression in TLS formation and its effect on OSCC prognosis. These findings highlight TCL1A as a novel target for OSCC immunotherapy.


Subject(s)
B-Lymphocytes , Carcinoma, Squamous Cell , Gene Expression Regulation, Neoplastic , Mouth Neoplasms , Proto-Oncogene Proteins , Tertiary Lymphoid Structures , Humans , Prognosis , Mouth Neoplasms/pathology , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/immunology , Tertiary Lymphoid Structures/pathology , Tertiary Lymphoid Structures/immunology , Tertiary Lymphoid Structures/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Female , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Male , Middle Aged , Cell Line, Tumor , Cell Proliferation
3.
Adv Healthc Mater ; 13(16): e2400526, 2024 06.
Article in English | MEDLINE | ID: mdl-38469978

ABSTRACT

The management of diabetic wounds (DWs) continues to pose a significant challenge in the field of medicine. DWs are primarily prevented from healing due to damage to macrophage efferocytosis and fibroblast dysfunction. Consequently, a treatment strategy that involves both immunoregulation and the promotion of extracellular matrix (ECM) formation holds promise for healing DWs. Nevertheless, existing treatment methods necessitate complex interventions and are associated with increased costs, for example, the use of cytokines and cell therapy, both of which have limited effectiveness. In this study, a new type of ruthenium (IV) oxide nanoparticles (RNPs)-laden hybrid hydrogel dressing with a double network of Pluronic F127 and F68 has been developed. Notably, the hybrid hydrogel demonstrates remarkable thermosensitivity, injectability, immunoregulatory characteristics, and healing capability. RNPs in hydrogel effectively regulate both fibroblasts and macrophages in a cascade manner, stimulating fibroblast differentiation while synergistically enhancing the efferocytosis of macrophage. The immunoregulatory character of the hydrogel aids in restoring the intrinsic stability of the immune microenvironment in the wound and facilitates essential remodeling of the ECM. This hydrogel therefore offers a novel approach for treating DWs through intercellular communication.


Subject(s)
Fibroblasts , Hydrogels , Macrophages , Wound Healing , Wound Healing/drug effects , Macrophages/drug effects , Macrophages/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/cytology , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Mice , RAW 264.7 Cells , Poloxamer/chemistry , Poloxamer/pharmacology , Diabetes Mellitus, Experimental/pathology , Male , Humans , Extracellular Matrix/metabolism , Nanoparticles/chemistry
4.
J Nanobiotechnology ; 21(1): 426, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37968665

ABSTRACT

BACKGROUND: The implementation of pyroptosis exhibits significant potential as a tactic to enhance tumor immune microenvironments. Previous applications of pyroptosis inducers have encountered various limitations, such as the development of drug resistance, manifestation of toxic side effects, and a deficiency in targeting capabilities. As a result, there is a growing demand for tumor therapeutic molecules that can overcome these obstacles. Therefore, the objective of this study is to develop a multifunctional nanospheres that addresses these challenges by enabling high-precision targeting of tumor cells and inducing effective pyroptosis. RESULTS: We prepared a mannose-modified MOF called mannose-doped Fe3O4@NH2-MIL-100 (M-FNM). M-FNM could enter CAL27 cells through MR-mediated endocytosis, which caused in a significant increase in the level of intracellular ROS. This increase subsequently triggered ER stress and activated the PERK-eIF2α-ATF4-CHOP signaling pathway. CHOP then mediated the downstream cascade of Caspase-1, inducing pyroptosis. In in vivo experiments, M-FNM demonstrated excellent targeting ability and exhibited anti-tumor effects. Additionally, M-FNM reshaped the immune microenvironment by promoting the infiltration of anti-tumor immune cells, primarily T lymphocytes. CONCLUSIONS: M-FNM significantly decreased tumor growth. This novel approach to induce pyroptosis in tumor cells using M-FNM may offer new avenues for the development of effective immunotherapies against cancer.


