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










Publication year range
1.
Adv Mater ; 36(21): e2312440, 2024 May.
Article in English | MEDLINE | ID: mdl-38332741

ABSTRACT

Delayed re-epithelization and weakened skin contractions are the two primary factors that hinder wound closure in large-scale acute or chronic wounds. However, effective strategies for targeting these two aspects concurrently are still lacking. Herein, an antioxidative active-shrinkage hydrogel (AHF@AS Gel) is constructed that can integratedly promote re-epithelization and skin constriction to accelerate large-scale acute and diabetic chronic wound closure. The AHF@AS Gel is encapsulated by antioxidative amino- and hydroxyl-modified C70 fullerene (AHF) and a thermosensitive active shrinkage hydrogel (AS Gel). Specifically, AHF relieves overactivated inflammation, prevents cellular apoptosis, and promotes fibroblast migration in vitro by reducing excessive reactive oxygen species (ROS). Notably, the AHF@AS Gel achieved ≈2.7-fold and ≈1.7-fold better re-epithelization in acute wounds and chronic diabetic wounds, respectively, significantly contributing to the promotion of wound closure. Using proteomic profiling and mechanistic studies, it is identified that the AHF@AS Gel efficiently promoted the transition of the inflammatory and proliferative phases to the remodeling phase. Notably, it is demonstrated that AS Gel alone activates the mechanosensitive epidermal growth factor receptor/Akt (EGFR/Akt) pathway and promotes cell proliferation. The antioxidative active shrinkage hydrogel offers a comprehensive strategy for acute wound and diabetic chronic wound closure via biochemistry regulation integrating with mechanical forces stimulation.


Subject(s)
Antioxidants , Hydrogels , Skin , Wound Healing , Hydrogels/chemistry , Antioxidants/chemistry , Antioxidants/pharmacology , Animals , Skin/metabolism , Skin/drug effects , Skin/pathology , Mice , Wound Healing/drug effects , Fullerenes/chemistry , Fullerenes/pharmacology , Reactive Oxygen Species/metabolism , ErbB Receptors/metabolism , Re-Epithelialization/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Cell Movement/drug effects , Humans , Proto-Oncogene Proteins c-akt/metabolism , Apoptosis/drug effects
2.
ACS Nano ; 18(3): 2131-2148, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38198697

ABSTRACT

Endotoxemia is a life-threatening multiple organ failure disease caused by bacterial endotoxin infection. Unfortunately, current single-target therapy strategies have failed to prevent the progression of endotoxemia. Here, we reported that alanine fullerene redox modulator (AFRM) remodeled the intestinal microenvironment for multiple targets endotoxemia mitigation by suppressing inflammatory macrophages, inhibiting macrophage pyroptosis, and repairing epithelial cell barrier integrity. Specifically, AFRM exhibited broad-spectrum and self-cascade redox regulation properties with superoxide dismutase (SOD)-like enzyme, peroxidase (POD)-like enzyme activity, and hydroxyl radical (•OH) scavenging ability. Guided by proteomics, we demonstrated that AFRM regulated macrophage redox homeostasis and down-regulated LPS/TLR4/NF-κB and MAPK/ERK signaling pathways to suppress inflammatory hyperactivation. Of note, AFRM could attenuate inflammation-induced macrophage pyroptosis via inhibiting the activation of gasdermin D (GSDMD). In addition, our results revealed that AFRM could restore extracellular matrix and cell-tight junction proteins and protect the epithelial cell barrier integrity by regulating extracellular redox homeostasis. Consequently, AFRM inhibited systemic inflammation and potentiated intestinal epithelial barrier damage repair during endotoxemia in mice. Together, our work suggested that fullerene based self-cascade redox modulator has the potential in the management of endotoxemia through synergistically remodeling the inflammation and epithelial barriers in the intestinal microenvironment.


