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










Publication year range
1.
Zhongguo Zhong Yao Za Zhi ; 49(5): 1144-1153, 2024 Mar.
Article in Chinese | MEDLINE | ID: mdl-38621961

ABSTRACT

Agaricus blazei is a rare medicinal and edible fungus with a crispy taste and delicious flavor. Both fruiting body and mycelium are rich in polysaccharides, sterols, terpenoids, peptides, lipids, polyphenols, and other active ingredients, which have strong pharmacological activities such as anti-tumor, lipid-lowering, glucose-lowering, immunomodulation, optimization of intestinal flora, and anti-oxidation. Therefore, it is a kind of fungal resource with a great prospect of edible and medicinal development. Among the reported chemical components of A. blazei, blazeispirol is a series of sterol compounds unique to A. blazei, which has a spiral structure and is different from classical steroids. It is an important active ingredient found in the mycelium of A. blazei and has significant hepatoprotective activity. It can be used as a phylogenetic and chemotaxonomic marker of A. blazei strains and is considered an excellent lead compound for drug development. According to the skeleton structure characteristics, the 17 discovered blazeispirol compounds can be divided into two types: blazeispirane and problazeispirane. In order to further explore the resource of blazeispirol compounds of A. blazei, the discovery, isolation, structure, biological activity, and biosynthetic pathways of blazeispirol compounds of A. blazei were systematically reviewed. Besides, the metabolic regulation strategies related to the fermentation synthesis of blazeispirol A by A. blazei were discussed. This review could provide a reference for the efficient synthesis and development of blazeispirol compounds, the research and development of related drugs and functional foods, and the quality improvement of A. blazei and other medicinal and edible fungi resources and derivatives.


Subject(s)
Agaricus , Neoplasms , Phylogeny , Polysaccharides , Steroids , Agaricus/chemistry , Agaricus/metabolism
2.
J Control Release ; 353: 591-610, 2023 01.
Article in English | MEDLINE | ID: mdl-36503071

ABSTRACT

Intracellular Methicillin-Resistant Staphylococcus aureus (MRSA) remains a major factor of refractory and recurrent infections, which cannot be well addressed by antibiotic therapy. Here, we design a cellular infectious microenvironment-activatable polymeric nano-system to mediate targeted intracellular drug delivery for macrophage reprogramming and intracellular MRSA eradication. The polymeric nano-system is composed of a ferrocene-decorated polymeric nanovesicle formulated from poly(ferrocenemethyl methacrylate)-block-poly(2-methacryloyloxyethyl phosphorylcholine) (PFMMA-b-PMPC) copolymer with co-encapsulation of clofazimine (CFZ) and interferon-γ (IFN-γ). The cellular-targeting PMPC motifs render specific internalization by macrophages and allow efficient intracellular accumulation. Following the internalization, the ferrocene-derived polymer backbone sequentially undergoes hydrophobic-to-hydrophilic transition, charge reversal and Fe release in response to intracellular hydrogen peroxide over-produced upon infection, eventually triggering endosomal escape and on-site cytosolic drug delivery. The released IFN-γ reverses the immunosuppressive status of infected macrophages by reprogramming anti-inflammatory M2 to pro-inflammatory M1 phenotype. Meanwhile, intracellular Fe2+-mediated Fenton reaction together with antibiotic CFZ contributes to increased intracellular hydroxyl radical (•OH) generation. Ultimately, the nano-system achieves robust potency in ablating intracellular MRSA and antibiotic-tolerant persisters by synchronous immune modulation and efficient •OH killing, providing an innovative train of thought for intracellular MRSA control.


Subject(s)
Anti-Bacterial Agents , Macrophages , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Interferon-gamma , Macrophages/immunology , Metallocenes/therapeutic use , Methicillin-Resistant Staphylococcus aureus/immunology , Polymers/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcal Infections/immunology , Nanostructures/therapeutic use
3.
Org Lett ; 24(45): 8283-8288, 2022 11 18.
Article in English | MEDLINE | ID: mdl-36331137

ABSTRACT

In this work, we successfully explored an unexpected dehydrogenation triggered by Pd/Cu-catalyzed C(sp3)-H arylation and intramolecular C-N coupling of amides to synthesize the bioactive 1,2-dihydroquinoline scaffold with good regioselectivity and good compatibility of functional groups. This strategy provides an alternative route to realize molecular complexity and diversity from simple and readily available molecules via multiple C-H bond activation. Preliminary mechanistic studies demonstrated that ß,γ-dehydrogenation is triggered by the arylation of the C(sp3)-H bond and the intramolecular C-N coupling.


