Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 181
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Environ Toxicol ; 39(7): 3808-3819, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38523403

ABSTRACT

Silicon dioxide (SiO2)-induced pulmonary fibrosis is potentially associated with the impairment of mitochondrial function. Previous research found that inhibition of macrophage receptor with collagenous structure (MARCO) could alleviate particle-induced lung injury by regulating phagocytosis and mitigating mitochondrial damage. The present study aims to explore the underlying anti-fibrosis mechanism of polyguanylic acid (PolyG, MARCO inhibitor) in a silicotic rat model. Hematoxylin and eosin and Masson staining were performed to visualize lung tissue pathological changes. Confocal microscopy, transmission electron microscope, western blot analysis, quantitative real-time PCR (qPCR), and adenosine triphosphate (ATP) content assay were performed to evaluate collagen content, mitochondrial function, and morphology changes in SiO2-induced rat pulmonary fibrosis. The results suggested that SiO2 exposure contributed to reactive oxygen species aggregation and the reduction of respiratory complexes and ATP synthesis. PolyG treatment could effectively reduce MARCO expression and ameliorate lung injury and fibrosis by rectifying the imbalance of mitochondrial respiration and energy synthesis. Furthermore, PolyG could maintain mitochondrial homeostasis by promoting peroxisome proliferator-activated receptor-coactivator 1 α (PGC1α)-mediated mitochondrial biogenesis and regulating fusion and fission. Together, PolyG could ameliorate SiO2-induced pulmonary fibrosis via inhibiting MARCO to protect mitochondrial function.


Subject(s)
Mitochondria , Pulmonary Fibrosis , Silicon Dioxide , Silicosis , Animals , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Silicosis/drug therapy , Silicosis/pathology , Silicosis/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Silicon Dioxide/toxicity , Male , Rats , Rats, Sprague-Dawley , Disease Models, Animal , Lung/drug effects , Lung/pathology , Lung/metabolism , Reactive Oxygen Species/metabolism
2.
Environ Geochem Health ; 45(8): 6585-6603, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37341891

ABSTRACT

Oxidative stress and inflammation are mechanisms underlying toxicity induced by fine particulate matter (PM2.5). The antioxidant baseline of the human body modulates the intensity of oxidative stress in vivo. This present study aimed to evaluate the role of endogenous antioxidants in alleviating PM2.5-induced pulmonary injury using a novel mouse model (LiasH/H) with an endogenous antioxidant capacity of approximately 150% of its wild-type counterpart (Lias+/+). LiasH/H and wild-type (Lias+/+) mice were randomly divided into control and PM2.5 exposure groups (n = 10), respectively. Mice in the PM2.5 group and the control group were intratracheally instilled with PM2.5 suspension and saline, respectively, once a day for 7 consecutive days. The metal content, major pathological changes in the lung, and levels of oxidative stress and inflammation biomarkers were examined. The results showed that PM2.5 exposure induced oxidative stress in mice. Overexpression of the Lias gene significantly increased the antioxidant levels and decreased inflammatory responses induced by PM2.5. Further study found that LiasH/H mice exerted their antioxidant function by activating the ROS-p38MAPK-Nrf2 pathway. Therefore, the novel mouse model is useful for the elucidation of the mechanisms of pulmonary injury induced by PM2.5.


Subject(s)
Lung Injury , Particulate Matter , Humans , Mice , Animals , Particulate Matter/toxicity , Lung Injury/chemically induced , Antioxidants/metabolism , Lung , Oxidative Stress , Inflammation/metabolism
3.
Small ; 18(35): e2203292, 2022 09.
Article in English | MEDLINE | ID: mdl-35859534

ABSTRACT

An effective therapeutic strategy against methicillin-resistant Staphylococcus aureus (MRSA) that does not promote further drug resistance is highly desirable. While phototherapies have demonstrated considerable promise, their application toward bacterial infections can be limited by negative off-target effects to healthy cells. Here, a smart targeted nanoformulation consisting of a liquid perfluorocarbon core stabilized by a lipid membrane coating is developed. Using vancomycin as a targeting agent, the platform is capable of specifically delivering an encapsulated photosensitizer along with oxygen to sites of MRSA infection, where high concentrations of pore-forming toxins trigger on-demand payload release. Upon subsequent near-infrared irradiation, local increases in temperature and reactive oxygen species effectively kill the bacteria. Additionally, the secreted toxins that are captured by the nanoformulation can be processed by resident immune cells to promote multiantigenic immunity that protects against secondary MRSA infections. Overall, the reported approach for the on-demand release of phototherapeutic agents into sites of infection could be applied against a wide range of high-priority pathogens.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Humans , Liposomes/pharmacology , Microbial Sensitivity Tests , Phototherapy , Staphylococcal Infections/drug therapy , Staphylococcal Infections/prevention & control
4.
Ecotoxicol Environ Saf ; 245: 114100, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36155331

