22,461 results on '"TRANSFERRIN receptors"'
Search Results
52. Transferrin and transferrin receptors update.
- Author
-
Kawabata, Hiroshi
- Subjects
- *
TRANSFERRIN , *TRANSFERRIN receptors , *IRON in the body , *ARENAVIRUSES , *PLASMODIUM vivax , *LIVER cells - Abstract
Abstract In vertebrates, transferrin (Tf) safely delivers iron through circulation to cells. Tf-bound iron is incorporated through Tf receptor (TfR) 1-mediated endocytosis. TfR1 can mediate cellular uptake of both Tf and H-ferritin, an iron storage protein. New World arenaviruses, which cause hemorrhagic fever, and Plasmodium vivax use TfR1 for entry into host cells. Human TfR2, another receptor for Tf, is predominantly expressed in hepatocytes and erythroid precursors, and holo-Tf dramatically upregulates its expression. TfR2 forms a complex with hemochromatosis protein, HFE, and serves as a component of the iron sensing machinery in hepatocytes. Defects in TfR2 cause systemic iron overload, hemochromatosis, through down-regulation of hepcidin. In erythroid cells, TfR2 forms a complex with the erythropoietin receptor and regulates erythropoiesis. TfR2 facilitates iron transport from lysosomes to mitochondria in erythroblasts and dopaminergic neurons. Administration of apo-Tf, which scavenges free iron, has been explored for various clinical conditions including atransferrinemia, iron overload, and tissue ischemia. Apo-Tf has also been shown to ameliorate anemia in animal models of β-thalassemia. In this review, I provide an update and summary on our knowledge of mammalian Tf and its receptors. Graphical abstract fx1 Highlights • Transferrin receptor 1 (TfR1) mediates cellular uptake of holo-Tf and H-ferritin. • New World arenaviruses and Plasmodium vivax use TfR1 for entry into host cells. • In hepatocytes, TfR2 forms a complex with HFE and serves as an iron sensor. • In erythroblasts, TfR2 forms a complex with EPO receptor and regulates erythropoiesis. • TfR2 facilitates iron transport from the lysosomes to mitochondria in erythroblasts. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
53. Antiproliferative and Apoptosis Triggering Potential of Paclitaxel-Based Targeted-Lipid Nanoparticles with Enhanced Cellular Internalization by Transferrin Receptors—a Study in Leukemia Cells
- Author
-
Yang Dai, Jingcao Huang, Bing Xiang, Huanling Zhu, and Chuan He
- Subjects
Leukemia ,Paclitaxel ,Lipid nanoparticles ,Apoptosis ,Cancer targeting ,Transferrin ,Materials of engineering and construction. Mechanics of materials ,TA401-492 - Abstract
Abstract Leukemia is a typical blood cancer that is characterized by the numerous duplication and proliferation of white blood cells. The main aim of this study was to develop PTX-loaded multifunctional nanoparticles and target to leukemia cells. In this study, transferrin-decorated paclitaxel-loaded lipid nanoparticle (TPLN) was prepared with an aim to increase the chemotherapeutic efficacy in the leukemia cells. Results clearly showed the superior targeting potential of TPLN to the HL-60 cancer cells compared to that of the paclitaxel-loaded nanoparticles (PLN). To be specific, TPLN showed a significantly higher cytotoxic effect in the cancer cells compared to that of the PLN indicating the superior targeting efficiency of the Tf-decorated nanoparticle system. The IC50 value of TPLN was 0.45 μg/ml compared to 2.8 μg/ml for PLN. TPLN induced a most remarkable apoptosis of the cancer cells and much of the cells were distorted with huge presence of the apoptotic body formation. Importantly, TPLN showed a remarkable reduction in the viable cells proportion to ~ 65% with around ~ 30% apoptosis cells (early and late apoptosis). Overall, results clearly showed the targeting potential of ligand-conjugated lipid nanoparticle system to the leukemia cells that might pave the way for the successful cancer treatment.
- Published
- 2018
- Full Text
- View/download PDF
54. Altered iron metabolism, inflammation, transferrin receptors, and ferritin expression in non-small-cell lung cancer
- Author
-
Kukulj, Suzana, Jaganjac, Morana, Boranic, Milivoj, Krizanac, Simun, Santic, Zarko, and Poljak-Blazi, Marija
- Published
- 2010
- Full Text
- View/download PDF
55. Nutrient transporter pattern in CD56dim NK cells: CD16 (FcγRIIIA)-dependent modulation and association with memory NK cell functional profile.
- Author
-
De Federicis, Davide, Capuano, Cristina, Ciuti, Daniel, Molfetta, Rosa, Galandrini, Ricciarda, and Palmieri, Gabriella
- Subjects
KILLER cells ,METABOLIC reprogramming ,TRANSFERRIN receptors ,GLUCOSE transporters ,HUMAN cytomegalovirus - Abstract
Background: Human memory NK cells represent a heterogeneous CD56
dim population that expands and persists in human cytomegalovirus (HCMV)-seropositive healthy individuals. They are characterized by the preferential, not fully overlapping, expression of NKG2C (activating receptor for HLA-E) and CD57 maturation marker, and by the lack of FcεRIγ adaptor chain. Hyperresponsiveness to Fcγ receptor IIIA (CD16) engagement represents the distinctive functional signature of memory NK cells. Although CD16 engagement was shown to acutely enhance glycolytic and oxidative pathways, its capability to induce a persisting metabolic reprogramming of human NK cells is poorly understood yet. Results: Here, we describe the peculiar nutrient transporter expression pattern of FcεRIγ- memory NK cells, characterized by higher levels of CD98 neutral amino acid antiporter and CD71 transferrin receptor, and lower expression of GLUT1 glucose transporter, with respect to FcεRIγ+ conventional NK cells. Although CD16 engagement acutely enhances glycolytic and oxidative pathways, its capability to induce a persisting metabolic reprogramming of human NK cells is poorly understood yet. Our results firstly show that sustained CD16 engagement by contact with IgG-opsonized target cells induces the mTORC1-dependent upregulation of CD98 and CD71 nutrient receptors on CD56dim NK cells, in a transporter-specific fashion, that is finely tuned by cell-dependent (grade of functional maturation, and memory or conventional lineage) and stimulus-dependent (time length and cooperation with cytokines) factors. We also demonstrate that CD98 antiporter function is required for CD16-dependent IFN-γ production, and that enhanced CD98-mediated neutral amino acid uptake associates with heightened memory NK cell functional response. Conclusion: Collectively, our work documents that CD16 engagement leads to a metabolic rewiring of human NK cells and suggests that a distinct nutrient transporter expression pattern may contribute to memory NK cell peculiar functional features. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
56. A Human Brain-Chip for Modeling Brain Pathologies and Screening Blood–Brain Barrier Crossing Therapeutic Strategies.
- Author
-
Chim, Shek Man, Howell, Kristen, Kokkosis, Alexandros, Zambrowicz, Brian, Karalis, Katia, and Pavlopoulos, Elias
- Subjects
INDUCED pluripotent stem cells ,MICROPHYSIOLOGICAL systems ,DRUG discovery ,TUMOR necrosis factors ,TRANSFERRIN receptors - Abstract
Background/Objectives: The limited translatability of preclinical experimental findings to patients remains an obstacle for successful treatment of brain diseases. Relevant models to elucidate mechanisms behind brain pathogenesis, including cell-specific contributions and cell-cell interactions, and support successful targeting and prediction of drug responses in humans are urgently needed, given the species differences in brain and blood-brain barrier (BBB) functions. Human microphysiological systems (MPS), such as Organ-Chips, are emerging as a promising approach to address these challenges. Here, we examined and advanced a Brain-Chip that recapitulates aspects of the human cortical parenchyma and the BBB in one model. Methods: We utilized human primary astrocytes and pericytes, human induced pluripotent stem cell (hiPSC)-derived cortical neurons, and hiPSC-derived brain microvascular endothelial-like cells and included for the first time on-chip hiPSC-derived microglia. Results: Using Tumor necrosis factor alpha (TNFα) to emulate neuroinflammation, we demonstrate that our model recapitulates in vivo-relevant responses. Importantly, we show microglia-derived responses, highlighting the Brain-Chip's sensitivity to capture cell-specific contributions in human disease-associated pathology. We then tested BBB crossing of human transferrin receptor antibodies and conjugated adeno-associated viruses. We demonstrate successful in vitro/in vivo correlation in identifying crossing differences, underscoring the model's capacity as a screening platform for BBB crossing therapeutic strategies and ability to predict in vivo responses. Conclusions: These findings highlight the potential of the Brain-Chip as a reliable and time-efficient model to support therapeutic development and provide mechanistic insights into brain diseases, adding to the growing evidence supporting the value of MPS in translational research and drug discovery. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
57. Iron status in children with acute COVID-19 and paediatric inflammatory multisystem syndrome during infection and after recovery
- Author
-
Mai S. El-Meshad, Angi Adel Alwakeel, Reham M. El-Farahaty, Hyam Sameh Nada, and Mayada S. Zeid
- Subjects
COVID-19 ,MIS-C ,Paediatrics ,Anaemia ,Soluble transferrin receptors (sTfRs) ,Iron ,Pediatrics ,RJ1-570 - Abstract
Abstract Background COVID-19 has significant effects on organ function, particularly on lung function and iron metabolism. Studies have shown increased levels of ferritin, an iron storage protein, in COVID-19 patients, indicating potential changes in iron utilization. Research has focused primarily on adults, with limited studies on paediatric patients and a lack of comparisons with MIS-C patients. This study aimed to assess iron status in paediatric COVID-19 patients using traditional and new biomarkers, soluble transferrin receptors (sTfR) and Reticulocyte hemoglobin equivalent (RET-He), to improve diagnosis and prognosis. Additionally, we sought to compare iron status between acute COVID-19 patients and MIS-C patients and evaluate the relationships among iron dysmetabolism, disease severity, and prognosis in paediatric patients. The study also involved monitoring iron status during and after infection to understand its impact on patient severity and prognosis. Methods A cohort study involving 49 patients aged 1 month to 18 years was conducted at the isolation department of Mansoura University Children's Hospital. The study included 36 patients with acute COVID-19 and 13 with multisystem inflammatory syndrome of childhood (MIS-C). Diagnosis was based on PCR from a deep nasopharyngeal swab or a positive antibody test. Follow-up of survivors was conducted 3 months after recovery. Blood samples were obtained during infection and at follow-up for CBC, Ret-He, iron kinetics, and sTfR analyses. Results Significant iron deficiency anaemia was observed in all patients during infection, with improvement after 3 months of recovery in survivors. The improvement was more obvious in MIS-C patients, with Hb and iron kinetics not significantly affected by disease severity. The STfR was significantly lower in nonsurvivors than in survivors. The ROC curve showed that a baseline sTfR ≤ 18 nmol/L was a statistically significant difference between nonsurvivors and survivors (area under the curve (AUC) = 0.810, p
- Published
- 2024
- Full Text
- View/download PDF
58. Oligomerized Transferrin Receptors Are Selectively Retained by a Lumenal Sorting Signal in a Long-Lived Endocytic Recycling Compartment
- Author
-
Marsh, Eric W., Leopold, Philip L., Jones, Nancy L., and Maxfield, Frederick R.