Subject(s)
Amides , Palladium , Amides/chemistry , Palladium/chemistry , Catalysis , Molecular Structure
4.
Biomaterials ; 281: 121358, 2022 02.
Article in English | MEDLINE | ID: mdl-34979416

ABSTRACT

The overexpression of glutathione (GSH) in cancer cells has long been regarded as the primary obstacle for reactive oxygen species (ROS)-involved anti-tumor therapies. To solve this issue, a ferric ion and selenite-codoped calcium phosphate (Fe/Se-CaP) nanohybrid here is fabricated to catabolize endogenous GSH, instead of directly deleting it, to trigger a ROS storm for tumor suppression. The selenite component in Fe/Se-CaP can catabolize GSH to superoxide anion (O2•-) and hydroxyl radicals (•OH) via cascade catalytic reactions, elevating oxidative stress while destroying antioxidant system. The doped Fe can further catalyze the soaring hydrogen peroxide (H2O2) originated from O2•- to •OH via Fenton reactions. Collectively, Fe/Se-CaP mediated self-augmented catabolism dynamic therapy finally induces apoptosis of cancer cells owing to the significant rise of ROS and, combined with CaP adjuvant, evokes adaptive immune responses to suppress tumor progression, providing an innovative train of thought for ROS-involved anti-tumor therapies.


Subject(s)
Glutathione , Hydrogen Peroxide , Glutathione/metabolism , Hydrogen Peroxide/metabolism , Iron , Reactive Oxygen Species/metabolism , Selenious Acid , Superoxides/metabolism
5.
Nanoscale ; 13(8): 4420-4431, 2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33616147

ABSTRACT

Traditional methods of depleting tumor-associated myeloid cells via chemotherapy can easily lead to the re-recruitment of them, eventually resulting in chemo-resistance and presenting obstacles in immunotherapy. Herein, we report a nano-educator (NE) that when loaded with all trans retinoic acid (ATRA) and anti-PD-1 antibodies (aPD-1) instructs myeloid cells to assist T cells towards revitalizing anti-PD-1 therapy. In vivo, ATRA converts myeloid-derived suppressor cells (MDSCs) into dendritic cells (DCs), which are essential for anti-PD-1 therapy, while intervening in the polarization of macrophages. Furthermore, aPD-1-armed T cells reboot anti-tumor immunity after suppression relief, which exposes tumor-specific antigens and in turn promotes the maturation of transformed DCs. The nano-platform provides shelter for vulnerable immunomodulatory agents and durable drug release to stimulate intensive immune modulation. We established three types of tumor-bearing mice models with different myeloid cell contents to show the spatiotemporal complementarity of ATRA and aPD-1. The NE re-educates the tumor's guard to assist T cells in enhanced immunotherapy, broadening the application of aPD-1 in the treatment of anti-PD-1-resistant tumors.


Subject(s)
Myeloid Cells , Myeloid-Derived Suppressor Cells , Animals , Cell Line, Tumor , Immunotherapy , Macrophages , Mice
6.
Biomaterials ; 245: 119986, 2020 07.
Article in English | MEDLINE | ID: mdl-32229331

ABSTRACT

Diabetes is an increasing health problem and associated with inflammatory complications that seriously affects the quality of life and survival of patients. Carbon monoxide (CO), owing to its anti-inflammatory and anti-apoptotic properties, has become a potential therapeutic molecule for the treatment of autoimmune diseases. Here, we constructed a mesoporous silica-based biomimetic CO nanogenerator (mMMn), which was loaded with manganese carbonyl and camouflaged with macrophage membrane. Driven by the active targeting of macrophage membrane to inflammatory sites, the as-designed mMMn could effectively accumulate in pancreatic tissue of type 1 diabetic mice, which was established by consecutive administration of streptozotocin (STZ). It was found that the local reactive oxygen species (ROS) within pancreas could trigger the continuous CO release from mMMn, which greatly ameliorated diabetes in mice with improved blood glucose homeostasis by alleviating inflammatory responses and inhibiting ß-cells apoptosis. The exogenous CO targeting to pancreatic tissue paves a novel way for the treatment of type 1 diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Animals , Apoptosis , Biomimetics , Blood Glucose , Carbon Monoxide , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Humans , Mice , Quality of Life , Streptozocin
7.
Adv Mater ; 32(16): e2000376, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32134530