ABSTRACT

Silicosis is a severe progressive lung disease without effective treatment methods. Previous evidence has demonstrated that endothelial cell to mesenchymal transition (EndoMT) plays an essential role in pulmonary fibrosis, and pulmonary fibrosis is associated with dysregulation of autophagy, while the relationship between autophagy and EndoMT has not yet been adequately studied. Herein, we established a mouse model of silicosis, and we found that the pharmacological induction of the AMPK/mTOR-dependent pathway using 100 mg/kg Metformin (Met) enhanced autophagy in vivo, and results of the Western blot showed that autophagy-related proteins, LC3 II/I ratio, and Beclin-1 increased while p62 decreased. In addition, Met treatment attenuated silica-induced pulmonary inflammation and decreased collagen deposition by suppressing EndoMT, and the proliferation of human umbilical vein endothelial cells (HUVECs) was also inhibited. Notably, the tube forming assay showed that Met also protected the vascular endothelial cells from silica-induced morphological damage. In conclusion, Met can alleviate inflammatory response and collagen deposition in the process of pulmonary fibrosis induced by silica via suppressing EndoMT through the AMPK/mTOR signaling pathway.


Subject(s)
Metformin , Pulmonary Fibrosis , Silicosis , AMP-Activated Protein Kinases , Animals , Autophagy , Autophagy-Related Proteins/pharmacology , Beclin-1 , Collagen , Human Umbilical Vein Endothelial Cells , Humans , Metformin/pharmacology , Mice , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Signal Transduction , Silicon Dioxide/toxicity , Silicosis/drug therapy , TOR Serine-Threonine Kinases
5.
Mol Pharm ; 17(10): 3857-3869, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32833457

ABSTRACT

Disulfiram (DSF) is an FDA-approved anti-alcoholic drug that has recently proven to be effective in cancer treatment. However, the short half-life in the bloodstream and the metal ion-dependent antitumor activity significantly limited the further application of DSF in the clinical field. To this end, we constructed a silk fibroin modified disulfiram/zinc oxide nanocomposites (SF/DSF@ZnO) to solubilize and stabilize DSF, and, more importantly, achieve pH triggered Zn2+ release and subsequent synergistic antitumor activity. The prepared SF/DSF@ZnO nanocomposites were spherical and had a high drug loading. Triggered by the lysosomal pH, SF/DSF@ZnO could induce the rapid release of Zn2+ under the acidic conditions and caused nanoparticulate disassembly along with DSF release. In vitro experiments showed that cytotoxicity of DSF could be enhanced by the presence of Zn2+, and further amplified when encapsulated into SF/DSF@ZnO nanocomposites. It was confirmed that the significantly amplified cytotoxicity of SF/DSF@ZnO was resulted from pH-triggered Zn2+ release, inhibited cell migration, and increased ROS production. In vivo study showed that SF/DSF@ZnO nanocomposites significantly increased the tumor accumulation and prolonged the retention time. In vivo antitumor experiments in the xenograft model showed that SF/DSF@ZnO exerted the highest tumor-inhibition rate among all the drug treatments. Therefore, this exquisite study established silk fibroin-modified disulfiram/zinc oxide nanocomposites, SF/DSF@ZnO, where ZnO not only acted as a delivery carrier but also served as a metal ion reservoir to achieve synergistic antitumor efficacy. The established DSF nanoformulation displayed excellent therapeutic potential in future cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Nanocomposites/administration & dosage , Neoplasms/drug therapy , Zinc/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Bombyx/chemistry , Cations, Divalent/pharmacokinetics , Cell Line, Tumor/transplantation , Disease Models, Animal , Disulfiram/administration & dosage , Disulfiram/chemistry , Disulfiram/pharmacokinetics , Drug Compounding/methods , Drug Liberation , Drug Screening Assays, Antitumor , Drug Synergism , Female , Fibroins/chemistry , Half-Life , Humans , Hydrogen-Ion Concentration , Mice , Neoplasms/pathology , Zinc Oxide/administration & dosage , Zinc Oxide/chemistry , Zinc Oxide/pharmacokinetics
6.
Epidemiol Infect ; 148: e236, 2020 10 05.
Article in English | MEDLINE | ID: mdl-33012300