Subject(s)
Metal-Organic Frameworks , Neoplasms , Humans , Pyroptosis , Apoptosis , Mannose , Metal-Organic Frameworks/pharmacology , Endoplasmic Reticulum Stress , eIF-2 Kinase/metabolism , eIF-2 Kinase/pharmacology , Tumor Microenvironment
5.
ACS Nano ; 17(19): 18669-18687, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37768738

ABSTRACT

Tissue engineering and regenerative medicine hold promise for improving or even restoring the function of damaged organs. Graphene-based materials (GBMs) have become a key player in biomaterials applied to tissue engineering and regenerative medicine. A series of cellular and molecular events, which affect the outcome of tissue regeneration, occur after GBMs are implanted into the body. The immunomodulatory function of GBMs is considered to be a key factor influencing tissue regeneration. This review introduces the applications of GBMs in bone, neural, skin, and cardiovascular tissue engineering, emphasizing that the immunomodulatory functions of GBMs significantly improve tissue regeneration. This review focuses on summarizing and discussing the mechanisms by which GBMs mediate the sequential regulation of the innate immune cell inflammatory response. During the process of tissue healing, multiple immune responses, such as the inflammatory response, foreign body reaction, tissue fibrosis, and biodegradation of GBMs, are interrelated and influential. We discuss the regulation of these immune responses by GBMs, as well as the immune cells and related immunomodulatory mechanisms involved. Finally, we summarize the limitations in the immunomodulatory strategies of GBMs and ideas for optimizing GBM applications in tissue engineering. This review demonstrates the significance and related mechanism of the immunomodulatory function of GBM application in tissue engineering; more importantly, it contributes insights into the design of GBMs to enhance wound healing and tissue regeneration in tissue engineering.


Subject(s)
Graphite , Tissue Engineering , Biocompatible Materials , Immunity , Immunomodulation
6.
Mater Today Bio ; 16: 100344, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35833197

ABSTRACT

Pulp injury is one of the most common clinical diseases, and severe cases are usually associated with the functional loss of the tooth, while the current clinical treatment modality is only a cavity filling procedure without the regeneration of the dentin-pulp complex, thus leading to a devitalized and brittle tooth. In this study, carbon dots (CDots) with excellent biocompatibility are prepared from ascorbic acid and polyethyleneimine via a hydrothermal method. The as-prepared CDots can enhance extracellular matrix (ECM) secretion of human dental pulp stem cells (DPSCs), giving rise to increased cell adhesion on ECM and a stronger osteogenic/odontogenic differentiation capacity of DPSCs. Further, the mechanism underlying CDots-enhanced ECM secretion is revealed by the transcriptome analysis, Western blot assay and molecular dynamics simulation, identifying that the pharmacological activities of CDots are originated from a reasonable activation of the autophagy, which is mediated by regulating phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin signaling pathway. Based on the abundant CDots-induced ECM and thereby the reinforcement of the cell-ECM adhesion, an intact dental pulp stem cell sheet can be achieved, which in return promote in vivo the efficient regeneration of dentin-pulp complex as well as blood vessels.

7.
J Colloid Interface Sci ; 601: 650-660, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34091312

ABSTRACT

Engineering a targetable nanoparticle to tumor cell is a challenge issue for clinical application. Our results demonstrated that the chemokine CXCL8 secreted by oral squamous cell carcinoma (OSCC) could act as a chemoattractant to attract dental pulp mesenchymal stem cell (DPSC), which expressed the CXCL8 binding receptor, CXCR2, to the OSCC. Therefore, to create OSCC targetable nanoparticles, we used DPSC membranes to modify nanoparticles of metal-organic framework nanoparticles (MOFs) resulting in a novel MOF@DPSCM nanoparticle. Interestingly, results from in vitro and in vivo experiments illustrated that MOF@DPSCM possessed specificity for the OSCC, and the MOF@DPSCM carried DOX (doxorubicin), MOF-DOX@DPSCM could induce CAL27 cell death in vitro and block CAL27 tumor growth in vivo. Our data suggest that this novel MOF-DOX@DPSCM nanoparticle is a potential targetable drug delivery system for the OSCC in the future clinical application.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mesenchymal Stem Cells , Metal-Organic Frameworks , Mouth Neoplasms , Nanoparticles , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Dental Pulp , Humans , Mouth Neoplasms/drug therapy , Squamous Cell Carcinoma of Head and Neck
8.
ACS Appl Mater Interfaces ; 12(45): 50287-50302, 2020 Nov 11.
Article in English | MEDLINE | ID: mdl-33121247