ABSTRACT

Traditional phototherapies face the issue that the insufficient penetration of light means it is difficult to reach deep lesions, which greatly reduces the feasibility of cancer therapy. Here, an implantable nitric oxide (NO)-release device is developed to achieve long-term, long-distance, remote-controllable gas therapy for cancer. The device consists of a wirelessly powered light-emitting diode (wLED) and S-nitrosoglutathione encapsulated with poly(dimethylsiloxane) (PDMS), obtaining the NO-release wLED (NO-wLED). It is found that NO release from the NO-wLED can be triggered by wireless charging and the concentration of produced NO reaches 0.43 × 10-6 m min-1 , which can achieve a killing effect on cancer cells. In vivo anticancer experiments exhibit obvious inhibitory effect on the growth of orthotopic cancer when the implanted NO-wLED is irradiated by wireless charging. In addition, recurrence of cancer can be prevented by NO produced from the NO-wLED after surgery. By illumination in the body, this strategy overcomes the poor penetration and long-wavelength dependence of traditional phototherapies, which also provides a promising approach for in vivo gas therapy remote-controlled by wireless charging.


Subject(s)
Colonic Neoplasms/therapy , Nitric Oxide/metabolism , Phototherapy/instrumentation , Wireless Technology , Animals , Cell Line, Tumor , Colonic Neoplasms/metabolism , Electric Power Supplies , Mice
8.
Biomaterials ; 223: 119472, 2019 12.
Article in English | MEDLINE | ID: mdl-31499254

ABSTRACT

Inflammation during photothermal therapy (PTT) of tumor usually results in adverse consequences. Here, a biomembrane camouflaged nanomedicine (mPDAB) containing polydopamine and ammonia borane was designed to enhance PTT efficacy and mitigate inflammation. Polydopamine, a biocompatible photothermal agent, can effectively convert light into heat for PTT. Ammonia borane was linked to the surface of polydopamine through the interaction of hydrogen bonding, which could destroy redox homoeostasis in tumor cells and reduce inflammation by H2 release in tumor microenvironment. Owing to the same origin of outer biomembranes, mPDAB showed excellent tumor accumulation and low systemic toxicity in a breast tumor model. Excellent PTT efficacy and inflammation reduction made the mPDAB completely eliminate the primary tumors, while also restraining the outgrowth of distant dormant tumors. The biomimetic nanomedicine shows potentials as a universal inflammation-self-alleviated platform to ameliorate inflammation-related disease treatment, including but not limited to PTT for tumor.


Subject(s)
Ammonia/chemistry , Boranes/chemistry , Breast Neoplasms/drug therapy , Hydrogen , Phototherapy/methods , Animals , Biocompatible Materials , COS Cells , Chlorocebus aethiops , Female , Gases , HeLa Cells , Homeostasis , Humans , Inflammation , Mammary Neoplasms, Experimental/drug therapy , Membranes, Artificial , Mice , Nanomedicine/methods , Neoplasm Transplantation , Oxidation-Reduction , Recurrence , Temperature , Tumor Microenvironment
9.
Nano Lett ; 19(8): 5568-5576, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31262183

ABSTRACT

Tumor cells adapt to reactive oxygen species (ROS) attacking by launching DNA damage repairing mechanisms such as nucleotide pool sanitizing enzyme mutt homologue 1 (MTH1) to mitigate the oxidatively induced DNA lesions, which could greatly limit the therapeutic efficiency of current oxidation therapy. Here, an amplified oxidative damage strategy for tumor therapy was proposed that was focused not only on the enhancement of ROS generation but also the inhibition of subsequent MTH1 enzyme activity simultaneously. In our formulation, mesoporous silica-coated Prussian blue nanoplatforms (PB@MSN) with excellent catalase-like activity and drug loading capability were employed to encapsulate MTH1 inhibitor TH287, followed by the modification of tetraphenylporphrin zinc (Zn-Por) via metallo-supramolecular coordination (PMPT), where Zn-Por behaved as photodynamic and fluorescence imaging agents, as well as acid-responsive gatekeepers. The intelligent PMPT nanosystems could induce the decomposition of H2O2 to relieve the hypoxic tumor environment, thus elevating the generation of singlet oxygen for improved oxidative damage. In the meantime, controllable-released TH287 from pores could hinder MTH1-mediated damage repairing process and aggravate oxidative damage, thereby resulting in cellular toxicity as well as tumor growth inhibition.