Subject(s)
Endotoxemia , Fullerenes , Mice , Animals , Endotoxemia/chemically induced , Endotoxemia/metabolism , Intestines , NF-kappa B/metabolism , Inflammation , Oxidation-Reduction , Lipopolysaccharides/pharmacology
3.
Proc Natl Acad Sci U S A ; 120(52): e2311673120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38109541

ABSTRACT

The unbalanced immune state is the dominant feature of myocardial injury. However, the complicated pathology of cardiovascular diseases and the unique structure of cardiac tissue lead to challenges for effective immunoregulation therapy. Here, we exploited oral fullerene nanoscavenger (OFNS) to maintain intestinal redox homeostasis to resolve systemic inflammation for effectively preventing distal myocardial injury through bidirectional communication along the heart-gut immune axis. Observably, OFNS regulated redox microenvironment to repair cellular injury and reduce inflammation in vitro. Subsequently, OFNS prevented myocardial injury by regulating intestinal redox homeostasis and recovering epithelium barrier integrity in vivo. Based on the profiles of transcriptomics and proteomics, we demonstrated that OFNS balanced intestinal and systemic immune homeostasis for remote cardioprotection. Of note, we applied this principle to intervene myocardial infarction in mice and mini-pigs. These findings highlight that locally addressing intestinal redox to inhibit systemic inflammation could be a potent strategy for resolving remote tissue injury.


Subject(s)
Fullerenes , Myocardial Infarction , Swine , Mice , Animals , Fullerenes/pharmacology , Swine, Miniature , Inflammation/pathology , Myocardial Infarction/prevention & control , Homeostasis , Intestinal Mucosa
4.
Adv Sci (Weinh) ; 10(35): e2302910, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37884486

ABSTRACT

Tumor immunotherapy offers a new paradigm to treat cancer; however, the existing regimens are accompanied by the dilemma of insufficient therapeutic outcomes and off-target adverse effects. The intestinal immune system contains a bulk of immune cells, which can be important contributors to the maintenance of systemic immune homeostasis. However, manipulating intestinal immunity to achieve systemic anti-tumor immunity is extremely challenging. Here, an oral immunotherapy strategy is reported using immune-enhancing fullerenes (IEF) that can reinvigorate anti-tumor immunity via immune cell-metabolic reprogramming of intestinal immune cells. Findings show that IEF can remodel anti-inflammatory macrophages into tumor-killing macrophages by regulating the energy metabolism pathway from oxidative phosphorylation (OXPHOS) to glycolysis. Consequently, IEF can reprogram the immunosuppressive intestinal immunity and enhance sys temic immunity in vivo, thereby boosting anti-tumor immunity and converting "cold" tumors into "hot" tumors. Oral immunotherapy strategy, modulating autoimmune cells in the intestine and achieving systemic anti-tumor immunity, can ensure safe and efficient tumor immunotherapy.


Subject(s)
Neoplasms , Humans , Immunotherapy , Immunosuppression Therapy , Neoplasms/drug therapy , Intestines
5.
Natl Sci Rev ; 10(1): nwac167, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36684514

ABSTRACT

Ferroptosis, an iron-dependent regulated cell death process driven by excessive lipid peroxides, can enhance cancer vulnerability to chemotherapy, targeted therapy and immunotherapy. As an essential upstream process for ferroptosis activation, lipid peroxidation of biological membranes is expected to be primarily induced by intrabilayer reactive oxygen species (ROS), indicating a promising strategy to initiate peroxidation by improving the local content of diffusion-limited ROS in the lipid bilayer. Herein, liposomes embedded with PEG-coated 3 nm γ-Fe2O3 nanoparticles in the bilayer (abbreviated as Lp-IO) were constructed to promote the intrabilayer generation of hydroxyl radicals (•OH) from hydrogen peroxide (H2O2), and the integration of amphiphilic PEG moieties with liposomal bilayer improved lipid membrane permeability to H2O2 and •OH, resulting in efficient initiation of lipid peroxidation and thus ferroptosis in cancer cells. Additionally, Lp-IO enabled traceable magnetic resonance imaging and pH/ROS dual-responsive drug delivery. Synergistic antineoplastic effects of chemotherapy and ferroptosis, and alleviated chemotherapeutic toxicity, were achieved by delivering doxorubicin (capable of xCT and glutathione peroxidase inhibition) with Lp-IO. This work provides an efficient alternative for triggering therapeutic lipid peroxidation and a ferroptosis-activating drug delivery vehicle for combination cancer therapies.