ABSTRACT

Forecasting the epidemics of the diseases is very valuable in planning and supplying resources effectively. This study aims to estimate the epidemiological trends of the coronavirus disease 2019 (COVID-19) prevalence and mortality using the advanced α-Sutte Indicator, and its prediction accuracy level was compared with the most frequently adopted autoregressive integrated moving average (ARIMA) method. Time-series analysis was performed based on the total confirmed cases and deaths of COVID-19 in the world, Brazil, Peru, Canada and Chile between 27 February 2020 and 30 June 2020. By comparing the prediction reliability indices, including the root mean square error, mean absolute error, mean error rate, mean absolute percentage error and root mean square percentage error, the α-Sutte Indicator was found to produce lower forecasting error rates than the ARIMA model in all data apart from the prevalence testing set globally. The α-Sutte Indicator can be recommended as a useful tool to nowcast and forecast the COVID-19 prevalence and mortality of these regions except for the prevalence around the globe in the near future, which will help policymakers to plan and prepare health resources effectively. Also, the findings of our study may have managerial implications for the outbreak in other countries.


Subject(s)
Betacoronavirus , Coronavirus Infections/epidemiology , Pneumonia, Viral/epidemiology , COVID-19 , Coronavirus Infections/mortality , Forecasting , Humans , Models, Statistical , Pandemics , Pneumonia, Viral/mortality , Prevalence , Reproducibility of Results , SARS-CoV-2
7.
Pharmacol Res ; 145: 104256, 2019 07.
Article in English | MEDLINE | ID: mdl-31054312

ABSTRACT

Islet transplantation is the experimental strategy to treat type 1 diabetes by transplanting isolated islets from a donor pancreas into the recipient. While significant progress has been made in the islet transplantation field, islet loss before and after transplantation is still the major obstacle that currently precludes its widespread application. Islet must survive from possible cellular damages during the isolation procedure, storage time, islet injection process and post-transplantation immune rejection, only then the survived islets could produce insulin, actively regulating the blood glucose level. Therefore, islet protection needs to be addressed, especially regarding oxidative stress and immune response induced islet cell damages in diabetic patients. Many clinical data have shown that mildly elevated bilirubin levels in the body negatively correlate to the occurrence of an array of diseases that are related to increased oxidative stress, especially diabetes, and its complications. Recent studies confirmed that bilirubin helps receivers to suppress immune reaction and enable prolonged tolerance to islet transplantation. In this paper, we will review the pharmacological mechanism of bilirubin to modulate oxidative cellular damage and chronic inflammatory reaction in both diabetes and islet transplantation process. Also, we will present the clinical evidence of a strong correlation in bilirubin and diabetes. More importantly, we will summarize undergoing therapeutic applications of bilirubin in islet transplantation and discuss formulation approaches designed to overcome bilirubin delivery issues for future use.


Subject(s)
Bilirubin/therapeutic use , Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation , Animals , Bilirubin/pharmacology , Diabetes Mellitus, Type 1/metabolism , Humans , Oxidative Stress/drug effects
8.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 50(3): 334-338, 2019 May.
Article in Zh | MEDLINE | ID: mdl-31631599

ABSTRACT

OBJECTIVE: To study the mechanism of renal injury in Lepr db/ db mice with the leptin receptor homozygous deficiency. METHODS: Ten male of 28-week-old Lepr db/+ mice with leptin receptor heterozygous deficiency were selected as control group and ten male Lepr db/ db mice with leptin receptor homozygous deficiency were used in this study. After fasting for 8 hours, the body mass, fasting blood glucose (FBG) and glycosylated hemoglobulin (HbA1c) of the mice were measured. Blood of the mice was obtained from femoral artery before euthanasia. Serum creatinine (CRE), blood urea nitrogen (BUN), superoxide dismutase (SOD), glutathione (GSH) and malonaldehyde (MDA) were detected by corresponding kits, and serum interleukin-1ß (IL-1ß), monocyte chemotactic protein-1 (MCP-1) and tumor necrosis factor-α (TNF-α) were measured using enzyme-linked immunosorbent assay (ELISA) method. The kidney was taken for pathological observation. The expression levels of nuclear factor E2-related factor 2 (Nrf2) and nuclear factor kappa B (NF-κB) in renal were analyzed by Western blot. The mitochondria of renal was isolated by the corresponding kit. Meanwhile, the expression level of lipoic acid synthase (LIAS) in renal mitochondria was measured by Western blot. RESULTS: The body mass, FPG, HbA1c, CRE and BUN levels of the Lepr db/ db mice were significantly increased in comparison with the Lepr db/+ mice ( P<0.05). Compared with the Lepr db/+ mice, the Lepr db/ db mice renal exhibited glomerular hypertrophy, thickened basement membrane and capillary wall, the mesangial matrix expansion and mesangial cell hyperplasia. Compared with the Lepr db/+ mice, the serum level of GSH in the Lepr db/ db mice was decreased significantly ( P<0.05). The levels of MDA and concentrations of MCP-1, IL-1ß and TNF-α in serum of the Lepr db/ db mice were higher than those of the Lepr db/+ mice ( P<0.05). Compared with the Lepr db/+ mice, the expression of LIAS and Nrf2 protein in the Lepr db/ db mice renal were decreased ( P<0.05), while the expression of NF-κB protein was increased ( P<0.05). CONCLUSION: LIAS, Nrf2 and NF-κB might play significant roles through regulation of oxidative stress and inflammation in the renal injury of Lepr db/ db mice.