- Published
- 1995
59. Intracellular Pools of Transferrin Receptors Result from Constitutive Internalization of Unoccupied Receptors
- Author
-
Ajioka, Richard S. and Kaplan, Jerry
- Published
- 1986
60. Identification of Transferrin Receptors on the Surface of Human Cultured Cells
- Author
-
Hamilton, Thomas A., Wada, H. Garrett, and Sussman, Howard H.
- Published
- 1979
61. Differential Tissue Localization of Oviduct and Erythroid Transferrin Receptors
- Published
- 1991
62. Subunit Structure of Cell Surface Proteins: Disulfide Bonding in Antigen Receptors, Ly-2/3 Antigens, and Transferrin Receptors of Murine T and B Lymphocytes
- Author
-
Goding, James W. and Harris, Alan W.
- Published
- 1981
63. The End2 Mutation in CHO Cells Slows the Exit of Transferrin Receptors from the Recycling Compartment but Bulk Membrane Recycling Is Unaffected
- Author
-
Presley, John F., Mayor, Satyajit, Dunn, Kenneth W., Johnson, Lester S., McGraw, Timothy E., and Maxfield, Frederick R.
- Published
- 1993
64. Supplementary Figures + Legend from Optimized Doxorubicin Chemotherapy for Diffuse Large B-cell Lymphoma Exploits Nanocarrier Delivery to Transferrin Receptors
- Author
-
Arumov, Artavazd, primary, Liyanage, Piumi Y., primary, Trabolsi, Asaad, primary, Roberts, Evan R., primary, Li, Lingxiao, primary, Ferreira, Braulio C.L.B., primary, Gao, Zhen, primary, Ban, Yuguang, primary, Newsam, Austin D., primary, Taggart, Melissa W., primary, Vega, Francisco, primary, Bilbao, Daniel, primary, Leblanc, Roger M., primary, and Schatz, Jonathan H., primary
- Published
- 2023
- Full Text
- View/download PDF
65. Value of Soluble Transferrin Receptors and sTfR/log Ferritin in the Diagnosis of Iron Deficiency Accompanied by Acute Infection
- Author
-
El-Gendy, Fady M., El-Hawy, Mahmoud A., Rizk, Mohamed S., El-Hefnawy, Sally M., and Mahmoud, Mohamed Z.
- Published
- 2017
- Full Text
- View/download PDF
66. Association of poly(rC)-binding protein-2 with sideroflexin-3 through TOM20 as an iron entry pathway to mitochondria.
- Author
-
Mi, Danyang, Yanatori, Izumi, Zheng, Hao, Kong, Yingyi, Hirayama, Tasuku, and Toyokuni, Shinya
- Subjects
- *
TRANSFERRIN , *TRANSFERRIN receptors , *CHRONIC myeloid leukemia , *FERRITIN , *MITOCHONDRIA , *MITOCHONDRIAL proteins , *STIMULATED emission , *IRON overload - Abstract
Iron is essential for all the lives and mitochondria integrate iron into heme and Fe-S clusters for diverse use as cofactors. Here, we screened mitochondrial proteins in KU812 human chronic myelogenous leukemia cells by glutathione S-transferase pulldown assay with PCBP2 to identify mitochondrial receptors for PCBP2, a major cytosolic Fe(II) chaperone. LC–MS analyses identified TOM20, sideroflexin-3 (SFXN3), SFXN1 and TOM70 in the affinity-score sequence. Stimulated emission depletion microscopy and proteinase-K digestion of mitochondria in HeLa cells revealed that TOM20 is located in the outer membrane of mitochondria whereas SFXN3 is located in the inner membrane. Although direct association was not observed between PCBP2 and SFXN3 with co-immunoprecipitation, proximity ligation assay demonstrated proximal localization of PCBP2 with TOM20 and there was a direct binding between TOM20 and SFXN3. Single knockdown either of PCBP2 and SFXN3 in K562 leukemia cells significantly decreased mitochondrial catalytic Fe(II) and mitochondrial maximal respiration. SFXN3 but not MFRN1 knockout (KO) in mouse embryonic fibroblasts decreased FBXL5 and heme oxygenase-1 (HO-1) but increased transferrin uptake and induced ferritin, indicating that mitochondrial iron entry through SFXN3 is distinct. MFRN1 KO revealed more intense mitochondrial Fe(II) deficiency than SFXN3 KO. Insufficient mitochondrial heme synthesis was evident under iron overload both with SFXN3 and MFRN KO, which was partially reversed by HO-1 inhibitor. Conversely, SFXN3 overexpression caused cytosolic iron deficiency with mitochondrial excess Fe(II), which further sensitized HeLa cells to RSL3-induced ferroptosis. In conclusion, we discovered a novel pathway of iron entry into mitochondria from cytosol through PCBP2-TOM20-SFXN3 axis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
67. Tetramethylpyrazine attenuates renal tubular epithelial cell ferroptosis in contrast-induced nephropathy by inhibiting transferrin receptor and intracellular reactive oxygen species.
- Author
-
Zhongqiang Zhu, Jun Li, Zhiyong Song, Tonglu Li, Zongping Li, and Xuezhong Gong
- Subjects
- *
TRANSFERRIN receptors , *CONTRAST induced nephropathy , *REACTIVE oxygen species , *FERRITIN , *EPITHELIAL cells , *CARRIER proteins , *CELL survival , *LIPID peroxidation (Biology) - Abstract
Contrast-induced nephropathy (CIN) is a leading cause of hospital-acquired acute kidney injury (AKI). Recently, ferroptosis was reported to be crucial for AKI pathogenesis. Our previous studies indicated antioxidant tetramethylpyrazine (TMP) prevent CIN in vivo. However, whether ferroptosis is involved in TMP nephroprotective mechanism against CIN is unclear. In the present study, we investigated the role of renal tubular epithelial cell ferroptosis in TMP reno-protective effect against CIN and the molecular mechanisms by which TMP regulates ferroptosis. Classical contrast-medium, Iohexol, was used to construct CIN models in rats and HK-2 cells. Results showed that tubular cell injury was accompanied by ferroptosis both in vivo and in vitro, including the typical features of ferroptosis, Fe2+ accumulation, lipid peroxidation and decreased glutathione peroxidase 4 (GPX4). Ferroptosis inhibition by classic inhibitors Fer-1 and DFO promoted cell viability and reduced intracellular ROS production. Additionally, TMP significantly inhibited renal dysfunction, reduced AKI biomarkers, prevented ROS production, inhibited renal Fe2+ accumulation and increased GPX4 expression. Expressions of various proteins associated with iron ion metabolism, including transferrin receptor (TFRC), divalent metal transporter 1, iron-responsive element binding protein 2, ferritin heavy chain 1, ferroportin 1, and heat shock factor binding protein 1, were examined using mechanistic analyses. Among these, TFRC changes were the most significant after TMP pretreatment. Results of siRNA knockdown and plasmid overexpression of TFRC indicated that TFRC is essential for TMP to alleviate ferroptosis and reduce LDH release, Fe2+ accumulation and intracellular ROS. Our findings provide crucial insights about the potential of TMP in treating AKI associated with ferroptosis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
68. Mechanism of Ziyuglycoside II-mediated Ferroptosis-related Proteins on the Proliferation and Metastasis of Human Lung Adenocarcinoma Cell Lines.