6.
J Mater Chem B ; 10(45): 9457-9465, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36346268

ABSTRACT

The development and progression of colorectal cancer (CRC) are highly dependent on the long-term inflammatory microenvironment with immune dysregulation in the colorectum. However, effective therapeutics are limited to targeting CRC. Here, we developed oral fullerene tablets (OFTs) that can act directly on the colorectal site by oral administration and reduce the inflammatory state at the tumor site for effective CRC therapy. In detail, OFTs scavenged reactive oxygen species (ROS), restrained the mutation of the wild-type P53, inhibited the activation of the inflammatory pathway nuclear factor-κB (NF-κB) and the signal transducer and activator of transcription 3 (STAT3) in the colorectum of CRC mice. Subsequently, OFTs could greatly reduce the infiltration of pro-inflammatory M1 macrophages and neutrophils at the tumor site, restoring the inflammatory microenvironment and immune homeostasis in the colorectal region, and ultimately achieving the inhibition of CRC. In addition, there were no significant toxic side effects of the long-term administration of OFTs. Our work provides an effective oral therapeutic strategy for CRC therapy by modulating the colorectal tumor inflammatory microenvironment and sheds light on the route for oral nano-materials in the clinical treatment of CRC.


Subject(s)
Colorectal Neoplasms , Fullerenes , Mice , Animals , Fullerenes/pharmacology , Fullerenes/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Signal Transduction , NF-kappa B/metabolism , Tablets , Tumor Microenvironment
7.
Adv Sci (Weinh) ; 9(29): e2201541, 2022 10.
Article in English | MEDLINE | ID: mdl-36031401

ABSTRACT

Malignant proliferation and metastasis are the hallmarks of cancer cells. Aminated [70]fullerene exhibits notable antineoplastic effects, promoting it a candidate for multi-targeted cancer drugs. It is an urgent need to reveal the structure-activity relationship for antineoplastic aminated fullerenes. Herein, three amphiphilic derivatives of [60]fullerene with clarified molecular structures are synthesized: TAPC-4, TAPC-3, and TCPC-4. TAPC-4 inhibits the proliferation of diverse tumor cells via G0/G1 cell cycle arrest, reverses the epithelial-mesenchymal transition, and abrogates the high mobility of tumor cells. TAPC-4 can be excreted from the organism and achieves an in vivo inhibition index of 75.5% in tumor proliferation and 87.5% in metastatic melanoma with a wide safety margin. Molecular dynamics simulations reveal that the amphiphilic molecular structure and the ending amino groups promote the targeting of TAPC-4 to heat shock protein Hsp90-beta, vimentin, and myosin heavy chain 9 (MYH9), probably resulting in the alteration of cyclin D1 translation, vimentin expression, and MYH9 location, respectively. This work initially emphasizes the dominant role of the amphiphilic structure and the terminal amino moieties in the antineoplastic effects of aminated fullerenes, providing fundamental support for their anti-tumor drug development.


Subject(s)
Antineoplastic Agents , Fullerenes , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cyclin D1 , Fullerenes/chemistry , Fullerenes/pharmacology , Fullerenes/therapeutic use , Heat-Shock Proteins , Myosin Heavy Chains , Vimentin
8.
Front Oncol ; 12: 779939, 2022.
Article in English | MEDLINE | ID: mdl-35433481