Subject(s)
Kidney/pathology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Receptors, Leptin/genetics , Sulfurtransferases/metabolism , Animals , Male , Mice , Mice, Knockout , Oxidative Stress
9.
J Cell Biochem ; 119(11): 8922-8936, 2018 11.
Article in English | MEDLINE | ID: mdl-29953665

ABSTRACT

Accumulating evidence suggests that autophagy plays a protective role in chondrocytes and prevents cartilage degeneration in osteoarthritis (OA). The objective of this study was to investigate the effect of diazoxide on chondrocyte death and cartilage degeneration and to determine whether these effects are correlated to autophagy in experimental OA. In this study, a cellular OA model was established by stimulating SW1353 cells with interleukin 1ß. A rat OA model was generated by transecting the anterior cruciate ligament combined with the resection of the medial menisci, followed by treatment with diazoxide or diazoxide combination with 3-methyladenine. The percentage of viable cells was evaluated using calcein-acetoxymethyl/propidium iodide double staining. The messenger RNA expression levels of collagen type II alpha 1 chain (COL2A1), matrix metalloproteinase 13 (MMP-13), TIMP metallopeptidase inhibitor 1 (TIMP-1), and a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5) were determined using quantitative real-time polymerase chain reaction. The cartilage thickness and joint space were evaluated using ultrasound. SW1353 cell degeneration and autophagosomes were observed using transmission electron microscopy. The expression levels of microtubule-associated protein 1 light chain 3 (LC3), beclin-1, P62, COL2A1, and MMP-13 were evaluated using immunofluorescence staining and Western blot analysis. Diazoxide significantly attenuated articular cartilage degeneration and SW1353 cell death in experimental OA. The restoration of autophagy was observed in the diazoxide-treated group. The beneficial effects of diazoxide were markedly blocked by 3-methyladenine. Diazoxide treatment also modulated the expression levels of OA-related biomarkers. These results demonstrated that diazoxide exerted a chondroprotective effect and attenuated cartilage degeneration by restoring autophagy via modulation of OA-related biomarkers in experimental OA. Diazoxide treatment might be a promising therapeutic approach to prevent the development of OA.


Subject(s)
Diazoxide/therapeutic use , Osteoarthritis/drug therapy , ADAMTS5 Protein/metabolism , Animals , Autophagy/drug effects , Biomarkers/blood , Blotting, Western , Cell Survival/drug effects , Chondrosarcoma/drug therapy , Chondrosarcoma/metabolism , Collagen Type II/metabolism , Humans , Male , Matrix Metalloproteinase 13/metabolism , Microscopy, Electron, Transmission , Osteoarthritis/metabolism , RNA, Messenger/metabolism , Rats
10.
Molecules ; 23(10)2018 Oct 06.
Article in English | MEDLINE | ID: mdl-30301216

ABSTRACT

Didymin (isosakuranetin 7-O-rutinoside) is an orally bioactive dietary flavonoid glycoside first found in citrus fruits. Traditionally, this flavonoid has long been used in Asian countries as a dietary antioxidant. Recent studies have provided newer insights into this pleiotropic compound, which could regulate multiple biological activities of many important signaling molecules in health and disease. Emerging data also presented the potential therapeutic application of dietary flavonoid glycoside didymin against cancer, neurological diseases, liver diseases, cardiovascular diseases, and other diseases. In this review, we briefly introduce the source and extraction methods of didymin, and summarize its potential therapeutic application in the treatment of various diseases, with an emphasis on molecular targets and mechanism that contributes to the observed therapeutic effects. The dietary flavonoid didymin can be used to affect health and disease with multiple therapeutic targets, and it is anticipated that this review will stimulate the future development of this potential dietary medicine.