- Author
-
Zhang, Jian-hui, Xie, Li-jun, Wang, Han-lu, Chen, Hui, Meng, Xiao-rong, Lin, Xing, Lin, Xin-fu, Liao, Li-sheng, Chen, Ting, Luo, Jie-Wei, Hong, Lu-yu, and Chen, Xin
- Subjects
- *
TRANSFERRIN receptors , *CELL lines , *CANCER cell growth , *GLUTAMATE transporters , *CELL migration - Abstract
Background: Ferroptosis is a novel type of regulated cell death and targeting ferroptosis may be a potential treatment strategy for lung cancer. Ziyuglycoside II (ZYG II) has a significant inhibitory effect on the growth of lung cancer cells. However, the selective anti-tumor effect of the ZYG II against lung cancer has not been systemically studied. Objectives: We combined ferrostatin-1 and erastin to explore the potential therapeutic mechanism of the ZYG II for lung adenocarcinoma. Materials and Methods: A549 and H1299 cells were randomly divided into the control, ZYG II, ferroptosis inhibitor group (ZYG II+ ferrostatin-1), and erastin group (ZYG II+ erastin). Cell proliferation was detected using the CCK-8 method. Cell migration and invasion were evaluated using the Transwell assay. The protein expression levels of Glutathione Peroxidase 4 (GPX4), Solute Carrier Family 7 Member 11 (SLC7A11), and Transferrin receptor 1 (TFR1) were measured using western blotting. Results: Compared with the control group, the cell proliferation, migration, and invasion abilities of the ZYG II group significantly decreased, the protein expression levels of GPX4 and SLC7A11 in the ZYG II group declined significantly, and the expression of TFR1 increased significantly (p < 0.05). After adding ferrostatin 1 (ZYG II+ Ferrostatin 1), the cell proliferation, migration, and invasion abilities of the inhibited cells were significantly increased, the expression of GPX4 and SLC7A11 increased significantly and the expression of TFR1 decreased significantly (p < 0.05). However, after adding the erastin (ZYG II+ erastin), the cell viability was further inhibited in A549, the expression levels of GPX4 and SLC7A11 were further inhibited and the expression of TFR1 was further increased (p < 0.05). Conclusion: ZYG II significantly inhibited the survival rate, proliferation, migration, and invasion ability of A549 and H1299 cells, possibly by inducing ferroptosis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
69. A conserved trypanosomatid differentiation regulator controls substrate attachment and morphological development in Trypanosoma congolense.
- Author
-
Silvester, Eleanor, Szoor, Balazs, Ivens, Alasdair, Awuah-Mensah, Georgina, Gadelha, Catarina, Wickstead, Bill, and Matthews, Keith R.
- Subjects
- *
TRANSFERRIN receptors , *RNA interference , *TRYPANOSOMA , *SMALL interfering RNA , *LIFE cycles (Biology) , *BLOOD parasites - Abstract
Trypanosomatid parasites undergo developmental regulation to adapt to the different environments encountered during their life cycle. In Trypanosoma brucei, a genome wide selectional screen previously identified a regulator of the protein family ESAG9, which is highly expressed in stumpy forms, a morphologically distinct bloodstream stage adapted for tsetse transmission. This regulator, TbREG9.1, has an orthologue in Trypanosoma congolense, despite the absence of a stumpy morphotype in that parasite species, which is an important cause of livestock trypanosomosis. RNAi mediated gene silencing of TcREG9.1 in Trypanosoma congolense caused a loss of attachment of the parasites to a surface substrate in vitro, a key feature of the biology of these parasites that is distinct from T. brucei. This detachment was phenocopied by treatment of the parasites with a phosphodiesterase inhibitor, which also promotes detachment in the insect trypanosomatid Crithidia fasciculata. RNAseq analysis revealed that TcREG9.1 silencing caused the upregulation of mRNAs for several classes of surface molecules, including transferrin receptor-like molecules, immunoreactive proteins in experimental bovine infections, and molecules related to those associated with stumpy development in T. brucei. Depletion of TcREG9.1 in vivo also generated an enhanced level of parasites in the blood circulation consistent with reduced parasite attachment to the microvasculature. The morphological progression to insect forms of the parasite was also perturbed. We propose a model whereby TcREG9.1 acts as a regulator of attachment and development, with detached parasites being adapted for transmission. Author summary: Trypanosoma congolense is a major cause of livestock trypanosomosis in sub-Saharan Africa where it contributes to lost economic productivity and poverty. These parasites differ in two key respects from the better-studied African trypanosomes, Trypanosoma brucei. Firstly, T. congolense adheres to surfaces in vitro and in the vasculature in vivo. Secondly, they lack a morphologically distinct transmission stage equivalent to the stumpy form of T. brucei. In the current work we identify a T congolense orthologue of a key developmental regulator in T. brucei, REG9.1, previously identified by a genome-wide RNAi screen. By RNA interference in transgenic T. congolense we demonstrate that TcREG9.1 functions both in parasite adherence in vitro and in vivo, and also in parasite development. The work provides a first insight into the unusual adherence and developmental biology of T. congolense and also suggests a possible model for how these characteristics interact to promote disease spread. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
70. Transferrin receptor 2 mitigates periodontitis‐driven alveolar bone loss.
- Author
-
Lösser, Lennart, Ledesma‐Colunga, Maria G., Sastre, Enrique Andrés, Scholtysek, Carina, Hofbauer, Lorenz C., Noack, Barbara, Baschant, Ulrike, and Rauner, Martina
- Subjects
- *
BONE resorption , *TRANSFERRIN receptors , *GENE expression , *IRON in the body , *HOMEOSTASIS , *ALVEOLAR process - Abstract
Periodontitis is associated with significant alveolar bone loss. Patients with iron overload suffer more frequently from periodontitis, however, the underlying mechanisms remain largely elusive. Here, we investigated the role of transferrin receptor 2 (Tfr2), one of the main regulators of iron homeostasis, in the pathogenesis of periodontitis and the dental phenotype under basal conditions in mice. As Tfr2 suppresses osteoclastogenesis, we hypothesized that deficiency of Tfr2 may exacerbate periodontitis‐induced bone loss. Mice lacking Tfr2 (Tfr2−/−) and wild‐type (Tfr2+/+) littermates were challenged with experimental periodontitis. Mandibles and maxillae were collected for microcomputed tomography and histology analyses. Osteoclast cultures from Tfr2+/+ and Tfr2−/− mice were established and analyzed for differentiation efficiency, by performing messenger RNA expression and protein signaling pathways. After 8 days, Tfr2‐deficient mice revealed a more severe course of periodontitis paralleled by higher immune cell infiltration and a higher histological inflammation index than Tfr2+/+ mice. Moreover, Tfr2‐deficient mice lost more alveolar bone compared to Tfr2+/+ littermates, an effect that was only partially iron‐dependent. Histological analysis revealed a higher number of osteoclasts in the alveolar bone of Tfr2‐deficient mice. In line, Tfr2‐deficient osteoclastic differentiation ex vivo was faster and more efficient as reflected by a higher number of osteoclasts, a higher expression of osteoclast markers, and an increased resorptive activity. Mechanistically, Tfr2‐deficient osteoclasts showed a higher p38‐MAPK signaling and inhibition of p38‐MAPK signaling in Tfr2‐deficient cells reverted osteoclast formation to Tfr2+/+ levels. Taken together, our data indicate that Tfr2 modulates the inflammatory response in periodontitis thereby mitigating effects on alveolar bone loss. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
71. Transferrin receptor levels and its rare variant are associated with human obesity.
- Author
-
Qiu, Jin, Zhang, Zhiyin, Hu, Yepeng, Guo, Yuhan, Liu, Caizhi, Chen, Yanru, Wang, Dongmei, Su, Junlei, Wang, Sainan, Ni, Mengshan, Xu, Sainan, Yu, Jian, Hu, Tianhui, Song, Gaojie, Ma, Xinran, Gu, Xuejiang, Wang, Jiqiu, and Xu, Lingyan
- Subjects
- *
TRANSFERRIN receptors , *OBESITY , *IRON in the body , *BROWN adipose tissue , *BODY mass index , *ADIPOSE tissue diseases - Abstract
Aim: Iron homeostasis is critical for functional respiratory chain complex of mitochondrial, thus potentially contributing to fat biology and energy homeostasis. Transferrin receptor (Tfrc) binds to transferrin for extracellular iron uptake and is recently reported to be involved in brown fat development and functionality. However, whether TFRC levels and variants are associated with human obesity is unknown. Methods: To investigate the association of TFRC levels and variants with human obesity, fat biopsies were obtained from surgery. Exon‐sequencing and genetic assessments were conducted of a case–control study. For TFRC levels assessment in fat biopsy, 9 overweight and 12 lean subjects were involved. For genetic study, obese (n = 1271) and lean subjects (n = 1455) were involved. TFRC levels were compared in abdominal mesenteric fat of pheochromocytoma patients versus control subjects, and overweight versus lean subjects. For genetic study, whole‐exome sequencing of obese and matched control subjects were conducted and analyzed. In addition, the possible disruption in protein stability of TFRC variant was assessed by structural and molecular analysis. Results: TFRC levels are increased in human browning adipose tissue and decreased in fat of overweight patients. Besides, TFRC levels are negatively correlated with body mass index and positively correlated with uncoupling protein 1 levels. Furthermore, a rare heterozygous missense variant p.I337V in TFRC shows a tendency to enrich in obese subjects. Structural and functional study reveals impaired protein stability of the TFRC variant compared to wild‐type. Conclusions: Reduced TFRC levels and its rare variant p.I337V with protein instability are associated with human obesity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
72. An investigation of the distributions of ferroptosis and necroptosis mediators in the maternal–fetal interface at different days of rat pregnancy.