ABSTRACT

Exosomes are small extracellular vesicles critical for intercellular signaling via their delivery of cargoes, including proteins, DNA, RNA, lipids, and metabolites. Exosomes play essential roles in remodeling the tumor microenvironment (TME) for tumor growth, metastasis, and drug resistance. Aminated fullerenes (e.g., C70-ethylenediamine [EDA]) exhibit antineoplastic effects by targeting multiple functional proteins. Nanosized C70-EDA with positive surface charges tends to be taken up by monocytes in the bloodstream and monocyte-derived macrophages in the TME. Herein, the alterations of monocytes and monocyte-derived exosomes by C70-EDA have been investigated. C70-EDA reprogramed THP-1 monocyte to an M2-like state and substantially increased the protein content in exosomes secreted by M2-like monocytes. Notably, C70-EDA-induced M2-like monocytes released exosomes that triggered the proliferation of recipient tumor cells, which may alleviate the antineoplastic efficacy of C70-EDA. As revealed by proteomic profiling of exosomes, this outcome is probably a result of Rho GTPase/p21-activated kinase (PAK) pathway activation in recipient tumor cells induced by upregulated exosomal proteins. This work indicates a promising strategy in which aminated fullerenes can be combined with PAK inhibitors for cancer therapy.

9.
Sci China Life Sci ; 65(6): 1146-1156, 2022 06.
Article in English | MEDLINE | ID: mdl-34735681

ABSTRACT

Ulcerative colitis (UC) is a long-term, recurrent inflammatory bowel disease for which no effective cure is yet available in the clinical setting. Repairing the barrier dysfunction of the colon and reducing intestinal inflammation are considered key objectives to cure UC. Here we demonstrate a novel therapeutic strategy based on a C60 fullerene suspension (C60FS) to treat dinitrobenzene sulfonic acid-induced UC in an animal model. C60FS can repair the barrier dysfunction of UC and effectively promote the healing of ulcers; it also manifests better treatment effects compared with mesalazine enema. C60FS can reduce the numbers of basophils in the blood of UC rats and mast cells in the colorectal tissue, thereby effectively alleviating inflammation. The expression of H1R, H4R, and VEGFR2 receptors in colorectal tissues is inhibited by C60FS, and the levels of histamine and prostaglandin in the rat blood are reduced. This work presents a reliable strategy based on fullerene to cure UC and provides a novel guide for UC treatment.


Subject(s)
Colitis, Ulcerative , Colorectal Neoplasms , Fullerenes , Nanoparticles , Animals , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Colorectal Neoplasms/metabolism , Disease Models, Animal , Fullerenes/metabolism , Fullerenes/therapeutic use , Inflammation/metabolism , Intestinal Mucosa/metabolism , Rats
10.
Langmuir ; 37(8): 2740-2748, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33586439

ABSTRACT

Hydrophilic fullerene derivatives get notable performance in various biological applications, especially in cancer therapy and antioxidation. The biological behaviors of functional fullerenes are much dependent on their surface physicochemical properties. The excellent reactive oxygen species-scavenging capabilities of functional fullerenes promote their outstanding performances in inhibiting pathological symptoms associated with oxidative stress, including neurodegenerative diseases, cardiovascular diseases, acute and chronic kidney disease, and diabetes. Herein, fullerene derivatives with reversed surface charges in aqueous solutions are prepared: cationic C60-EDA and anionic C60-(EDA-EA). Under the driving force of membrane potential (negative inside) in the cell and mitochondria, C60-EDA is much rapidly taken in by cells and transported into mitochondria compared with C60-(EDA-EA) that is enriched in lysosomes. With high cellular uptake and mitochondrial enrichment, C60-EDA exhibits stronger antioxidation capabilities in vitro than C60-(EDA-EA), indicating its better performance in the therapy of oxidation-induced diseases. It is revealed that the cellular uptake rate, subcellular location, and intracellular antioxidation behavior of fullerene derivatives are primarily mediated by their surface charges, providing new strategies for the design of fullerene drugs and their biological applications.


