Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.126
Filter
1.
Anim Reprod Sci ; 263: 107437, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38395011

ABSTRACT

Short-term sperm storage is a straightforward and cost-effective method of managing logistics in large scale fish hatchery operations but may result in decline in sperm quality. For effective artificial reproduction of fish, use of an appropriate additive to optimize sperm storage conditions is essential. In this study, it was investigated the effect of purified seminal plasma transferrin (Tf) at 10 µg/ml on relevant parameters in common carp Cyprinus carpio sperm during short-term storage. We compared sperm motility and curvilinear velocity, adenosine triphosphate (ATP) content and DNA fragmentation of fresh spermatozoa to that stored for 24, 48, 72, and 144 h with or without Tf. The percentage of motile cells and the curvilinear velocity of spermatozoa in stored samples for 72 h with transferrin supplementation were greater compared to samples with no added protein. The ATP content in samples without added transferrin was reduced (P < 0.05) after 72 h of storage, in contrast to the levels observed in transferrin-supplemented sperm. A time-dependent increase in DNA fragmentation was observed. Significantly lower DNA damage, expressed as percent tail DNA (10.99 ±â€¯1.28) and olive tail moment (0.54 ±â€¯0.12), was recorded in Tf-supplemented samples stored for 48 h compared to that with no Tf. Hence, it is concluded that the beneficial effects of transferrin on common carp sperm could serve as an additional tool for developing and enhancing short-term sperm preservation procedures commonly used in aquaculture.


Subject(s)
Carps , Semen Preservation , Male , Animals , Semen/metabolism , Transferrin/pharmacology , Adenosine Triphosphate/metabolism , Sperm Motility , Spermatozoa , Semen Preservation/veterinary , Semen Preservation/methods , DNA/metabolism
2.
Clin Sci (Lond) ; 137(18): 1431-1449, 2023 09 27.
Article in English | MEDLINE | ID: mdl-37694282

ABSTRACT

Skeletal mandibular hypoplasia (SMH) is one of the most common skeletal craniofacial deformities in orthodontics, which was often accompanied by impaired chondrogenesis and increasing apoptosis of condylar chondrocytes. Therefore, protecting chondrocytes from apoptosis and promoting chondrogenesis in condylar growth is vital for treatment of SMH patients. Transferrin (TF) was highly expressed in condylar cartilage of newborn mice and was gradually declined as the condyle ceased growing. Interestingly, serum level of TF in SMH patients was significantly lower than normal subjects. Hence, the aim of our study was to investigate the effect of TF on survival and differentiation of chondrocytes and condylar growth. First, we found that TF protected chondrogenic cell line ATDC5 cells from hypoxia-induced apoptosis and promoted proliferation and chondrogenic differentiation in vitro. Second, TF promoted chondrogenic differentiation and survival through activating autophagic flux. Inhibiting autophagic flux markedly blocked the effects of TF. Third, TF significantly activated ULK1-ATG16L1 axis. Silencing either transferrin receptor (TFRC), ULK1/2 or ATG16 significantly blocked the autophagic flux induced by TF, as well as its effect on anti-apoptosis and chondrogenic differentiation. Furthermore, we established an organoid culture model of mandible ex vivo and found that TF significantly promoted condylar growth. Taken together, our study unraveled a novel function of TF in condylar growth that TF protected chondrocytes from hypoxia-induced apoptosis and promoted chondrogenic differentiation through inducing autophagy via ULK1-ATG16L1 axis, which demonstrated that TF could be a novel growth factor of condylar growth and shed new light on developing treatment strategy of SMH patients.


Subject(s)
Chondrogenesis , Transferrin , Humans , Animals , Mice , Transferrin/pharmacology , Cell Differentiation , Autophagy , Hypoxia , Autophagy-Related Protein-1 Homolog , Intracellular Signaling Peptides and Proteins , Autophagy-Related Proteins
3.
Haematologica ; 108(10): 2703-2714, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37165842

ABSTRACT

ß-thalassemia is an inherited anemia characterized by ineffective erythropoiesis. Blood transfusions are required for survival in transfusion-dependent ß-thalassemia and are also occasionally needed in patients with non-transfusion-dependent ß-thalassemia. Patients with transfusion-dependent b-thalassemia often have elevated transferrin saturation (TSAT) and non-transferrin-bound iron (NTBI) levels, which can lead to organ iron overload, oxidative stress, and vascular damage. Vamifeport is an oral ferroportin inhibitor that was previously shown to ameliorate anemia, ineffective erythropoiesis, and dysregulated iron homeostasis in the Hbbth3/+ mouse model of ß-thalassemia, under non-transfused conditions. Our study aimed to assess the effects of oral vamifeport on iron-related parameters (including plasma NTBI levels) and ineffective erythropoiesis following blood transfusions in Hbbth3/+ mice. A single dose of vamifeport prevented the transient transfusion-mediated NTBI increase in Hbbth3/+ mice. Compared with vehicle treatment, vamifeport significantly increased hemoglobin levels and red blood cell counts in transfused mice. Vamifeport treatment also significantly improved ineffective erythropoiesis in the spleens of Hbbth3/+ mice, with additive effects observed when treatment was combined with repeated transfusions. Vamifeport corrected leukocyte counts and significantly improved iron-related parameters (serum transferrin, TSAT and erythropoietin levels) versus vehicle treatment in Hbbth3/+ mice, irrespective of transfusion status. In summary, vamifeport prevented transfusion-mediated NTBI formation in Hbbth3/+ mice. When given alone or combined with blood transfusions, vamifeport also ameliorated anemia, ineffective erythropoiesis, and dysregulated iron homeostasis. Administering vamifeport together with repeated blood transfusions additively ameliorated anemia and ineffective erythropoiesis in this mouse model, providing preclinical proof-of-concept for the efficacy of combining vamifeport with blood transfusions in ß-thalassemia.