Subject(s)
Antioxidants/therapeutic use , Citrus/chemistry , Flavonoids/therapeutic use , Glycosides/therapeutic use , Cardiovascular Diseases/diet therapy , Dietary Supplements , Flavonoids/chemistry , Glycosides/chemistry , Humans , Neoplasms/diet therapy , Nervous System Diseases/diet therapy
11.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 35(2): 237-243, 2018 04 25.
Article in Zh | MEDLINE | ID: mdl-29745529

ABSTRACT

The chemical extraction method was used to prepare the rat uterine decellularized scaffolds, and to investigate the feasibility of preparing the extracellular matrix (ECM) hydrogel. The rat uterus were collected and extracted by 1%sodium dodecyl sulfate (SDS), 3% TritonX-100 and 4% sodium deoxycholate (SDC) in sequence. Scanning electron microscopy, histochemical staining and immunohistochemistry was used to assess the degree of decellularization of rat uterine scaffold. The prepared decellularized scaffold was digested with pepsin to obtain a uterine ECM hydrogel, and the protein content of ECM was determined by specific ELISA kit. Meanwhile, the mechanical characteristic of ECM hydrogel was measured. The results showed that the chemical extraction method can effectively remove the cells effectively in the rat uterine decellularized scaffold, with the ECM composition preserved completely. ECM hydrogel contains a large amount of ECM protein and shows a good stability, which provides a suitable supporting material for the reconstruction of endometrium in vitro.

12.
Biomacromolecules ; 17(6): 2168-77, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27196997

ABSTRACT

Effective wound healing requires complicated, coordinated interactions and responses at protein, cellular, and tissue levels involving growth factor expression, cell proliferation, wound closure, granulation tissue formation, and vascularization. In this study, we develop a heparin-based coacervate consisting of poly(ethylene argininylaspartate digylceride) (PEAD) as a storage matrix, heparin as a bridge, and fibroblast growth factor-2 (FGF2) as a cargo (namely heparin-FGF2@PEAD) for wound healing. First, in vitro characterization demonstrates the loading efficiency and control release of FGF2 from the heparin-FGF2@PEAD coacervate. The following in vivo studies examine the wound healing efficiency of the heparin-FGF2@PEAD coacervate upon delivering FGF2 to full-thickness excisional skin wounds in vivo, in comparison with the other three control groups with saline, heparin@PEAD as vehicle, and free FGF2. Collective in vivo data show that controlled release of FGF2 to the wounds by the coacervate significantly accelerates the wound healing by promoting cell proliferation, stimulating the secretion of vascular endothelial growth factor (VEGF) for re-epithelization, collagen deposition, and granulation tissue formation, and enhancing the expression of platelet endothelial cell adhesion molecule (CD31) and alpha-smooth muscle actin (α-SMA) for blood vessel maturation. In parallel, no obvious wound healing effect is found for the control, vehicle, and free FGF2 groups, indicating the important role of the coavervate in the wound healing process. This work designs a suitable delivery system that can protect and release FGF2 in a sustained and controlled manner, which provides a promising therapeutic potential for topical treatment of wounds.


Subject(s)
Cell Proliferation/drug effects , Dermis/cytology , Fibroblast Growth Factor 2/administration & dosage , Heparin/chemistry , Regeneration/physiology , Vascular Endothelial Growth Factor A/metabolism , Wound Healing/drug effects , Administration, Cutaneous , Administration, Topical , Animals , Cells, Cultured , Dermis/drug effects , Fibroblast Growth Factor 2/chemistry , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Regeneration/drug effects
13.
Cardiovasc Drugs Ther ; 30(3): 247-61, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26947349