- Author
-
Tatar, Musa and Tüfekci, Kıymet Kübra
- Subjects
- *
TRANSFERRIN receptors , *TRANSFERRIN , *DECIDUA , *YOLK sac , *PREGNANCY , *RATS , *LABORATORY rats , *PROTEIN expression - Abstract
Ferroptosis and necroptosis are recognized as playing major roles in the regulation of various physiological processes. However, the physiological role of the cell death mediated by these two pathways in the developmental process has not yet been clearly established. This study investigated ferroptosis and necroptosis signalling pathways in maternal–fetal tissue in the different gestational days (GD) of rat pregnancy using immunohistochemical and western blot methods in order to fill this gap. Twenty‐four female Wistar albino rats were mated and divided into three groups. Maternal–fetal tissue samples were collected on GD 5, 12 and 19 of pregnancy. Expression and total protein levels of the markers glutathione peroxidase‐4, soluble transporter family 7 member 11, transferrin receptor, receptor‐interacting serine/threonine‐protein kinase 1, receptor‐interacting serine/threonine‐protein kinase 3 and mixed lineage kinase domain‐like protein were investigated on both the maternal and fetal surfaces of the placenta using immunohistochemical and western blot methods. The results showed varying levels of protein expression of both ferroptosis and necroptosis mediators in the GD 5, 12 and 19 of pregnancy. Immunohistochemical analyses revealed that these mediators were located on both the maternal (decidua and metrial gland) and fetal surfaces (labyrinth zone, yolk sac and basal zone) and that their expression levels changed in the different GD. The findings revealed the existence of important ferroptosis and necroptosis pathway mediators in rat maternal–fetal tissue. These results may provide a molecular framework for a better understanding of the communication between the placenta, decidua and fetus during the developmental process. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
73. IST1 regulates select recycling pathways.
- Author
-
Clippinger, Amy K., Naismith, Teresa V., Yoo, Wonjin, Jansen, Silvia, Kast, David J., and Hanson, Phyllis I.
- Subjects
- *
TRANSFERRIN receptors , *ENDOSOMES , *CELL membranes , *CLATHRIN , *MEMBRANE proteins , *MANNOSE - Abstract
ESCRTs (Endosomal Sorting Complex Required for Transports) are a modular set of protein complexes with membrane remodeling activities that include the formation and release of intraluminal vesicles (ILVs) to generate multivesicular endosomes. While most of the 12 ESCRT‐III proteins are known to play roles in ILV formation, IST1 has been associated with a wider range of endosomal remodeling events. Here, we extend previous studies of IST1 function in endosomal trafficking and confirm that IST1, along with its binding partner CHMP1B, contributes to scission of early endosomal carriers. Functionally, depleting IST1 impaired delivery of transferrin receptor from early/sorting endosomes to the endocytic recycling compartment and instead increased its rapid recycling to the plasma membrane via peripheral endosomes enriched in the clathrin adaptor AP‐1. IST1 is also important for export of mannose 6‐phosphate receptor from early/sorting endosomes. Examination of IST1 binding partners on endosomes revealed that IST1 interacts with the MIT domain‐containing sorting nexin SNX15, a protein previously reported to regulate endosomal recycling. Our kinetic and spatial analyses establish that SNX15 and IST1 occupy a clathrin‐containing subdomain on the endosomal perimeter distinct from those previously implicated in cargo retrieval or degradation. Using live‐cell microscopy, we see that SNX15 and CHMP1B alternately recruit IST1 to this subdomain or the base of endosomal tubules. These findings indicate that IST1 contributes to a subset of recycling pathways from the early/sorting endosome. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
74. Multimodal highly fluorescent-magnetic nanoplatform to target transferrin receptors in cancer cells.
- Author
-
Cabral Filho, Paulo E., Cabrera, Mariana P., Cardoso, Ana L.C., Santana, Otacilio A., Geraldes, Carlos F.G.C., Santos, Beate S., Pedroso de Lima, Maria C., Pereira, Giovannia A.L., and Fontes, Adriana
- Subjects
- *
TRANSFERRIN receptors , *HELA cells , *MAGNETIC resonance imaging , *FERRIC oxide , *FLUORESCENCE spectroscopy - Abstract
Abstract Background Site-specific multimodal nanoplatforms with fluorescent-magnetic properties have great potential for biological sciences. For this reason, we developed a multimodal nanoprobe (BNPs-Tf), by covalently conjugating an optical-magnetically active bimodal nanosystem, based on quantum dots and iron oxide nanoparticles, with the human holo-transferrin (Tf). Methods The Tf bioconjugation efficiency was evaluated by the fluorescence microplate assay (FMA) and the amount of Tf immobilized on BNPs was quantified by fluorescence spectroscopy. Moreover, relaxometric and fluorescent properties of the BNPs-Tf were evaluated, as well as its ability to label specifically HeLa cells. Cytotoxicity was also performed by Alamar Blue assay. Results The FMA confirmed an efficient bioconjugation and the fluorescence spectroscopy analysis indicated that 98% of Tf was immobilized on BNPs. BNPs-Tf also presented a bright fluorescence and a transversal/longitudinal relaxivities ratio (r 2 / r 1) of 65. Importantly, the developed BNPs-Tf were able to label, efficiently and specifically, the Tf receptors in HeLa cells, as shown by fluorescence and magnetic resonance imaging assays. Moreover, this multimodal system did not cause noteworthy cytotoxicity. Conclusions The prepared BNPs-Tf hold great promise as an effective and specific multimodal, highly fluorescent-magnetic, nanoplatform for fluorescence analyses and T 2 -weighted images. General significance This study developed an attractive and versatile multimodal nanoplatform that has potential to be applied in a variety of in vitro and in vivo studies, addressing biological processes, diagnostic, and therapeutics. Moreover, this work opens new possibilities for designing other efficient multimodal nanosystems, considering other biomolecules in their composition able to provide them important functional properties. Graphical abstract Unlabelled Image Highlights • Transferrin (Tf) was efficiently conjugated to bimodal nanoparticles (BNPs). • BNPs-Tf presented high fluorescence and transversal relaxivity. • BNPs-Tf labeled efficiently cell Tf receptors by fluorescence /relaxometric assays. • BNPs-Tf did not cause significant toxicity in HeLa cells, under the conditions applied. • BNPs-Tf hold great promise for fluorescence analyses and T 2 -weighted images. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
75. Complex and Dynamic Interactions between Parvovirus Capsids, Transferrin Receptors, and Antibodies Control Cell Infection and Host Range.
- Author
-
Callaway, Heather M., Welsch, Kathrin, Weichert, Wendy, Allison, Andrew B., Hafenstein, Susan L., Kai Huang, Iketani, Sho, and Parrish, Colin R.
- Subjects
- *
CAPSIDS , *TRANSFERRIN receptors , *IMMUNOGLOBULINS , *IMMUNE response , *BINDING sites - Abstract
Antibody and receptor binding are key virus-host interactions that control host range and determine the success of infection. Canine and feline parvovirus capsids bind the transferrin receptor type 1 (TfR) to enter host cells, and specific structural interactions appear necessary to prepare the stable capsids for infection. Here, we define the details of binding, competition, and occupancy of wild-type and mutant parvovirus capsids with purified receptors and antibodies. TfR-capsid binding interactions depended on the TfR species and varied widely, with no direct relationship between binding affinity and infection. Capsids bound feline, raccoon, and black-backed jackal TfRs at high affinity but barely bound canine TfRs, which mediated infection efficiently. TfRs from different species also occupied capsids to different levels, with an estimated 1 to 2 feline TfRs but 12 black-backed jackal TfRs binding each capsid. Multiple alanine substitutions within loop 1 on the capsid surface reduced TfR binding but substitutions within loop 3 did not, suggesting that loop 1 directly engaged the TfR and loop 3 sterically affected that interaction. Binding and competition between different TfRs and/or antibodies showed complex relationships. Both antibodies 14 and E competed capsids off TfRs, but antibody E could also compete capsids off itself and antibody 14, likely by inducing capsid structural changes. In some cases, the initial TfR or antibody binding event affected subsequent TfR binding, suggesting that capsid structure changes occur after TfR or antibody binding and may impact infection. This shows that precise, host-specific TfR-capsid interactions, beyond simple attachment, are important for successful infection. IMPORTANCE Host receptor binding is a key step during viral infection and may control both infection and host range. In addition to binding, some viruses require specific interactions with host receptors in order to infect, and anti-capsid antibodies can potentially disrupt these interactions, leading to neutralization. Here, we examine the interactions between parvovirus capsids, the receptors from different hosts, and anti-capsid antibodies. We show that interactions between parvovirus capsids and host-specific TfRs vary in both affinity and in the numbers of receptors bound, with complex effects on infection. In addition, antibodies binding to two sites on the capsids had different effects on TfR-capsid binding. These experiments confirm that receptor and antibody binding to parvovirus capsids are complex processes, and the infection outcome is not determined simply by the affinity of attachment. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
76. Decreased expression of membrane α4β1, α5β1 integrins and transferrin receptor on erythroblasts in splenectomized patients with β-thalassemia intermedia. Parallel assessment of serum soluble transferrin receptors levels
- Author
-
Kossiva, Lydia, Paterakis, George, Tassiopoulos, Stergios, Papadhimitriou, Stefanos I., Voukouti, Eugenia, Gligori, Ioanna, and Rombos, Yannis
- Published
- 2003
- Full Text
- View/download PDF
77. Serum transferrin receptors in detection of iron deficiency in pregnancy
- Author
-
Rusia, U., Flowers, C., Madan, N., Agarwal, N., Sood, S. K., and Sikka, M.