Subject(s)
Iron Overload , Thalassemia , beta-Thalassemia , Humans , Mice , Animals , beta-Thalassemia/drug therapy , Erythropoiesis , Iron/metabolism , Transferrin/pharmacology , Iron Overload/drug therapy , Iron Overload/etiology , Blood Transfusion
4.
ACS Appl Mater Interfaces ; 15(24): 28879-28890, 2023 Jun 21.
Article in English | MEDLINE | ID: mdl-37249181

ABSTRACT

Ferroptosis, an iron-dependent cell death driven by the lethal levels of lipid peroxidation (LPO), becomes a promising anticancer strategy. However, the anticancer efficacy of ferroptosis is often hindered by the activation of nuclear factor erythrocyte 2-associated factor 2 (Nrf2), which is an indispensable regulator of the cellular antioxidant balance by preventing the accumulation of intracellular reactive oxygen species (ROS). Herein, we present a rational design of a Tf-targeted cascade nanoplatform TPM@AM based on mesoporous polydopamine (MPDA) co-encapsulating a ferroptosis inducer (artesunate, ART) and an Nrf2-specific inhibitor (ML385) to enhance intracellular ROS and therefore amplify ferrotherapy. Transferrin (Tf) can specifically recognize the transferrin receptor (TfR) on the surface of the cell membrane, which binds and transports iron into cells. When TPM@AM is endocytosed, the high-acid tumor microenvironment and laser irradiation trigger the collapse of MPDA to release ART and ML385. Furthermore, MPDA endows the nanoplatform with photothermal capability. The nanoplatform exhibits high efficiency for synergistic tumor suppression, representing a spatiotemporal controllable therapeutic strategy for precise synergistic cancer therapy.


Subject(s)
Ferroptosis , Iron/metabolism , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Transferrin/pharmacology
5.
ACS Appl Mater Interfaces ; 15(22): 26484-26495, 2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37218712

ABSTRACT

Cisplatin (CDDP) is a widely used chemotherapeutic drug with proven efficacy for treating tumors. However, its use has been associated with severe side effects and eventually leads to drug resistance, thus limiting its clinical application in patients with ovarian cancer (OC). Herein, we aimed to investigate the success rate of reversing cisplatin resistance using a synthetic, multitargeted nanodrug delivery system comprising a Mn-based metal-organic framework (Mn-MOF) containing niraparib (Nira) and CDDP alongside transferrin (Tf) conjugated to the surface (Tf-Mn-MOF@Nira@CDDP; MNCT). Our results revealed that MNCT can target the tumor site, consume glutathione (GSH), which is highly expressed in drug-resistant cells, and then decompose to release the encapsulated Nira and CDDP. Nira and CDDP play a synergistic role in increasing DNA damage and apoptosis, exhibiting excellent antiproliferation, migration, and invasion activities. In addition, MNCT significantly inhibited tumor growth in tumor-bearing mice and exhibited excellent biocompatibility without side effects. Furthermore, it depleted GSH, downregulated multidrug-resistant transporter protein (MDR) expression, and upregulated tumor suppressor protein phosphatase and tensin homolog (PTEN) expression, consequently reducing DNA damage repair and reversing cisplatin resistance. These results indicate that multitargeted nanodrug delivery systems can provide a promising clinical approach to overcoming cisplatin resistance. This study provides an experimental basis for further investigation into multitargeted nanodrug delivery systems to reverse cisplatin resistance in patients with OC.