Subject(s)
Breast Neoplasms/drug therapy , DNA Repair Enzymes/antagonists & inhibitors , Oxidative Stress/drug effects , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Photosensitizing Agents/therapeutic use , Porphyrins/therapeutic use , Pyrimidines/therapeutic use , Animals , Breast Neoplasms/metabolism , DNA Repair Enzymes/metabolism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Female , Humans , MCF-7 Cells , Mice, Nude , Optical Imaging , Phosphoric Monoester Hydrolases/metabolism , Photochemotherapy , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Pyrimidines/administration & dosage , Reactive Oxygen Species/metabolism
10.
Biomaterials ; 217: 119303, 2019 10.
Article in English | MEDLINE | ID: mdl-31271859

ABSTRACT

Here, a highly cooperative liquid metal nanoparticle-enzyme (LM@GOX) was constructed for combinational starvation/photothermal therapy of tumor. It was found that the enzyme activity of glucose oxidase (GOX) could be strengthened along with the increased temperature within a given range and its optimal activity is around about 43-60 °C. Utilizing the photothermal conversion ability of liquid metal (LM), the GOX catalytic efficiency could be photo-controlled with improved starvation therapeutic efficiency. Furthermore, due to the accelerating blood flow during the photothermal therapy (PTT), the hypoxic situation in tumor tissues could also be relieved, which would contribute to conquering the hypoxia-suppressed GOX catalysis. In the meanwhile, the severe thermo-resistance of tumor cells during PTT process could be overcome by GOX induced decrease of adenosine triphosphate (ATP) and heat shock proteins (HSPs) level, eventually leading to an improved therapeutic effect of PTT. Both in vitro and in vivo studies proved that LM@GOX could significantly inhibit the growth of solid tumor under NIR illumination by a win-win cooperative starvation/photothermal therapy.


Subject(s)
Glucose Oxidase/metabolism , Hyperthermia, Induced , Light , Metal Nanoparticles/chemistry , Neoplasms/therapy , Phototherapy , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/pharmacology , Body Weight/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Glucose/pharmacology , Heat-Shock Proteins/metabolism , Humans , Infrared Rays , Metal Nanoparticles/ultrastructure , Mice, Inbred BALB C , Optical Imaging , Tumor Burden/drug effects
11.
ACS Nano ; 13(8): 8618-8629, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31246413

ABSTRACT

Natural nanoparticles have been extensively studied due to their diverse properties and easy accessibility. Here, the nanoparticles extracted from cuttlefish ink (CINPs) with significant antitumor efficacy are explored. These CINPs, with spherical morphology, good dispersibility, and biocompatibility, are rich in melanin and contain a variety of amino acids and monosaccharides. Through the activation of mitogen-activated protein kinase (MAPK) signaling pathway, CINPs can efficiently reprogram tumor-associated macrophages (TAMs) from immune-suppressive M2-like phenotype to antitumor M1-like phenotype. Besides, under near-infrared (NIR) irradiation, CINPs exhibit high photothermal effect and tumor cell killing ability, which make them a potential candidate in photothermal therapy (PTT) of tumor. In vivo, CINPs can increase the proportion of M1 macrophages and foster the recruitment of cytotoxic T lymphocytes (CTLs) to tumors, leading to reduced primary tumor growth and lung metastasis. In combination with their photothermal effect, which can induce tumor-specific antigens release, CINPs could almost completely inhibit tumor growth accompanied by more active immune responses. Collectively, these CINPs described here can provide both tumor immunotherapy and PTT, implying that CINPs are promising for tumor treatment.


Subject(s)
Immunotherapy , Ink , Nanoparticles/chemistry , Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Decapodiformes/chemistry , Humans , Hyperthermia, Induced , Indoles/chemistry , Indoles/pharmacology , Macrophages/drug effects , Mice , Phototherapy , T-Lymphocytes, Cytotoxic/drug effects
12.
ACS Nano ; 13(5): 5523-5532, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31046229