- Published
- 1999
- Full Text
- View/download PDF
78. Iron parameters analysis in dogs with myxomatous mitral valve disease.
- Author
-
Kumiega, Ewa, A Kobak, Kamil, Noszczyk-Nowak, Agnieszka, and Kasztura, Monika
- Subjects
TRANSFERRIN ,IRON in the body ,DOGS ,MITRAL valve ,TRANSFERRIN receptors ,BODY size ,BIOMARKERS - Abstract
Background: Myxomatous mitral valve disease (MMVD) is the most common acquired cardiovascular disease in small breed dogs. In contrast to human patients with heart failure (HF), iron deficiency (ID) prevalence in dogs with MMVD is weakly known. The study aimed to assess the usability of ID markers in serum and reticulocyte parameters from whole blood of dogs with MMVD to evaluate early ID symptoms. Results: Sixty-eight dogs (43 male and 25 female) were included in the study. MMVD dogs were assigned according to the 2019 ACVIM guidelines for groups B1 (n = 9), B2 (n = 10), C (n = 27) and D (n = 10). Groups were also combined into B1 and B2 as non-symptomatic HF and C with D as symptomatic HF. Healthy controls were 12 dogs. Serum iron concentration below the reference range in dogs with MMVD was 12.5%. Other ID indices, such as %SAT, UIBC, and TIBC were similar in the MMVD groups and healthy controls (p > 0.05 for all parameters). Statistical comparison between control group and 4 groups of different stages of MMVD showed that significant differences occur only in serum transferrin. The assessment of ferritin and soluble transferrin receptors using Western Blotting did not show differences between control (n = 7) and MMVD (n = 33) dogs. Study has shown positive correlation between ID parameters and echocardiographic indices such as LA/Ao and LVIDdN, and some biochemical parameters. A significant increase in reticulocytes percentage, assessed manually, was observed in the HF group of animals (p = 0.027) compared to the control group. Conclusions: Studies have shown that ID parameters in serum are not significantly different in dogs with MMVD compared to healthy dogs. However, there is a clear correlation between atrial size and normalised left ventricular size to body size and some biochemical parameters, including ID parameters and therefore the severity of MMVD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
79. Glycosylation and Characterization of Human Transferrin in an End-Stage Kidney Disease.
- Author
-
Miljuš, Goran, Penezić, Ana, Pažitná, Lucia, Gligorijević, Nikola, Baralić, Marko, Vilotić, Aleksandra, Šunderić, Miloš, Robajac, Dragana, Dobrijević, Zorana, Katrlík, Jaroslav, and Nedić, Olgica
- Subjects
TRANSFERRIN ,CHRONIC kidney failure ,GLYCOSYLATION ,TRANSFERRIN receptors ,PERITONEAL dialysis ,IRON metabolism - Abstract
Chronic kidney disease (CKD) is a global health concern affecting approximately one billion individuals worldwide. End-stage kidney disease (ESKD), the most severe form of CKD, is often accompanied by anemia. Peritoneal dialysis (PD), a common treatment for ESKD, utilizes the peritoneum for solute transfer but is associated with complications including protein loss, including transferrin (Tf) a key protein involved in iron transport. This study investigated Tf characteristics in ESKD patients compared to healthy individuals using lectin microarray, spectroscopic techniques and immunocytochemical analysis to assess Tf interaction with transferrin receptors (TfRs). ESKD patients exhibited altered Tf glycosylation patterns, evidenced by significant changes in lectin reactivity compared to healthy controls. However, structural analyses revealed no significant differences in the Tf secondary or tertiary structures between the two groups. A functional analysis demonstrated comparable Tf-TfR interaction in both PD and healthy samples. Despite significant alterations in Tf glycosylation, structural integrity and Tf-TfR interaction remained preserved in PD patients. These findings suggest that while glycosylation changes may influence iron metabolism, they do not impair Tf function. The study highlights the importance of a glucose-free dialysis solutions in managing anemia exacerbation in PD patients with poorly controlled anemia, potentially offering a targeted therapeutic approach to improve patient outcomes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
80. Transcytosis-Driven Treatment of Neurodegenerative Disorders by mRNA-Expressed Antibody–Transferrin Conjugates.
- Author
-
Niazi, Sarfaraz K. and Magoola, Matthias
- Subjects
NEURODEGENERATION ,TRANSFERRIN receptors ,CARRIER proteins ,BLOOD-brain barrier ,TRANSFERRIN - Abstract
The recent setbacks in the withdrawal and approval delays of antibody treatments of neurodegenerative disorders (NDs), attributed to their poor entry across the blood–brain barrier (BBB), emphasize the need to bring novel approaches to enhance the entry across the BBB. One such approach is conjugating the antibodies that bind brain proteins responsible for NDs with the transferrin molecule. This glycoprotein transports iron into cells, connecting with the transferrin receptors (TfRs), piggybacking an antibody–transferrin complex that can subsequently release the antibody in the brain or stay connected while letting the antibody bind. This process increases the concentration of antibodies in the brain, enhancing therapeutic efficacy with targeted delivery and minimum systemic side effects. Currently, this approach is experimented with using drug-transferring conjugates assembled in vitro. Still, a more efficient and safer alternative is to express the conjugate using mRNA technology, as detailed in this paper. This approach will expedite safer discoveries that can be made available at a much lower cost than the recombinant process with in vitro conjugation. Most importantly, the recommendations made in this paper may save the antibodies against the NDs that seem to be failing despite their regulatory approvals. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
81. The PvRBP2b-TfR1 interaction is not essential for reticulocytes invasion by Plasmodium vivax isolates from Cambodia.
- Author
-
Feufack-Donfack, Lionel B., Baldor, Léa, Roesch, Camille, Tat, Baura, Orban, Agnes, Seng, Dynang, Salvador, Jeremy, Khim, Nimol, Carias, Lenore, King, Christopher L., Russell, Bruce, Nosten, Francois, Ong, Alice SM, Mao, Haitong, Renia, Laurent, Lo, Eugenia, Witkowski, Benoit, and Popovici, Jean
- Subjects
CARRIER proteins ,PLASMODIUM vivax ,RETICULOCYTES ,POLYMORPHISM (Zoology) ,CAMBODIANS ,TRANSFERRIN receptors ,MONOCLONAL antibodies - Abstract
Plasmodium vivax is the most widespread of the different Plasmodium species able to infect humans and is responsible for most malaria cases outside Africa. An effective, strain-transcending vaccine that alleviates or suppresses erythrocyte invasion would be a game-changer in eliminating vivax malaria. Recently, the binding of P. vivax Reticulocyte Binding Protein 2b (PvRBP2b) to human Transferrin receptor (TfR1) has been described as essential for reticulocyte invasion, making this parasite protein an appealing vaccine candidate. Here, using P. vivax Cambodian clinical isolates in robust ex vivo invasion assays, we show that anti-PvRBP2b polyclonal and monoclonal antibodies that inhibit binding of PvRBP2b to TfR1 do not block P. vivax invasion into reticulocytes even at high concentrations. Anti-TfR1 antibodies do not inhibit P. vivax invasion either. Combinations at high concentrations of human monoclonal antibodies targeting different PvRBP2b epitopes do not inhibit invasion. Combinations of anti-PvRBP2b with anti-PvDBP do not enhance invasion inhibition caused by anti-PvDBP alone. We also show that the invasion of Cambodian P. vivax is trypsin-resistant while TfR1 is trypsin-sensitive, and we demonstrate that TfR1 is not recycled following trypsin treatment. We determined the PvRBP2b sequence of all isolates used in the invasion assays and analyzed polymorphism within epitopes recognized by anti-PvRBP2b antibodies. We show that polymorphism does not explain the absence of neutralization. Anti-PvRBP2b polyclonal antibodies recognized all four isolates tested in immunofluorescence assays while not inhibiting P. vivax invasion. Overall, our results demonstrate that PvRBP2b binding to TfR1 is not essential for invasion into reticulocytes of P. vivax Cambodian strains questioning the relevance of PvRBP2b as vaccine candidate. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