Subject(s)
Antineoplastic Agents , Nanoparticles , Ovarian Neoplasms , Humans , Female , Animals , Mice , Cisplatin/pharmacology , Cisplatin/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Transferrin/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Nanoparticles/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm
6.
Acta Biomater ; 166: 524-535, 2023 08.
Article in English | MEDLINE | ID: mdl-37088161

ABSTRACT

Cerebral ischemia-reperfusion injury (CIRI) is a complex pathological condition with high mortality. In particular, reperfusion can stimulate overproduction of reactive oxygen species (ROS) and activation of inflammation, causing severe secondary injuries to the brain. Despite tremendous efforts, it remains urgent to rationally design antioxidative agents with straightforward and efficient ROS scavenging capability. Herein, a potent antioxidative agent was explored based on iridium oxide nano-agglomerates (Tf-IrO2 NAs) via the facile transferrin (Tf)-templated biomineralization approach, and innovatively applied to treat CIRI. Containing some small-size IrO2 aggregates, these NAs possess intrinsic hydroxyl radicals (•OH)-scavenging ability and multifarious enzyme activities, such as catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GPx). Moreover, they also showed improved blood-brain barrier (BBB) penetration and enhanced accumulation in the ischemic brain via Tf receptor-mediated transcytosis. Therefore, Tf-IrO2 NAs achieved robust in vitro anti-inflammatory and cytoprotection effects against oxidative stress. Importantly, mice were effectively protected against CIRI by enhanced ROS scavenging activity in vivo, and the therapeutic mechanism was systematically verified. These findings broaden the idea of expanding Ir-based NAs as potent antioxidative agents to treat CIRI and other ROS-mediated diseases. STATEMENT OF SIGNIFICANCE: (1) The ROS-scavenging activities of IrO2 are demonstrated comprehensively, which enriched the family of nano-antioxidants. (2) The engineering Tf-IrO2 nano-agglomerates present unique multifarious enzyme activities and simultaneous transferrin targeting and BBB crossing ability for cerebral ischemia-reperfusion injury therapy. (3) This work may open an avenue to enable the use of IrO2 to alleviate ROS-mediated inflammatory and brain injury diseases.


Subject(s)
Brain Ischemia , Reperfusion Injury , Mice , Animals , Reactive Oxygen Species/metabolism , Transferrin/pharmacology , Iridium/pharmacology , Iridium/therapeutic use , Brain Ischemia/drug therapy , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Oxidative Stress , Antioxidants/pharmacology , Reperfusion
7.
Reprod Fertil Dev ; 35(5): 363-374, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36780707

ABSTRACT

CONTEXT: One of the main problems of porcine in vitro maturation (IVM) is incomplete cytoplasmatic maturation. Nuclear and cytoplasmic maturation will determine the future success of fertilisation and embryo development. Insulin-transferrin-selenium (ITS) has insulin-like and antioxidant effects, and metformin (M) is an insulin-sensitiser and antioxidant drug. AIMS: To assess the effects of adding ITS and/or M in porcine IVM media on cytoplasmic maturation and early embryo development. METHODS: Cumulus -oocyte complexes (COC) were IVM with M (10-4 M), ITS (0.1% v/v), M+ITS or no adding (Control). KEY RESULTS: ITS increased glucose consumption compared to Control and M (P <0.01), and M+ITS did not differ from ITS or Control. Redox balance: M, ITS and M+ITS increased glutathione (P <0.01) and decreased lipid peroxidation (P <0.005). The viability of cumulus cells by flow cytometry increased with M (P <0.005) and decreased with ITS (P <0.001); M+ITS did not differ from Control. After IVF, M increased penetration and decreased male pronucleus (P <0.05). Embryo development: cleavage increased with M (P <0.05), and blastocysts increased with ITS and M+ITS (P <0.05). The number of blastocyst cells increased with ITS (P <0.05). CONCLUSIONS: Adding ITS and M+ITS to porcine IVM media benefits embryo development to blastocysts, but ITS alone has better effects than M+ITS. IMPLICATIONS: ITS is an excellent tool to improve IVM and embryo development after IVF in pigs.


Subject(s)
Metformin , Selenium , Male , Animals , Swine , Selenium/pharmacology , Insulin/pharmacology , Transferrin/pharmacology , Metformin/pharmacology , Fertilization in Vitro/veterinary , Oocytes , Embryonic Development , Blastocyst , In Vitro Oocyte Maturation Techniques/veterinary
8.
Adv Mater ; 35(10): e2209603, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36524741

ABSTRACT

Glutathione (GSH)-activatable probes hold great promise for in vivo cancer imaging, but are restricted by their dependence on non-selective intracellular GSH enrichment and uncontrollable background noise. Here, a holographically activatable nanoprobe caging manganese tetraoxide is shown for tumor-selective contrast enhancement in magnetic resonance imaging (MRI) through cooperative GSH/albumin-mediated cascade signal amplification in tumors and rapid elimination in normal tissues. Once targeting tumors, the endocytosed nanoprobe effectively senses the lysosomal microenvironment to undergo instantaneous decomposition into Mn2+ with threshold GSH concentration of ≈ 0.12 mm for brightening MRI signals, thus achieving high contrast tumor imaging and flexible monitoring of GSH-relevant cisplatin resistance during chemotherapy. Upon efficient up-regulation of extracellular GSH in tumor via exogenous injection, the relaxivity-silent interstitial nanoprobe remarkably evolves into Mn2+ that are further captured/retained and re-activated into ultrahigh-relaxivity-capable complex by stromal albumin in the tumor, and simultaneously allows the renal clearance of off-targeted nanoprobe in the form of Mn2+ via lymphatic vessels for suppressing background noise to distinguish tiny liver metastasis. These findings demonstrate the concept of holographic tumor activation via both tumor GSH/albumin-mediated cascade signal amplification and simultaneous background suppression for precise tumor malignancy detection, surveillance, and surgical guidance.