ABSTRACT

Nucleic acid transfer has shown significant potential in the treatment of bone damage because of its long lasting local effect and lower cost. Nonviral vectors, such as nanomaterials, with higher biocompatibility are increasedly applied in the study of bone defect repair. Carbon dots with various reactive groups on the surface not only provide a unique surface to carry therapeutic genes, but also some carbon dots have been reported to promote osteogenic differentiation. The bone regeneration effect of carbon dots in vivo, however, is rarely investigated. MiR-2861 has revealed osteogenic differentiation effects. In the current study, we created ascorbic acid-PEI carbon dots (CD), which were able to carry miR-2861, by the microwave-assisted pyrolysis method. Results demonstrated that CD had excellent fluorescence stability leading to good fluorescence imaging in vitro and in vivo. CD was efficiently internalized into bone marrow stromal cells (BMSCs) through the clathrin-mediated endocytosis pathway and distributed in the mitochondria, endoplasmic reticulum, lysosome, and nucleus. Results from alkaline phosphatase staining, alizarin red staining, and reverse transcription real-time PCR (RT-QPCR) showed that our CD indeed had osteogenic effects in vitro. Flow cytometry data indicated that CD could efficiently deliver miR-2861 into BMSCs in vitro, and CD carrying miR-2861 (CD@miR) had the strongest osteogenic effects. Analyses of hematology, serum biochemistry, and histology showed that CD and CD@miR did not have cytotoxicity and had higher biocompatibility in vivo. Most interestingly, CD and miR-2861 in the CD@miR could act synergistically to promote osteogenic differentiation in vitro and new bone regeneration in vivo remarkably. Our results clearly indicate that the osteogenic CD delivering osteogenic therapeutic gene, miR-2861, can obtain much stronger bone regeneration ability, suggesting that our CD has great potential in future clinical application.


Subject(s)
Ascorbic Acid/chemistry , Carbon/chemistry , MicroRNAs/pharmacology , Polyethyleneimine/chemistry , Quantum Dots/chemistry , Animals , Bone Regeneration/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Drug Carriers/chemistry , MicroRNAs/chemistry , MicroRNAs/genetics , Microwaves , Molecular Structure , Optical Imaging , Osteogenesis/drug effects , Particle Size , Rats , Rats, Wistar , Surface Properties
9.
Biomater Sci ; 8(10): 2840-2852, 2020 May 19.
Article in English | MEDLINE | ID: mdl-32307492

ABSTRACT

Bone defects are still an unsolved clinical issue that must be overcome. Carbon dots have shown very promising effects in biological therapy. In the current study, we explored their effects on osteogenesis. Furthermore, we revealed the mechanisms in order to develop novel therapeutic approaches to manage the bone defect. For this study, ascorbic acid carbon dots (CDs) were created by a one-step microwave-assisted method. Results showed that the CDs effectively enhanced matrix mineralization, promoted osteogenic differentiation in vitro, and promoted new bone regeneration in the skull defect model in vivo. Furthermore, our data demonstrated that the ER stress and PERK-eIF2α-ATF4 pathway were activated by the CD-induced increase in intracellular calcium. Taken together, our findings suggest that the PERK pathway plays a critical role in CD-induced osteogenic differentiation, and the CDs created herein have the potential to be used to repair bone defects in clinical practice.


Subject(s)
Activating Transcription Factor 4/metabolism , Ascorbic Acid/pharmacology , Bone Regeneration/drug effects , Carbon/pharmacology , Osteoblasts/drug effects , Protein Serine-Threonine Kinases/metabolism , Quantum Dots/chemistry , eIF-2 Kinase/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Mice , Mice, Inbred C57BL , Microwaves , Osteogenesis/drug effects , Particle Size , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...