ABSTRACT

Carbon monoxide (CO) is regarded as a potential therapeutic agent with multiple beneficial functions for biomedical applications. In this study, a versatile CO nanogenerator (designated as PPOSD) was fabricated and developed for tumor therapy and anti-inflammation. Partially oxidized tin disulfide (SnS2) nanosheets (POS NSs) were decorated with a tumor-targeting polymer (polyethylene glycol-cyclo(Asp-d-Phe-Lys-Arg-Gly), PEG-cRGD), followed by the loading of chemotherapeutic drug doxorubicin (DOX) to prepare polymer@POS@DOX, or PPOSD. After injected intravenously, PPOSD could selectively accumulate in tumor tissue via the cRGD-mediated tumor recognition. Upon 561 nm laser irradiation, the POS moiety in PPOSD can photoreduce CO2 to CO, which significantly sensitized the chemotherapeutic effect of DOX. The POS in PPOSD can also act as a photothermal agent for effective photothermal therapy (PTT) of the tumor upon 808 nm laser irradiation. Furthermore, the generated CO can simultaneously decrease the inflammatory reaction caused by PTT. Blood analysis and hematoxylin-eosin staining of major organs showed that no obvious systemic toxicity was induced after the treatment, suggesting good biosafety of PPOSD. This versatile CO nanogenerator will find great potential for both enhanced tumor inhibition and anti-inflammation.


Subject(s)
Carbon Monoxide/pharmacology , Inflammation/drug therapy , Neoplasms/drug therapy , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carbon Dioxide/chemistry , Carbon Monoxide/chemistry , Cell Line, Tumor , Disulfides/chemistry , Disulfides/pharmacology , Doxorubicin/pharmacology , Humans , Inflammation/pathology , Mice , Nanoparticles/chemistry , Neoplasms/pathology , Xenograft Model Antitumor Assays
13.
Biomaterials ; 207: 76-88, 2019 07.
Article in English | MEDLINE | ID: mdl-30959408

ABSTRACT

Various negative effects accompanying with the instability of bare liquid metal (LM) nanoparticles, including undesirable spontaneous coalescence, continuous photothermal performance deterioration and difficult multi-step functionalization, severely hinder its applications in biomedical area. In this study, we proposed a new concept of immobilized liquid metal nanoparticles based on a surface mesoporous silica coating strategy (LM@MSN). Strikingly, it was found that unsteady and vulnerable LM nanoparticles after immobilization exhibited enhanced stabilization and sustainable photothermal performance even with a long and repeated light irradiation in acidic environments. Moreover, integrating the properties of easy surface functionalization and high drug loading efficiency from silica shell, immobilized LM nanoparticle was further used for photothermal involved combinational therapy. The classical anticancer drug doxorubicin (DOX) was encapsulated in pores of silica shell and the hyaluronic acid (HA) was decorated on LM@MSN to construct LM@MSN/DOX@HA for tumor targeted combination therapy. Both in vitro and in vivo studies proved that LM@MSN/DOX@HA could significantly inhibit solid tumor growth under near infrared (NIR) irradiation by synergistic photothermal/chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Doxorubicin/therapeutic use , Metal Nanoparticles/chemistry , Antineoplastic Agents/chemistry , Doxorubicin/chemistry , Drug Delivery Systems/methods , Porosity , Silicon Dioxide/chemistry
14.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 30(6): 641-4, 2013 Dec.
Article in Chinese | MEDLINE | ID: mdl-24327137

ABSTRACT

OBJECTIVE: To detect the underlying genetic defect in two Chinese families with hereditary multiple exostoses and provide genetic counseling. METHODS: Potential mutations in EXT1 and EXT2 genes in the probands were detected by direct sequencing of PCR-amplified exons. Suspected mutations were verified in all available family members and 200 unrelated healthy controls. RESULTS: A heterozygous frameshift mutation c.346_356delinsTAT in exon 1 of EXT1 and a heterozygous deletion mutation c.2009-2012del(TCAA) in exon 10 of EXT1 were respectively detected in affected members from the two families. The same mutations were not detected in unaffected members and 200 unrelated healthy controls. No mutations in EXT2 were detected in the two families. CONCLUSION: Two novel mutations of EXT1 have been detected in association with hereditary multiple exostoses in two Chinese families. Above results have provided a basis for genetic counseling for the two families and expanded the spectrum of EXT1 mutations.


Subject(s)
Exostoses, Multiple Hereditary/enzymology , Exostoses, Multiple Hereditary/genetics , N-Acetylglucosaminyltransferases/genetics , Sequence Deletion , Adolescent , Adult , Aged , Asian People/genetics , Child , Child, Preschool , DNA Mutational Analysis/methods , Female , Heterozygote , Humans , Male , Middle Aged , Pedigree , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...