Subject(s)
Albumins , Glutathione , Magnetic Resonance Imaging , Metal Nanoparticles , Molecular Probes , Neoplasms , Glutathione/administration & dosage , Glutathione/pharmacokinetics , Glutathione/pharmacology , Molecular Probes/administration & dosage , Molecular Probes/pharmacokinetics , Molecular Probes/pharmacology , Albumins/administration & dosage , Albumins/pharmacokinetics , Albumins/pharmacology , Magnetic Resonance Imaging/methods , Contrast Media/administration & dosage , Contrast Media/pharmacokinetics , Contrast Media/pharmacology , Image Enhancement/methods , Holography/methods , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology , Metal Nanoparticles/administration & dosage , Transferrin/administration & dosage , Transferrin/pharmacokinetics , Transferrin/pharmacology , Tissue Distribution , A549 Cells , Humans , Animals , Mice , Mice, Inbred BALB C , Mice, Nude , Cisplatin/administration & dosage , Cisplatin/pharmacokinetics , Cisplatin/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology
9.
J Pharm Pharmacol ; 75(1): 49-56, 2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36173891

ABSTRACT

OBJECTIVES: Resveratrol (Res) was a naturally occurring polyphenol compound. It has various beneficial effects, including anti-inflammatory, anti-oxidant and anti-cancer effects. However, the anti-cancer activity was hindered by its low targeting and drug release performance. Thus, we synthesized transferrin-cathepsin B cleavable peptide modified mesoporous silica nanoparticle encapsulated Res (Tf-Res-MSN). METHODS: Res was encapsulated in mesoporous silica nanoparticles (MSN), which was a kind of drug carrier complex. Tf was modified to recognize the cancer cells. Cathepsin B cleavable peptide (Pep) was used to combine Res-MSN complex and Tf to construct the final product. Pep was used as linker and trigger for Res release. KEY FINDINGS: The smart nanocarriers were increased the drug release performance of Res in human breast cancer (MCF-7) cells. The physicochemical properties of Tf-Res-MSN were assessed by zeta potential, UV-Prove, diffraction scanning calorimetry (DSC), nitrogen physisorption analysis and transmission electron microscope (TEM). MTT assay, AO and Annexin V-FITC/PI staining were performed to explore the anti-tumour activity of Tf-Res-MSN. The results showed that Tf-Res-MSN significantly decreased cell viability and increased cell apoptosis. The inhibition rate and apoptotic rate of Tf-Res-MSN in MCF-7 cells were 95.75% and 80.8%, respectively. CONCLUSION: Our study demonstrated that Tf-Res-MSN was a valuable technique with potential value in breast cancer applications.


Subject(s)
Breast Neoplasms , Nanoparticles , Humans , Female , Drug Delivery Systems/methods , MCF-7 Cells , Resveratrol/pharmacology , Cathepsin B/pharmacology , Silicon Dioxide , Transferrin/pharmacology , Transferrin/therapeutic use , Drug Carriers/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Peptides/pharmacology , Nanoparticles/chemistry , Apoptosis , Porosity
10.
Int J Mol Sci ; 23(21)2022 Oct 22.
Article in English | MEDLINE | ID: mdl-36361544

ABSTRACT

Iron is essential for retinal metabolism, but an excess of ferrous iron causes oxidative stress. In glaucomatous eyes, retinal ganglion cell (RGC) death has been associated with dysregulation of iron homeostasis. Transferrin (TF) is an endogenous iron transporter that controls ocular iron levels. Intraocular administration of TF is neuroprotective in various models of retinal degeneration, preventing iron overload and reducing iron-induced oxidative stress. Herein, we assessed the protective effects of TF on RGC survival, using ex vivo rat retinal explants exposed to iron, NMDA-induced excitotoxicity, or CoCl2-induced hypoxia, and an in vivo rat model of ocular hypertension (OHT). TF significantly preserved RGCs against FeSO4-induced toxicity, NMDA-induced excitotoxicity, and CoCl2-induced hypoxia. TF protected RGCs from apoptosis, ferroptosis, and necrosis. In OHT rats, TF reduced RGC loss by about 70% compared to vehicle-treated animals and preserved about 47% of the axons. Finally, increased iron staining was shown in the retina of a glaucoma patient's eye as compared to non-glaucomatous eyes. These results indicate that TF can interfere with different cell-death mechanisms involved in glaucoma pathogenesis and demonstrate the ability of TF to protect RGCs exposed to elevated IOP. Altogether, these results suggest that TF is a promising treatment against glaucoma neuropathy.