ABSTRACT

This study aims to investigate the preclinical performance and mechanism of a novel strategy of aFGF-loaded heparin-modified microbubbles (aFGF-HMB) combined with ultrasound-targeted microbubble destruction (UTMD) technique for diabetic cardiomyopathy (DCM) prevention. Type 1 diabetic rats were induced by streptozotocin. Twelve weeks after intervention, indexes from transthoracic echocardiography and cardiac catheterization showed that the left ventricular function in the aFGF-HMB/UTMD group was significantly improved compared with diabetes control (DM). From Picrosirius Red staining and TUNEL staining, the aFGF-HMB/UTMD group showed significant difference from the other groups. The cardiac collagen volume fraction (CVF) and myocardial cell apoptosis index (AI) in aFGF-HMB/UTMD group decreased to 7.2 % and 7.11 % respectively, compared with the DM group (CVF = 24.5 % and AI =20.3 % respectively). The results of myocardial microvascular density (MCD) also proved the strongest inhibition of aFGF-HMB/UTMD group on DCM progress. CD31 staining of aFGF-HMB/UTMD group reached 22 n/hrp, much higher than that of DM group (9 n/hrp). These results confirmed that the abnormalities including left ventricular dysfunction, myocardial fibrosis, cardiomyocytes apoptosis and microvascular rarefaction could be suppressed by twice weekly aFGF treatments for 12 consecutive weeks (free aFGF or aFGF-HMB+/-UTMD), with the strongest improvements observed in the aFGF-HMB/UTMD group (P < 0.05 vs free aFGF or aFGF-HMB). Western blot analyses of heart tissue further revealed the highest aFGF, anti-apoptosis protein (Bcl-2), VEGF-C, pAkt, pFoxo-3a levels and strongest reduction in pro-apoptosis proteins (Bax) level in aFGF-HMB/UTMD group. Overall, aFGF-HMB combined with UTMD technique might be developed as an effective strategy to prevent DCM in future clinical therapy.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Diabetic Cardiomyopathies/drug therapy , Fibroblast Growth Factor 1/administration & dosage , Heparin/administration & dosage , Hypoglycemic Agents/administration & dosage , Animals , Apoptosis/drug effects , Diabetes Mellitus, Experimental/diagnostic imaging , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/diagnostic imaging , Diabetes Mellitus, Type 1/metabolism , Diabetic Cardiomyopathies/diagnostic imaging , Diabetic Cardiomyopathies/metabolism , Echocardiography , Fibroblast Growth Factor 1/pharmacokinetics , Fibroblast Growth Factor 1/therapeutic use , Heart/diagnostic imaging , Heparin/chemistry , Heparin/therapeutic use , Hypoglycemic Agents/therapeutic use , Male , Microbubbles , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats, Sprague-Dawley , Ultrasonic Waves , bcl-2-Associated X Protein/metabolism
14.
AAPS PharmSciTech ; 17(6): 1376-1382, 2016 Dec.
Article in English | MEDLINE | ID: mdl-26753818

ABSTRACT

A critical issue for alcohol-induced liver disease (ALD) therapeutics is the lack of a highly efficient delivery system. In this study, a Puerarin-propylene glycol-liposome system was prepared for the purpose of targeting puerarin, an isoflavon, to the liver. Transmission electron microscope (TEM) results showed the liposomes to be spherical in shape with an average diameter of 182 nm with a polydispersity index of 0.239. The zeta potential of the particles was about -30 mV. The entrapment efficiency of puerarin was above 90%. MTT-based assay in HpeG2 cells showed no significant cytotoxicity in the presence of up to 25% concentration of the system containing 3% puerarin. In vivo performance of this system was studied in mice. Pharmacokinetics and distribution of puerarin-PG-liposome system was studied relative to puerarin solution at the same dose levels. The results show that puerarin-PG-liposome prolonged drug retention time and decreased elimination of puerarin in mice (AUC of liposome system and solution was 9.5 and 4.0 mg h L-1, respectively). Furthermore, propylene glycol (PG)-liposome system enhanced puerarin distribution into liver and spleen, while decreasing puerarin distribution in other tissues. Overall, the puerarin-PG-liposome system showed enhanced therapeutic effect in mice with ALD.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Isoflavones/chemistry , Isoflavones/pharmacology , Liposomes/chemistry , Liver/drug effects , Propylene Glycol/chemistry , Animals , Cell Line, Tumor , Drug Delivery Systems/methods , Ethanol/adverse effects , Hep G2 Cells , Humans , Isoflavones/pharmacokinetics , Liver/metabolism , Mice , Particle Size , Spleen/metabolism , Tissue Distribution
15.
Yao Xue Xue Bao ; 50(1): 99-103, 2015 Jan.
Article in Zh | MEDLINE | ID: mdl-25924483

ABSTRACT

Ultrasonic microbubbles were used to open blood-brain barriers (BBB) with a reversed and limited behavior feature in the study, which could improve the brain-targeted delivery of anti-tumor drugs. The glioma rat model was prepared. Low-frequency ultrasound was combined with microbubbles to affect the permeability of BBB compared with the permeability of independently administered Evans blue (EB) crossing BBB. Time point and length of ultrasound were investigated whether they affect the permeability of BBB and the damage of brain tissue. The effect of the growth time of glioma on BBB permeability was explored. Only glioma had a very little impact on BBB permeability. However, ultrasonic microbubbles opened the BBB with the features of temporary, limited and reversed behavior and improved EB and magnetic resonance imaging contrast agent penetrating BBB. A length of 30 s ultrasound is appropriate for opening BBB and no damage of brain tissue. Drugs should be injected before ultrasound so that they enter into brain as BBB opening. Ultrasonic microbubbles can open BBB effectively and safely, which improve drugs penetrating BBB under proper time point and length.