82. Iron transport pathways in the human malaria parasite Plasmodium falciparum revealed by RNA-sequencing.
- Author
-
Wunderlich, Juliane, Kotov, Vadim, Votborg-Novél, Lasse, Ntalla, Christina, Geffken, Maria, Peine, Sven, Portugal, Silvia, and Strauss, Jan
- Subjects
PROTEIN structure prediction ,IRON in the body ,ERYTHROCYTES ,TRANSFERRIN receptors ,HEME oxygenase ,TRANSFERRIN ,FERRITIN - Abstract
Host iron deficiency is protective against severe malaria as the human malaria parasite Plasmodium falciparum depends on bioavailable iron from its host to proliferate. The essential pathways of iron acquisition, storage, export, and detoxification in the parasite differ from those in humans, as orthologs of the mammalian transferrin receptor, ferritin, or ferroportin, and a functional heme oxygenase are absent in P. falciparum. Thus, the proteins involved in these processes may be excellent targets for therapeutic development, yet remain largely unknown. Here, we show that parasites cultured in erythrocytes from an iron-deficient donor displayed significantly reduced growth rates compared to those grown in red blood cells from healthy controls. Sequencing of parasite RNA revealed diminished expression of genes involved in overall metabolism, hemoglobin digestion, and metabolite transport under low-iron versus control conditions. Supplementation with hepcidin, a specific ferroportin inhibitor, resulted in increased labile iron levels in erythrocytes, enhanced parasite replication, and transcriptional upregulation of genes responsible for merozoite motility and host cell invasion. Through endogenous GFP tagging of differentially expressed putative transporter genes followed by confocal live-cell imaging, proliferation assays with knockout and knockdown lines, and protein structure predictions, we identified six proteins that are likely required for ferrous iron transport in P. falciparum. Of these, we localized Pf VIT and Pf ZIPCO to cytoplasmic vesicles, Pf MRS3 to the mitochondrion, and the novel putative iron transporter Pf E140 to the plasma membrane for the first time in P. falciparum. Pf NRAMP/ Pf DMT1 and Pf CRT were previously reported to efflux Fe
2+ from the digestive vacuole. Our data support a new model for parasite iron homeostasis, in which Pf E140 is involved in iron uptake across the plasma membrane, Pf MRS3 ensures non-redundant Fe2+ supply to the mitochondrion as the main site of iron utilization, Pf VIT transports excess iron into cytoplasmic vesicles, and Pf ZIPCO exports Fe2+ from these organelles in case of iron scarcity. These results provide new insights into the parasite's response to differential iron availability in its environment and into the mechanisms of iron transport in P. falciparum as promising candidate targets for future antimalarial drugs. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
83. Low miR-936-mediated upregulation of Pim-3 drives sorafenib resistance in liver cancer through ferroptosis inhibition by activating the ANKRD18A/Src/NRF2 pathway.
- Author
-
Li, Xiao, Cui, Mengna, Xu, Long, and Guo, Qie
- Subjects
MOUSE leukemia viruses ,LIVER cancer ,TRANSFERRIN receptors ,LIVER cells ,REACTIVE oxygen species ,SORAFENIB - Abstract
Objective: Sorafenib, a multikinase inhibitor, is currently the standard treatment for advanced liver cancer. However, its application has become limited by the development of drug resistance. We intended to explore the mechanisms underlying the development of sorafenib resistance, therefore identifying an effective strategy to overcome sorafenib resistance remain challenges. Methods: Here, the follow-up of liver cancer patients undergoing sorafenib therapy, as well as animal tumor challenge and treatment were performed. The sorafenib-resistant liver cancer cell lines Huh7/SOR and HepG2/SOR were also established. miRNA and mRNA microarray analyses, TargetScan prediction, dual luciferase reporter assay, RNA pull-down assay, co-mmunoprecipitation (Co-IP) and pull-down assays, a transcription factor-specific NRF2 assay, an iron detection assay, a lipid peroxidation quantification assay, a ROS measurement assay, and GSH/GSSG and GSH-px standard quantitative assays were used. Results: We showed that upregulation of the provirus-integrating site for Moloney murine leukemia virus 3 (Pim-3) predicted poor response and unsatisfactory prognosis in sorafenib-treated liver cancer patients. Similarly, Pim-3 expression was positively associated with sorafenib resistance in liver cancer cells. Furthermore, microRNA-936 (miR-936) targeted the 3'-noncoding region (3'-UTR) of Pim-3 but exhibited lower expression in sorafenib-resistant liver cancer cells than in their parental cells. The high expression of Pim-3 mediated by miR-936 insufficiency activated the ANKRD18A/Src/NRF2 pathway which rearranged the expression of the indicated markers involved in iron distribution and lipid peroxidation homeostasis. MiR-936 overexpression and GV102-Pim-3-shRNA significantly attenuated the activity of the ANKRD18A/Src/NRF2 pathway to decrease the expression of Ankyrin repeat domain-containing protein 18A (ANKRD18A), Src, and Nuclear factor (erythroid-derived 2)-like 2 (NRF2), especially decreasing NRF2 nuclear retention and transcriptional activity. The transcriptional activity of NRF2 prompted cell ferroptosis because the transfection of miR-936 mimics, GV102-Pim-3-shRNA and GV102-NRF2-shRNA plasmid increased the expression of transferrin receptor 1 (TFR1) and divalent metal transporter 1 (DMT1) but decreased the expression of solute carrier family 7 member 11 (SLC7A11), glutathione peroxidase 4 (GPX4), quinone oxidoreductase 1 (NQO1), and heme oxygenase-1 (HO-1), thus facilitating the accumulation of intracellular Fe
2+ , lipid peroxides, and reactive oxygen species (ROS) but reducing the glutathione (GSH) level. Moreover, the elevated expression of Pim-3, resulting from the absence of miR-936 enhances sorafenib resistance in liver cancer by inhibiting cell ferroptosis. Conclusion: Pim-3 can be regarded as a target in the treatment of sorafenib-resistant liver cancer. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
84. Triggered ferroptotic albumin-tocopherol nanocarriers for treating drug-resistant breast cancer.
- Author
-
Gao, Qianqian, Liu, Tingting, Sun, Li, Yao, Yongliang, Li, Fang, and Mao, Lingxiang
- Subjects
TRANSFERRIN receptors ,BIOMATERIALS ,INDOCYANINE green ,REACTIVE oxygen species ,GLUTATHIONE peroxidase - Abstract
Ferroptosis is considered an effective method to overcome drug-resistant tumors. This study aims to use three FDA-approved biological materials, human serum albumin, D-α-tocopherol succinate, and indocyanine green, to construct a novel biocompatible nanomaterial named HTI-NPs, exploring its effect in drug-resistant breast cancer (MCF-7/ADR cells). The research results indicate that HTI-NPs can selectively inhibit the proliferation of MCF-7/ADR cells in vitro , accompanied by upregulating transferrin receptor, generating reactive oxygen species, and downregulating glutathione peroxidase 4. Under laser irradiation, HTI-NPs can promote ferroptosis by inhibiting glutathione expression through photodynamic therapy. Notably, HTI-NPs exhibit good inhibitory effects on MCF-7/ADR xenograft tumors in vivo. In conclusion, HTI-NPs represent a biocompatible nanomaterial that induces ferroptosis, providing new insights and options for treating drug-resistant breast cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
85. Overexpression of Nfs1 Cysteine Desulphurase Relieves Sevoflurane‐Induced Neurotoxicity and Cognitive Dysfunction in Neonatal Mice Via Suppressing Oxidative Stress and Ferroptosis.
- Author
-
Zhang, Yang, Liu, Xinru, Xie, Lijuan, Hong, Jin, Zhuang, Qin, Ren, Li, Li, Xiaohong, and Zhang, Congli
- Subjects
IRON in the body ,YOUNG adults ,TRANSFERRIN receptors ,IRON metabolism ,OXYGEN detectors - Abstract
Clinical evidence suggests that multiple exposures to sevoflurane in young people may be detrimental to cognitive development. Iron accumulation in the hippocampus is associated with sevoflurane‐induced neurotoxicity and cognitive deficits. The cysteine desulphurase, Nfs1, the rate‐limiting enzyme for the biosynthesis of iron–sulphur clusters, plays a role in cellular iron homeostasis. However, the impact of Nfs1‐mediated ferroptosis on sevoflurane‐induced neurotoxicity and cognitive impairments in neonatal mice remains undetermined. Neonatal mice at postnatal Day 6 received 3% sevoflurane daily for 3 consecutive days. Cognitive function was assessed using the Morris water maze test, and neurotoxicity was evaluated through terminal deoxynucleotidyl transferase dUTP nick end labeling and immunofluorescence staining. Here, HT22 hippocampal neurons were employed for in‐vitro experiments, and Fe2+ accumulation was measured. Ferroptosis‐related genes, including glutathione peroxidase 4 (GPX4), transferrin receptor 1 (TFR1) and ferritin, in the hippocampus and HT22 cells were observed, along with oxidative stress‐related indicators such as reactive oxygen species (ROS), methionine adenosyltransferase (MAT), glutathione (GSH) and lipid peroxidation (LPO). Transmission electron microscopy was utilized to examine the mitochondrial microstructure. Sevoflurane exposure significantly decreased Nfs1 expression in the hippocampus of mice and HT22 cells. This exposure resulted in cognitive impairments and neuronal damage in the hippocampus, which were alleviated by overexpression of Nfs1. Intracellular and mitochondrial iron accumulation occurred in HT22 cells following sevoflurane treatment. Sevoflurane exposure also significantly reduced GSH levels and increased levels of malondialdehyde, ROS and LPO in the hippocampus or HT22 cells. Additionally, sevoflurane exposure decreased GPX4 expression but increased TFR1 and ferritin expression in the hippocampus or HT22 cells. Overexpression of Nfs1 reversed the sevoflurane‐induced alterations in ferroptosis‐related genes and oxidative stress‐related indicators. Furthermore, overexpression of Nfs1 alleviated sevoflurane‐induced mitochondrial dysfunction. However, Nfs1 knockdown alone did not result in cognitive impairments, ferroptosis or oxidative stress. The overexpression of Nfs1 mitigated sevoflurane‐induced neurotoxicity and cognitive impairment by modulating oxidative stress and ferroptosis through the regulation of iron metabolism and transport. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
86. Strenuous training combined with erythropoietin induces red cell volume expansion-mediated hypervolemia and alters systemic and skeletal muscle iron homeostasis.