Subject(s)
Glaucoma , Neuroprotective Agents , Ocular Hypertension , Animals , Rats , Disease Models, Animal , Glaucoma/metabolism , Hypoxia , Intraocular Pressure , Iron/metabolism , N-Methylaspartate , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Ocular Hypertension/metabolism , Transferrin/pharmacology
11.
Int J Mol Sci ; 23(19)2022 Sep 23.
Article in English | MEDLINE | ID: mdl-36232527

ABSTRACT

Egg-derived peptides play important roles in insulin secretion and sensitivity, oxidative stress, and inflammation, suggesting their possible involvement in obesity management. Hence, the aim of this study is to explore the alleviating effects of IRW (lle-Arg-Trp) and IQW (lle-Gln-Trp) on obesity via the mouse model induced by a high-fat diet. The entire experimental period lasted eight weeks. The results demonstrated that IQW prevented weight gain (6.52%), decreased the glucose, low-density lipoprotein (LDL), malonaldehyde, triglycerides, total cholesterol (TC), and leptin levels, and increased the concentration of adiponectin (p < 0.05, n = 8). Although IRW failed to prevent weight gain, it reduced the concentration of glucose, high-density lipoprotein (HDL), LDL, and leptin, and increased the concentration of adiponectin (p < 0.05, n = 8). Moreover, IRW and IQW increased glucose tolerance and insulin resistance based on the results of the intraperitoneal glucose test and insulin tolerance test (p < 0.05, n = 8). The quantitative polymerase chain reaction results revealed that IRW and IQW downregulated the mRNA expression of DGAT1 (Diacylglycerol O-Acyltransferase 1), DGAT2 (Diacylglycerol O-Acyltransferase 2), TNF-α, IL-6, and IL-1ß of liver tissue (p < 0.05, n = 8). The results of the 16S ribosomal RNA amplicon sequencing showed that IQW and IRW tended to reduce the relative abundance of Firmicutes and Parabacteroides, and that IRW enhanced the abundance of Bacteroides (p < 0.05, n = 8). Collectively, IRW and IQW supplementation could alleviate the progression of obesity due to the fact that the supplementation reduced lipid deposition, maintained energy balance, and reprogrammed gut microbiota.


Subject(s)
Gastrointestinal Microbiome , Insulins , Adiponectin/metabolism , Animals , Cholesterol/pharmacology , Diacylglycerol O-Acyltransferase/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Egg Proteins/metabolism , Glucose/pharmacology , Insulins/metabolism , Interleukin-6/metabolism , Leptin/metabolism , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Malondialdehyde , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Obesity/etiology , Peptides/pharmacology , RNA, Messenger , RNA, Ribosomal, 16S/metabolism , Transferrin/pharmacology , Triglycerides/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Weight Gain
12.
Nanoscale ; 14(35): 12773-12788, 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-36001382

ABSTRACT

Spatiotemporal targeting of anti-glioma drugs remains a pressing issue in glioblastoma (GBM) treatment. We challenge this issue by developing a minimally invasive in situ implantable hydrogel implant comprising transferrin-targeted temozolomide-miltefosine nanovesicles in the surgically resected GBM cavity (tumour bed). Injection of the "nanovesicle in hydrogel system" in orthotopic GBM-bearing mice improved drug penetration into the peri-cavitary region (∼4.5 mm in depth) with the potential to act as a bridge therapy in the immediate postoperative period, before the initiation of adjuvant radiotherapy. The controlled and sustained release of temozolomide over a month in the surgical cavity eradicated the microscopic GBM cells present within the tumour bed, thereby augmenting the efficacy of adjuvant therapy. The drug (temozolomide and miltefosine) combination was tolerable and efficiently inhibited tumour growth, causing significant prolongation of the survival of tumour-bearing mice compared to that with the free drug. Direct implantation at the target site in the brain resulted in spatiotemporal anti-glioma activity with minimal extracranial and systemic distribution. Nanovesicle in flexible hydrogel systems can be used as potential platforms for the post-surgical management of GBM before initiating adjuvant radiation therapy.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Nanoparticles , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Delayed-Action Preparations/therapeutic use , Glioblastoma/pathology , Glioma/drug therapy , Hydrogels/pharmacology , Hydrogels/therapeutic use , Mice , Phosphorylcholine/analogs & derivatives , Polymers/therapeutic use , Temozolomide/pharmacology , Temozolomide/therapeutic use , Transferrin/pharmacology , Xenograft Model Antitumor Assays
13.
Transfus Apher Sci ; 61(2): 103319, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34801431

ABSTRACT

BACKGROUND: In transfusion-related iron overload, haem-derived iron accumulation in monocytes/macrophages is the initial event. When iron loading exceeds the ferritin storage capacity, iron is released into the plasma. When iron loading exceeds transferrin binding capacity, labile, non-transferrin-bound iron (NTBI) appears and causes organ injury. Haemin-induced cell death has already been investigated; however, whether NTBI induces cell death in monocytes/macrophages remains unclear. MATERIAL AND METHODS: Human monocytic THP-1 cells were treated with haemin or NTBI, particularly ferric ammonium citrate (FAC) or ferrous ammonium sulfate (FAS). The intracellular labile iron pool (LIP) was measured using an iron-sensitive fluorescent probe. Ferritin expression was measured by western blotting. RESULTS: LIP was elevated after haemin treatment but not after FAC or FAS treatment. Reactive oxygen species (ROS) generation and cell death induction were remarkable after haemin treatment but not after FAC or FAS treatment. Ferritin expression was not different between the FAC and haemin treatments. The combination of an iron chelator and a ferroptosis inhibitor significantly augmented the suppression of haemin cytotoxicity (p = 0.011). DISCUSSION: The difference in LIP suggests the different iron traffic mechanisms for haem-derived iron and NTBI. The Combination of iron chelators and antioxidants is beneficial for iron overload therapy.