Subject(s)
Fullerenes , Antioxidants/pharmacology , Organelles , Oxidative Stress , Reactive Oxygen Species
11.
ACS Appl Mater Interfaces ; 12(51): 56862-56873, 2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33305958

ABSTRACT

Functional fullerene derivatives exhibit fantastic inhibitory capabilities against cancer survival and metastasis, but the absence of clarified biological molecular targets and ambiguous regulation mechanisms set barriers for their clinical transformation. Cancer metastasis is the primary cause of mortality and initiated with increased cell migration, making cell motility regulation a high-value therapeutic target in precision medicine. Herein, a critical molecular target of the aminated fullerene derivative (C70-EDA), myosin heavy chain 9 (MYH9), was initially identified by a pull-down assay and MS screening. MYH9 is a cytoplasm-located protein and is responsible for cell motility and epithelial-mesenchymal transition regulation. Omics data from large-scale clinical samples reveals that MYH9 gets overexpressed in various cancers and correlates with unfavorable prognosis, indicating that it is a potential antineoplastic target. It is unveiled that C70-EDA binds to the C-terminal of MYH9, triggering the transport of MYH9 from the cytoplasm to the cell edge, blocking the MYH9-involved cell mobility, and inhibiting the metastasis-associated EMT process. This work provides a precise biological target and new strategies for fullerene applications in cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Fullerenes/pharmacology , Myosin Heavy Chains/metabolism , A549 Cells , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Biotin/analogs & derivatives , Biotin/metabolism , Biotin/pharmacology , Drug Screening Assays, Antitumor , Epithelial-Mesenchymal Transition/drug effects , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Fluorescein-5-isothiocyanate/pharmacology , Fullerenes/chemistry , Fullerenes/metabolism , Humans , Protein Binding
12.
Nanoscale ; 7(38): 16061-70, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26372069

ABSTRACT

Drug delivery systems (DDSs) with biocompatibility and precise drug delivery are eagerly needed to overcome the paradox in chemotherapy that high drug doses are required to compensate for the poor biodistribution of drugs with frequent dose-related side effects. In this work, we reported a metal-organic framework (MOF) based tumor targeting DDS developed by a one-pot, and organic solvent-free "green" post-synthetic surface modification procedure, starting from the nanoscale MOF MIL-101. Owing to the multifunctional surface coating, premature drug release from this DDS was prevented. Due to the pH responsive benzoic imine bond and the redox responsive disulfide bond at the modified surface, this DDS exhibited tumor acid environment enhanced cellular uptake and intracellular reducing environment triggered drug release. In vitro and in vivo results showed that DOX loaded into this DDS exhibited effective cancer cell inhibition with much reduced side effects.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Delivery Systems , Animals , COS Cells , Chlorocebus aethiops , HeLa Cells , Humans , Nanoparticles , Tissue Distribution
13.
Phys Rev Lett ; 110(16): 167003, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-23679633

ABSTRACT

Recently, a debate has arisen over which of the two distinct parts of the Fermi surface of Sr(2)RuO(4) is the active part for the chiral p-wave superconductivity exhibited. Early theories proposed p-wave pairing on the two-dimensional γ band, whereas a recent proposal focuses on the one-dimensional (α, ß) bands whose nesting pockets are the source of the strong incommensurate spin density wave (SDW) fluctuations. We apply a renormalization group theory to study quasi-one-dimensional repulsive Hubbard chains and explain the form of SDW fluctuations, reconciling the absence of long-range order with their nesting Fermi surface. The mutual exclusion of p-wave pairing and SDW fluctuations in repulsive Hubbard chains favors the assignment of the two-dimensional γ band as the source of p-wave pairing.

14.
Phys Rev Lett ; 108(25): 257002, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-23004642

ABSTRACT

By using Bogoliubov-de Gennes equations, we study superconducting (SC) states in a quasi-two-dimensional system of radius R. It is shown that no vortices exist in s-wave SC samples with R < R(c) ~ ξ(0), the T = 0 coherence length. We predict that chiral p-wave states exhibit superconductivity for R < R(c) only in the presence of a vortex with opposite chirality. This induced SC phase is a consequence of nonzero chirality of the pairing order parameter and implies the presence of chiral edge currents. Our study may be applied to sharply probing the pairing symmetry of unconventional superconductors.

SELECTION OF CITATIONS
SEARCH DETAIL
...