Subject(s)
Drug Delivery Systems , Glioma/drug therapy , Microbubbles , Animals , Blood-Brain Barrier , Contrast Media , Magnetic Resonance Imaging , Permeability , Rats , Ultrasonics
16.
Nanomedicine ; 10(4): 755-64, 2014 May.
Article in English | MEDLINE | ID: mdl-24200526

ABSTRACT

Lipid nanoparticles with solid matrix have been given increasing attention due to their biodegradable status and ability to entrap a variety of biologically active compounds. In this study, new phospholipid-based gelatin nanoparticles encapsulating basic fibroblast growth factor (bFGF) were developed to target the brain via nasal administration. Treatment effects were assessed by quantifying rotational behavior, monoamine neurotransmitter levels and tyrosine hydroxylase expression in 6-hydroxydopamine induced hemiparkinsonian rats. The gelatin nanostructured lipid carriers (GNLs) were prepared by a water-in-water emulsion method and then freeze-dried. The GNLs possessed better profile than gelatin nanoparticles (GNs), with particle size 143±1.14nm and Zeta potential -38.2±1.2mV. The intranasal GNLs efficiently enriched exogenous bFGF in olfactory bulb and striatum without adverse impact on the integrity of nasal mucosa and showed obvious therapeutic effects on hemiparkinsonian rats. Thus, GNLs are attractive carriers for nose-to-brain drug delivery, especially for unstable macromolecular drugs such as bFGF. FROM THE CLINICAL EDITOR: This team of authors reports the development of phospholipid-based gelatin nanoparticles encapsulating basic fibroblast growth factor to target the brain via intranasal administration. A rat model of hemiparkinsonism was applied demonstrating a good safety profile and an obvious therapeutic effect.


Subject(s)
Drug Carriers/pharmacology , Fibroblast Growth Factor 2/pharmacology , Gelatin/pharmacology , Lipids/pharmacology , Nanoparticles , Parkinson Disease, Secondary/drug therapy , Administration, Intranasal , Animals , Corpus Striatum/physiopathology , Drug Carriers/chemistry , Fibroblast Growth Factor 2/chemistry , Gelatin/chemistry , Lipids/chemistry , Olfactory Bulb/physiopathology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/physiopathology , Rats
17.
Drug Dev Ind Pharm ; 40(11): 1523-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24564824

ABSTRACT

Ca-alginate-poly-l-lysine-alginate (APA-Ca) and Ba-alginate-poly-l-lysine-alginate (APA-Ba) microcapsules were prepared and their thickness and surface were examined by light microscopy and scanning electron microscopy. Specifically, light microscopy with frozen section was used to visualize and quantify the thickness of APA membrane, and monitor temporal changes in the thickness of microcapsules during a month long culture in vitro. The section graph of APA microcapsule represents the accurate measurement of layer thickness of APA-Ca with diameter 900 ± 100 and 500 ± 100 µm at 6.01 ± 1.02 and 9.54 ± 2.42 µm (p < 0.05), and layer thickness of APA-Ba with diameter 900 ± 100 and 500 ± 100 µm at 5.47 ± 0.90 and 8.21 ± 1.97 µm (p < 0.05), regardless of the alginate composition used to generate the microcapsules. The microcapsule was stable during the culture for 30 days in vitro. Field emission scanning electron microscopy with freeze drying method was used to detect the surface and thickness of dried microcapsules. From the results, the outer surface of APA-Ca and APA-Ba membrane were smooth and dense, the film thickness of the APA-Ca was about 450-690 nm, while the APA-Ba was approximately 335 nm. In vivo experiment, little significant difference was seen in the change of film thickness of microcapsules in intrapertioneal site for 30 days after transplantation (p > 0.05), except that the recovery of APA-Ba was higher than the APA-Ca microcapsules. The paper showed an easy method to prepare APA-Ca and APA-Ba, and examine their thickness and surface, which could be utilized to study other types of microcapsules.