Subject(s)
Iron Overload , Iron , Cell Death , Ferritins , Hemin/pharmacology , Humans , Iron/metabolism , Reactive Oxygen Species/metabolism , Transferrin/metabolism , Transferrin/pharmacology
14.
Front Endocrinol (Lausanne) ; 13: 1009507, 2022.
Article in English | MEDLINE | ID: mdl-36778593

ABSTRACT

Background and objective: Glycation is a common post-transcriptional modification of proteins. Previous studies have shown that advanced glycation end modified transferrin (AGE-Tf) levels in diabetic rat kidney tissues were increased; however, its role in diabetic nephropathy remains unclear. In this study, differences in glycation degree and Tf sites induced by differing high glucose concentrations in vitro and the effect on total iron binding capacity (TIBC) were observed. Moreover, the effect of AGE-Tf on human renal tubular epithelial cells (HK-2) was investigated. Methods: In vitro Tf was incubated with increasing glucose concentrations (0 mM, 5.6 mM, 11.1 mM, 33.3 mM, 100 mM, 500 mM, and 1,000 mM) for AGE-Tf. Differences in AGE-Tf glycation degree and TIBC level were analyzed via colorimetric method. The AGE-Tf glycation sites were identified with LC-MS/MS. HK-2 cells were treated with AGE-Tf prepared with different glucose concentrations (33.3 mM and 500 mM) in vitro. The effects of AGE-Tf on HK-2 cell viability, proliferation, oxidative stress index, and Tf receptor expression levels were then observed. Results: With increasing glucose concentrations (100 mM, 500 mM, and 1,000 mM) in vitro, Tf glycation degree was significantly increased. The TIBC levels of AGE-Tf were decreased significantly with increasing glucose concentrations (33.3 mM, 100 mM, 500 mM, and 1,000 mM). Four glycated modification sites in Tf and 17 glycated modification sites were detected in AGE-Tf (500 mM) by LC-MS/MS. The structural types of AGEs were CML, G-H1, FL-1H2O, FL, and MG-H1. No significant differences were found in the survival rate of HK-2 cells among the AGE-Tf (500 mM), AGE-Tf (33.3 mM), and Tf groups (all p > 0.05). The apoptosis rate of HK-2 cells in the AGE-Tf (500 mM) group was significantly higher than that in the AGE-Tf (33.3 mM) group. Additionally, both of them were significantly higher than that in the Tf group (both p < 0.05). The MDA levels of HK-2 cells in the AGE-Tf (500 mM) and AGE-Tf (33.3 mM) groups were higher than that in the Tf group, but not significantly (both p > 0.05). The T-AOC level of HK-2 in the AGE-Tf (500 mM) group was significantly lower than that in the AGE-Tf (33.3 mM) and Tf groups (both p < 0.001). The GSH level of HK-2 cells in the AGE-Tf (500 mM) group was significantly lower than that in the Tf group (p < 0.05). The expression level of TfR in the AGE-Tf (500 mM) group was also significantly lower than that in the Tf group (p < 0.05). Conclusion: The degree and sites of Tf glycation were increased in vitro secondary to high-glucose exposure; however, the binding ability of Tf to iron decreased gradually. After HK-2 was stimulated by AGE-Tf in vitro, the apoptosis of cells was increased, antioxidant capacity was decreased, and TfR expression levels were downregulated.


Subject(s)
Maillard Reaction , Transferrin , Rats , Animals , Humans , Transferrin/pharmacology , Chromatography, Liquid , Tandem Mass Spectrometry , Iron , Glucose/pharmacology
15.
J Nanobiotechnology ; 19(1): 289, 2021 Sep 26.
Article in English | MEDLINE | ID: mdl-34565383