- Author
-
Ryan, Benjamin J., E. Barney Jr., David, McNiff, Julie L., Drummer, Devin J., Howard, Emily E., Gwin, Jess A., Carrigan, Christopher T., Murphy, Nancy E., Wilson, Marques A., Pasiakos, Stefan M., McClung, James P., and Margolis, Lee M.
- Subjects
IRON in the body ,MUSCLE proteins ,TRANSFERRIN receptors ,BLOOD volume ,VASTUS lateralis - Abstract
Strenuous physical training increases total blood volume (BV) through expansion of plasma volume (PV) and red cell volume (RCV). In contrast, exogenous erythropoietin (EPO) treatment increases RCV but decreases PV, rendering BV stable or slightly decreased. This study aimed to determine the combined effects of strenuous training and EPO treatment on BV and markers of systemic and muscle iron homeostasis. In this longitudinal study, eight healthy nonanemic males were treated with EPO (50 IU/kg body mass, three times per week, sc) across 28 days of strenuous training (4 days/wk, exercise energy expenditures of 1,334 ± 24 kcal/day) while consuming a controlled, energy-balanced diet providing 39 ± 4 mg/day iron. Before (PRE) and after (POST) intervention, BV compartments were measured using carbon monoxide rebreathing, and markers of iron homeostasis were assessed in blood and skeletal muscle (vastus lateralis). Training + EPO increased (P < 0.01) RCV (13 ± 6%) and BV (5 ± 4%), whereas PV remained unchanged (P = 0.86). The expansion of RCV was accompanied by a large decrease in whole body iron stores, as indicated by decreased (P < 0.01) ferritin (−77 ± 10%) and hepcidin (−49 ± 23%) concentrations in plasma. Training + EPO decreased (P < 0.01) muscle protein abundance of ferritin (−25 ± 20%) and increased (P < 0.05) transferrin receptor (47 ± 56%). These novel findings illustrate that strenuous training combined with EPO results in both increased total oxygen-carrying capacity and hypervolemia in young healthy males. The decrease in plasma and muscle ferritin suggests that the marked upregulation of erythropoiesis alters systemic and tissue iron homeostasis, resulting in a decline in whole body and skeletal muscle iron stores. NEW & NOTEWORTHY: Strenuous exercise training combined with erythropoietin (EPO) treatment increases blood volume, driven exclusively by red cell volume expansion. This hematological adaptation results in increased total oxygen-carrying capacity and hypervolemia. The marked upregulation of erythropoiesis with training + EPO reduces whole body iron stores and circulating hepcidin concentrations. The finding that the abundance of ferritin in muscle decreased after training + EPO suggests that muscle may release iron to support red blood cell production. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
87. Copy Number Variations of Plasmodium vivax DBP1, EBP/DBP2, and RBP2b in Ethiopians Who Are Duffy Positive and Duffy Negative.
- Author
-
Pestana, Kareen, Ford, Anthony, Rama, Rei, Abagero, Beka, Kepple, Daniel, Tomida, Junya, Popovici, Jean, Yewhalaw, Delenasaw, and Lo, Eugenia
- Subjects
ANTIGEN receptors ,PLASMODIUM vivax ,TRANSFERRIN receptors ,CAMBODIANS ,ERYTHROCYTES - Abstract
Recent evidence challenges the belief that individuals who are Duffy-negative are resistant to Plasmodium vivax due to lacking the Duffy antigen receptor for chemokines. Erythrocyte-binding protein (EBP/DBP2) has shown moderate binding to Duffy-negative erythrocytes in vitro. Reticulocyte-binding protein 2b (RBP2b) interactions with transferrin receptor 1 suggest involvement in Duffy-negative infections. Gene copy number variations in PvDBP1 , PvEBP / DBP2 , and PvRBP2b were investigated in Duffy-positive and Duffy-negative P vivax infections from Ethiopia. Among Duffy-positive samples, 34% displayed PvDBP1 duplications (Cambodian type). In Duffy-negative infections, 30% showed duplications, mostly Cambodian type. For PvEBP / DBP2 and PvRBP2b , Duffy-positive samples exhibited higher duplication rates (1–8 copies for PvEBP / DBP2 , 46%; 1–5 copies for PvRBP2b , 43%) as compared with Duffy-negative samples (20.8% and 26%, respectively). The range of copy number variations was lower in Duffy-negative infections. Demographic and clinical factors associated with gene multiplications in both Duffy types were explored, enhancing understanding of P vivax evolution in Africans who are Duffy negative. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
88. Vitamin D inhibits ferroptosis and mitigates the kidney injury of prediabetic mice by activating the Klotho/p53 signaling pathway.
- Author
-
Chen, Hao, Zhang, Yujing, Miao, Yufan, Song, Hanlu, Tang, Lulu, Liu, Wenyi, Li, Wenjie, Miao, Jinxin, and Li, Xing
- Subjects
TRANSFERRIN receptors ,KIDNEY injuries ,REACTIVE oxygen species ,GLUTATHIONE peroxidase ,DIABETIC nephropathies - Abstract
Diabetic nephropathy (DN) is a serious public health problem worldwide, and ferroptosis is deeply involved in the pathogenesis of DN. Prediabetes is a critical period in the prevention and control of diabetes and its complications, in which kidney injury occurs. This study aimed to explore whether ferroptosis would induce kidney injury in prediabetic mice, and whether vitamin D (VD) supplementation is capable of preventing kidney injury by inhibiting ferroptosis, while discussing the potential mechanisms. High-fat diet (HFD) fed KKAy mice and high glucose (HG) treated HK-2 cells were used as experimental subjects in the current study. Our results revealed that serious injury and ferroptosis take place in the kidney tissue of prediabetic mice; furthermore, VD intervention significantly improved the kidney structure and function in prediabetic mice and inhibited ferroptosis, showing ameliorated iron deposition, enhanced antioxidant capability, reduced reactive oxygen species (ROS) and lipid peroxidation accumulation. Meanwhile, VD up-regulated Klotho, solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) expression, and down-regulated p53, transferrin receptor 1 (TFR1) and Acyl-Coenzyme A synthetase long-chain family member 4 (ACSL4) expression. Moreover, we demonstrated that HG-induced ferroptosis is antagonized by treatment of VD and knockdown of Klotho attenuates the protective effect of VD on ferroptosis in vitro. In conclusion, ferroptosis occurs in the kidney of prediabetic mice and VD owns a protective effect on prediabetic kidney injury, possibly by via the Klotho/p53 pathway, thus inhibiting hyperglycemia-induced ferroptosis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
89. Investigation of the Impact of the H310A FcRn Region Mutation on 89Zr-Immuno-PET Brain Imaging with a BBB-Shuttle Anti‑Amyloid Beta Antibody.
- Author
-
Wuensche, Thomas E., Stergiou, Natascha, Mes, Iris, Verlaan, Mariska, Kooijman, Esther J. M., Windhorst, Albert D., Jensen, Allan, Asuni, Ayodeji A., Bang-Andersen, Benny, van Dongen, Guus A. M. S., Vugts, Danielle J., and Beaino, Wissam
- Subjects
POSITRON emission tomography ,THERAPEUTICS ,RADIOCHEMICAL purification ,TRANSFERRIN receptors ,PEPTIDES - Abstract
Purpose: In the emerging field of antibody treatments for neurodegenerative diseases, reliable tools are needed to evaluate new therapeutics, diagnose and select patients, monitor disease progression, and assess therapy response. Immuno-PET combines the high affinity and exceptional specificity of monoclonal antibodies with the non-invasive imaging technique positron emission tomography (PET). Its application in neurodegenerative disease brain imaging has been limited due to the marginal uptake across the blood–brain barrier (BBB). The emergence of BBB-shuttle antibodies with enhanced uptake across the BBB extended immuno-PET to brain imaging. We recently reported about specific brain uptake of a bispecific aducanumab mTfR antibody in APP/PS1 TG mice using
89 Zr-immuno-PET. However, a sufficient target-to-background ratio was reached at a relatively late scanning time point of 7 days post-injection. To investigate if a better target-to-background ratio could be achieved earlier, an aducanumab BBB-shuttle with a mutated Fc region for reduced FcRn affinity was evaluated. Procedures: AduH310A -8D3 and Adu-8D3 were modified with DFO*-NCS and subsequently radiolabeled with89 Zr. The potential influence of the H310A mutation, modification with DFO*-NCS, and subsequent radiolabeling on the in vitro binding to amyloid-beta and mTfR1 was investigated via amyloid-beta peptide ELISA and FACS analysis using mTfR1 transfected CHO-S cells. Blood kinetics, brain uptake, in vivo PET imaging and target engagement of radiolabeled AduH310A -8D3 were evaluated and compared to non-mutated Adu-8D3 in APP/PS1 TG mice and wild-type animals as controls. Results: Radiolabeling was performed with sufficient radiochemical yields and radiochemical purity. In vitro binding to amyloid-beta and mTfR1 showed no impairment. [89 Zr]Zr-AduH310A -8D3 showed faster blood clearance and earlier differentiation of amyloid-beta-related brain uptake compared to [89 Zr]Zr-Adu-8D3. However, only half of the brain uptake was observed for [89 Zr]Zr-AduH310A -8D3. Conclusions: Although a faster blood clearance of AduH310A -8D3 was observed, it was concluded that no beneficial effects for89 Zr-immuno-PET imaging of brain uptake were obtained. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
90. Intracellular Iron Deficiency and Abnormal Metabolism, Not Ferroptosis, Contributes to Homocysteine-Induced Vascular Endothelial Cell Death.