Subject(s)
Alginates/chemistry , Barium/chemistry , Calcium/chemistry , Capsules/chemistry , Polylysine/analogs & derivatives , Chemistry, Pharmaceutical , Drug Stability , Microscopy , Polylysine/chemistry , Surface Properties
18.
Adv Healthc Mater ; 13(13): e2304304, 2024 05.
Article in English | MEDLINE | ID: mdl-38306647

ABSTRACT

Acute lung injury (ALI) is a serious inflammatory disease that causes impairment of pulmonary function. Phenotypic modulation of macrophage in the lung using fibroblast growth factor 21 (FGF21) may be a potential strategy to alleviate lung inflammation. Consequently, achieving specific delivery of FGF21 to the inflamed lung and subsequent efficient FGF21 internalization by macrophages within the lung becomes critical for effective ALI treatment. Here, an apoptotic cell membrane-coated zirconium-based metal-organic framework UiO-66 is reported for precise pulmonary delivery of FGF21 (ACM@U-FGF21) whose design is inspired by the process of efferocytosis. ACM@U-FGF21 with apoptotic signals is recognized and internalized by phagocytes in the blood and macrophages in the lung, and then the intracellular ACM@U-FGF21 can inhibit the excessive secretion of pro-inflammatory cytokines by these cells to relieve the inflammation. Utilizing the homologous targeting properties inherited from the source cells and the spontaneous recruitment of immune cells to inflammatory sites, ACM@U-FGF21 can accumulate preferentially in the lung after injection. The results prove that ACM@U-FGF21 effectively reduces inflammatory damage to the lung by modulating lung macrophage polarization and suppressing the excessive secretion of pro-inflammatory cytokines by activated immune cells. This study demonstrates the usefulness of efferocytosis-inspired ACM@U-FGF21 in the treatment of ALI.


Subject(s)
Acute Lung Injury , Fibroblast Growth Factors , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Animals , Mice , Fibroblast Growth Factors/metabolism , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Phagocytosis/drug effects , Macrophages/metabolism , Macrophages/drug effects , Apoptosis/drug effects , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Mice, Inbred C57BL , Male , Zirconium/chemistry , Cytokines/metabolism , Lung/pathology , Lung/metabolism , RAW 264.7 Cells , Humans , Nanoparticles/chemistry
19.
Int J Pharm ; 656: 124115, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38614430

ABSTRACT

Fibroblast growth factor 21 (FGF21) shows great therapeutic potential in metabolic, neurodegenerative and inflammatory diseases. However, current FGF21 administration predominantly relies on injection rather than oral ingestion due to its limited stability and activity post-gastrointestinal transit, thereby hindering its clinical utility. Milk-derived exosomes (mEx) have emerged as a promising vehicle for oral drug delivery due to their ability to maintain structural integrity in the gastrointestinal milieu. To address the challenge associated with oral delivery of FGF21, we encapsulated FGF21 within mEx (mEx@FGF21) to protect its activity post-oral administration. Additionally, we modified the surface of mEx@FGF21 by introducing transferrin (TF) to enhance intestinal absorption and transport, designated TF-mEx@FGF21. In vitro results demonstrated that the surface modification of TF promoted FGF21 internalization by intestinal epithelial cells. Orally administered TF-mEx@FGF21 showed promising therapeutic effects in septic mice. This study represents a practicable strategy for advancing the clinical application of oral FGF21 delivery.


Subject(s)
Fibroblast Growth Factors , Inflammation , Sepsis , Fibroblast Growth Factors/administration & dosage , Animals , Administration, Oral , Mice , Sepsis/drug therapy , Inflammation/drug therapy , Male , Exosomes , Transferrin/administration & dosage , Transferrin/chemistry , Mice, Inbred C57BL , Milk , Humans , Drug Delivery Systems , Intestinal Absorption/drug effects
20.
Biomater Sci ; 12(4): 821-836, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38168805

ABSTRACT

Islet transplantation holds significant promise as a curative approach for type 1 diabetes (T1D). However, the transition of islet transplantation from the experimental phase to widespread clinical implementation has not occurred yet. One major hurdle in this field is the challenge of insufficient vascularization and subsequent early loss of transplanted islets, especially in non-intraportal transplantation sites. The establishment of a fully functional vascular system following transplantation is crucial for the survival and secretion function of islet grafts. This vascular network not only ensures the delivery of oxygen and nutrients, but also plays a critical role in insulin release and the timely removal of metabolic waste from the grafts. This review summarizes recent advances in effective strategies to improve graft revascularization and enhance islet survival. These advancements include the local release and regulation of angiogenic factors (e.g., vascular endothelial growth factor, VEGF), co-transplantation of vascular fragments, and pre-vascularization of the graft site. These innovative approaches pave the way for the development of effective islet transplantation therapies for individuals with T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans Transplantation , Islets of Langerhans , Humans , Islets of Langerhans/metabolism , Diabetes Mellitus, Type 1/surgery , Biocompatible Materials , Vascular Endothelial Growth Factor A/metabolism , Islets of Langerhans Transplantation/physiology , Neovascularization, Physiologic
SELECTION OF CITATIONS
SEARCH DETAIL