ABSTRACT

BACKGROUND: Effective treatment of glioma requires a nanocarrier that can cross the blood-brain barrier (BBB) to target the tumor lesion. In the current study, elemene (ELE) and cabazitaxel (CTX) liposomes were prepared by conjugating liposomes with transferrin (Tf) and embedding the cell membrane proteins of RG2 glioma cells into liposomes (active-targeting biomimetic liposomes, Tf-ELE/CTX@BLIP), which exhibited effective BBB infiltration to target glioma. RESULTS: The findings showed that Tf-ELE/CTX@BLIP was highly stable. The liposomes exhibited highly significant homologous targeting and immune evasion in vitro and a 5.83-fold intake rate compared with classical liposome (ELE/CTX@LIP). Bioluminescence imaging showed increased drug accumulation in the brain and increased tumor penetration of Tf-ELE/CTX@BLIP in orthotopic glioma model nude mice. Findings from in vivo studies indicated that the antitumor effect of the Tf-ELE/CTX@BLIP led to increased survival time and decreased tumor volume in mice. The average tumor fluorescence intensity after intravenous administration of Tf-ELE/CTX@BLIP was 65.2, 12.5, 22.1, 6.6, 2.6, 1.5 times less compared with that of the control, CTX solution, ELE solution, ELE/CTX@LIP, ELE/CTX@BLIP, Tf-ELE/CTX@LIP groups, respectively. Histopathological analysis showed that Tf-ELE/CTX@BLIP were less toxic compared with administration of the CTX solution. CONCLUSION: These findings indicate that the active-targeting biomimetic liposome, Tf-ELE/CTX@BLIP, is a promising nanoplatform for delivery of drugs to gliomas.


Subject(s)
Biomimetics/methods , Glioma/therapy , Liposomes/administration & dosage , Sesquiterpenes/pharmacology , Taxoids/pharmacokinetics , Transferrin/metabolism , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Brain , Cell Line, Tumor , Drug Delivery Systems/methods , Glioma/pathology , Mice , Mice, Nude , Sesquiterpenes/metabolism , Sesquiterpenes/therapeutic use , Taxoids/metabolism , Taxoids/therapeutic use , Transferrin/pharmacology , Transferrin/therapeutic use
16.
Cells ; 10(7)2021 07 02.
Article in English | MEDLINE | ID: mdl-34359837

ABSTRACT

Induced pluripotent stem (iPS) cells constitute a perfect tool to study human embryo development processes such as myogenesis, thanks to their ability to differentiate into three germ layers. Currently, many protocols to obtain myogenic cells have been described in the literature. They differ in many aspects, such as media components, including signaling modulators, feeder layer constituents, and duration of culture. In our study, we compared three different myogenic differentiation protocols to verify, side by side, their efficiency. Protocol I was based on embryonic bodies differentiation induction, ITS addition, and selection with adhesion to collagen I type. Protocol II was based on strong myogenic induction at the embryonic bodies step with BIO, forskolin, and bFGF, whereas cells in Protocol III were cultured in monolayers in three special media, leading to WNT activation and TGF-ß and BMP signaling inhibition. Myogenic induction was confirmed by the hierarchical expression of myogenic regulatory factors MYF5, MYOD, MYF6 and MYOG, as well as the expression of myotubes markers MYH3 and MYH2, in each protocol. Our results revealed that Protocol III is the most efficient in obtaining myogenic cells. Furthermore, our results indicated that CD56 is not a specific marker for the evaluation of myogenic differentiation.


Subject(s)
Cell Culture Techniques , Culture Media/pharmacology , Embryoid Bodies/drug effects , Fibroblasts/drug effects , Induced Pluripotent Stem Cells/drug effects , Muscle Development/drug effects , Muscle Fibers, Skeletal/drug effects , Biomarkers/metabolism , Cell Differentiation/drug effects , Colforsin/pharmacology , Collagen Type I/pharmacology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , Humans , Indoles/pharmacology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Insulin/pharmacology , Muscle Development/genetics , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Myogenin/genetics , Myogenin/metabolism , Oximes/pharmacology , Selenium/pharmacology , Transferrin/pharmacology
17.
Exp Hematol ; 99: 12-20.e3, 2021 07.
Article in English | MEDLINE | ID: mdl-34077792

ABSTRACT

Red blood cell production, or erythropoiesis, is a proliferative process that requires tight regulation. Erythropoietin (Epo) is a glycoprotein cytokine that plays a major role in erythropoiesis by triggering erythroid progenitors/precursors of varying sensitivity. The concentration of Epo in bone marrow is hypothesized to be suboptimal, and the survival of erythroid cells has been suggested to depend on Epo sensitivity. However, the key factors that control Epo sensitivity remain unknown. Two types of transferrin receptors (TfRs), TfR1 and TfR2, are known to play a role in iron uptake in erythroid cells. Here, we hypothesized that TfRs may additionally modulate Epo sensitivity during erythropoiesis by modulating Epo receptor (EpoR) signaling. Using an Epo-sensitive UT-7 (UT7/Epo) erythroid cell and human erythroid progenitor cell models, we report that iron-loaded transferrin, that is, holo-transferrin (holo-Tf), synergizes with suboptimal Epo levels to improve erythroid cell survival, proliferation, and differentiation. This is accomplished via the major signaling pathways of erythropoiesis, which include signal transducer and activator of transcription 5 (STAT5), mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK), and phosphoinositide-3-kinase (PI3K)/AKT. Furthermore, we found that this cooperation is improved by, but does not require, the internalization of TfR1. Interestingly, we observed that loss of TfR2 stabilizes EpoR levels and abolishes the beneficial effects of holo-Tf. Overall, these data reveal novel signaling properties of TfRs, which involve the regulation of erythropoiesis through EpoR signaling.


Subject(s)
Antigens, CD/metabolism , Cell Proliferation/drug effects , Erythroblasts/metabolism , Erythropoietin/pharmacology , Iron/pharmacology , MAP Kinase Signaling System/drug effects , Receptors, Transferrin/metabolism , Transferrin/pharmacology , Cell Line , Cell Survival/drug effects , Erythropoietin/metabolism , Humans , Iron/metabolism , Transferrin/metabolism
18.
J Nanobiotechnology ; 19(1): 115, 2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33892746

ABSTRACT

BACKGROUND: Many studies have found that ruthenium complexes possess unique biochemical characteristics and inhibit tumor growth or metastasis. RESULTS: Here, we report the novel dual-targeting ruthenium candidate 2b, which has both antitumor and antimetastatic properties and targets tumor sites through the enhanced permeability and retention (EPR) effect and transferrin/transferrin receptor (TF/TFR) interaction. The candidate 2b is composed of ruthenium-complexed carboline acid and four chloride ions. In vitro, 2b triggered DNA cleavage and thus blocked cell cycle progression and induced apoptosis via the PARP/ATM pathway. In vivo, 2b inhibited not only Lewis lung cancer (LLC) tumor growth but also lung metastasis. We detected apoptosis and decreased CD31 expression in tumor tissues, and ruthenium accumulated in the primary tumor tissue of C57BL/6 mice implanted with LLC cells. CONCLUSIONS: Thus, we conclude that 2b targets tumors, inhibits tumor growth and prevents lung metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/pharmacology , Lung Neoplasms/drug therapy , Nanomedicine/methods , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Ruthenium/chemistry , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/pathology , Cell Cycle , Cell Line, Tumor , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Receptors, Transferrin/drug effects , Transferrin/pharmacology
19.
Nanoscale Horiz ; 6(4): 348-356, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33687417

ABSTRACT

Non-apoptotic cell death such as ferroptosis and pyroptosis has shed new light on cancer treatment, whereas combinational therapy using both these mechanisms has not yet been fully explored. Herein, a dual-inductive nano-system to realize ferroptosis/pyroptosis mediated anti-cancer effects is presented. The nanodrug (Tf-LipoMof@PL) is constructed with a piperlongumine (PL) loaded metal-organic framework (MOF) coated with transferrin decorated pH sensitive lipid layer. Intracellular iron was enriched with an iron-containing MOF, whose endocytosis can be further facilitated by transferrin decorated on the lipid layer, which provides a prerequisite for the occurrence of ferroptosis and pyroptosis. Piperlongumine as the ferroptosis inducer can strengthen the ferroptotic cell death, and provide H2O2 for the dual induction system to increase ROS generation through Fenton reaction. On the basis of validation of both ferroptosis and pyroptosis, the dual-inductive nanodrug demonstrated ideal anticancer effects in the xenograft mice model, which proved that the ferroptosis/pyroptosis dual-inductive nanoplatform could be an effective and promising anticancer modality.


Subject(s)
Antineoplastic Agents/therapeutic use , Dioxolanes/therapeutic use , Ferroptosis/drug effects , Metal-Organic Frameworks/therapeutic use , Neoplasms/drug therapy , Pyroptosis/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Dioxolanes/chemistry , Dioxolanes/pharmacology , Female , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Mice, Inbred BALB C , Nanostructures/chemistry , Nanostructures/therapeutic use , Reactive Oxygen Species/metabolism , Transferrin/chemistry , Transferrin/pharmacology
20.
Sci Rep ; 11(1): 4544, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33633284

ABSTRACT

Doxorubicin (DOX) is considered one of the most powerful chemotherapeutic agents but its clinical use has several limitations, including cardiomyopathy and cellular resistance to the drug. By using transferrin (Tf) as a drug carrier, however, the adverse effects of doxorubicin as well as drug resistance can be reduced. The main objective of this study was to determine the exact nature and extent to which mitochondrial function is influenced by DOX-Tf conjugate treatment, specifically in human breast adenocarcinoma cells. We assessed the potential of DOX-Tf conjugate as a drug delivery system, monitoring its cytotoxicity using the MTT assay and ATP measurements. Moreover, we measured the alterations of mitochondrial function and oxidative stress markers. The effect of DOX-Tf was the most pronounced in MDA-MB-231, triple-negative breast cancer cells, whereas non-cancer endothelial HUVEC-ST cells were more resistant to DOX-Tf conjugate than to free DOX treatment. A different sensitivity of two investigate breast cancer cell lines corresponded to the functionality of their cellular antioxidant systems and expression of estrogen receptors. Our data also revealed that conjugate treatment mediated free radical generation and altered the mitochondrial bioenergetics in breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Doxorubicin/pharmacology , Energy Metabolism/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Transferrin/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic/drug effects , Homeostasis , Humans , Matrix Metalloproteinases/metabolism , Mitochondria/genetics , Oxidation-Reduction , Oxygen Consumption
SELECTION OF CITATIONS
SEARCH DETAIL
...