- Author
-
Shi, Wenting, Zhang, Jing, Zhao, Wairong, Yue, Meiyan, Ma, Jie, Zeng, Silu, Tang, Jingyi, Wang, Yu, and Zhou, Zhongyan
- Subjects
VASCULAR endothelial cells ,IRON metabolism ,CELL cycle ,TRANSFERRIN receptors ,METABOLIC regulation ,DEFEROXAMINE - Abstract
Background/Objectives: Homocysteine (Hcy) and iron are factors co-related with the progression of cardiovascular diseases. The vascular endothelium is an important barrier for physiological homeostasis, and its impairment initiates cardiovascular injury. However, the mechanism underlying Hcy-caused vascular endothelial cell injury and the participation of iron are not fully elucidated. This study aims to investigate the Hcy-induced vascular endothelial injury and iron metabolism dysfunction as well as the underlying molecular mechanism. Methods: Human umbilical vein endothelial cells (HUVECs) were employed as the experimental model to examine the Hcy-induced endothelial injury and its underlying mechanism via various biochemical assays. Results: Hcy suppressed the cell viability and proliferation and caused cell death in a concentration-dependent manner. Hcy induced cell cycle arrest, apoptosis, and autophagy as well as impairment of intracellular energy metabolism. Hcy disrupted the intracellular antioxidant system and mitochondrial function by increasing intracellular ROS, MDA and mitochondrial content, and decreasing the SOD activity and mitochondrial membrane potential. Hcy significantly reduced the GSH-Px activity along with the accumulation of intracellular GSH in a concentration-dependent manner. Ferroptosis inhibitors, Ferrostatin-1 (Fer-1), and Deferoxamine (DFO) significantly decreased the Hcy-caused cytotoxicity accompanied by a reduction in dysregulated mitochondria content, but only DFO ameliorated the elevation of intracellular ROS, and neither Fer-1 nor DFO affected the Hcy-caused reduction in intracellular ATP. In addition, Hcy decreased the intracellular concentration of iron, and supplementing Hcy with various concentrations of Fe
3+ increased the cell viability and decreased the LDH release in a concentration-dependent manner. Hcy dramatically decreased the mRNA expression level of transferrin receptor while increasing the mRNA expression levels of transferrin, ferritin light chain, ferritin heavy chain, ferroportin, and SLC7A11. Moreover, Hcy suppressed the protein expression of phospho-Akt, phospho-mTOR, Beclin-1, LC3A/B, Nrf2, HO-1, phospho-MEK1/2, phospho-ERK1/2, and Caspase-3 in concentration- and time-dependent manners. Conclusions: Hcy-induced vascular endothelial injury is likely to be associated with apoptosis and autophagy, but not ferroptosis. The key underlying mechanisms are involved in the disruption of the intracellular antioxidant system and iron metabolism via regulation of PI3K/Akt/mTOR, MAPKs, Nrf2/HO-1, and iron metabolism. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
91. Osteocyte ferroptosis induced by ATF3/TFR1 contributes to cortical bone loss during ageing.
- Author
-
Yin, Ying, Chen, Guang‐Jin, Yang, Chen, Wang, Jia‐Jia, Peng, Jin‐Feng, Huang, Xiao‐Fei, Tang, Qing‐Ming, and Chen, Li‐Li
- Subjects
COMPACT bone ,TRANSCRIPTION factors ,IRON overload ,OLDER people ,OSTEOCYTES ,TRANSFERRIN receptors - Abstract
Cortical bone loss is intricately associated with ageing and coincides with iron accumulation. The precise role of ferroptosis, characterized by iron overload and lipid peroxidation, in senescent osteocytes remains elusive. We found that ferroptosis was a crucial mode of osteocyte death in cortical bone during ageing. Using a single‐cell transcriptome analysis, we identified activating transcription factor 3 (ATF3) as a critical driver of osteocyte ferroptosis. Elevated ATF3 expression in senescent osteocytes promotes iron uptake by upregulating transferrin receptor 1 while simultaneously inhibiting solute carrier family 7‐member 11‐mediated cystine import. This process leads to an iron overload and lipid peroxidation, culminating in ferroptosis. Importantly, ATF3 inhibition in aged mice effectively alleviated ferroptosis in the cortical bone and mitigated cortical bone mass loss. Taken together, our findings establish a pivotal role of ferroptosis in cortical bone loss in older adults, providing promising prevention and treatment strategies for osteoporosis and fractures. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
92. Antiproliferative and Apoptosis Triggering Potential of Paclitaxel-Based Targeted-Lipid Nanoparticles with Enhanced Cellular Internalization by Transferrin Receptors—a Study in Leukemia Cells
- Author
-
Dai, Yang, Huang, Jingcao, Xiang, Bing, Zhu, Huanling, and He, Chuan
- Published
- 2018
- Full Text
- View/download PDF
93. Stoichiometries of transferrin receptors 1 and 2 in human liver
- Author
-
Chloupková, Maja, Zhang, An-Sheng, and Enns, Caroline A.
- Published
- 2010
- Full Text
- View/download PDF
94. Investigation of Trends in the Research on Transferrin Receptor-Mediated Drug Delivery via a Bibliometric and Thematic Analysis.
- Author
-
Kaur, Tarnjot, Upadhyay, Jyoti, Pukale, Sudeep, Mathur, Ashish, and Ansari, Mohd Nazam
- Subjects
BIBLIOMETRICS ,TRANSFERRIN ,THEMATIC analysis ,TRANSFERRIN receptors ,DRUG delivery systems ,BLOOD-brain barrier ,GENE therapy - Abstract
This study systematically reviews and characterizes the existing literature on transferrin/transferrin receptor-mediated drug delivery. Transferrin is an iron-binding protein. It can be used as a ligand to deliver various proteins, genes, ions, and drugs to the target site via transferrin receptors for therapeutic or diagnostic purposes via transferrin receptors. This study is based on a cross-sectional bibliometric analysis of 583 papers limited to the subject areas of pharmacology, toxicology, and pharmaceutics as extracted from the Scopus database in mid-September 2022. The data were analyzed, and we carried out a performance analysis and science mapping. There was a significant increase in research from 2018 onward. The countries that contributed the most were the USA and China, and most of the existing research was found to be from single-country publications. Research studies on transferrin/transferrin receptor-mediated drug delivery focus on drug delivery across the blood–brain barrier in the form of nanoparticles. The thematic analysis revealed four themes: transferrin/transferrin receptor-mediated drug delivery to the brain, cancer cells, gene therapy, nanoparticles, and liposomes as drug delivery systems. This study is relevant to academics, practitioners, and decision makers interested in targeted and site-specific drug delivery. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
95. Data from Optimized Doxorubicin Chemotherapy for Diffuse Large B-cell Lymphoma Exploits Nanocarrier Delivery to Transferrin Receptors
- Author
-
Jonathan H. Schatz, Roger M. Leblanc, Daniel Bilbao, Francisco Vega, Melissa W. Taggart, Austin D. Newsam, Yuguang Ban, Zhen Gao, Braulio C.L.B. Ferreira, Lingxiao Li, Evan R. Roberts, Asaad Trabolsi, Piumi Y. Liyanage, and Artavazd Arumov
- Abstract
New treatments are needed to address persistent unmet clinical needs for diffuse large B-cell lymphoma (DLBCL). Overexpression of transferrin receptor 1 (TFR1) is common across cancer and permits cell-surface targeting of specific therapies in preclinical and clinical studies of various solid tumors. Here, we developed novel nanocarrier delivery of chemotherapy via TFR1-mediated endocytosis, assessing this target for the first time in DLBCL. Analysis of published datasets showed novel association of increased TFR1 expression with high-risk DLBCL cases. Carbon–nitride dots (CND) are emerging nanoparticles with excellent in vivo stability and distribution and are adaptable to covalent conjugation with multiple substrates. In vitro, linking doxorubicin (Dox) and transferrin (TF) to CND (CND–Dox–TF, CDT) was 10–100 times more potent than Dox against DLBCL cell lines. Gain- and loss-of-function studies and fluorescent confocal microscopy confirmed dependence of these effects on TFR1-mediated endocytosis. In contrast with previous therapeutics directly linking Dox and TF, cytotoxicity of CDT resulted from nuclear entry by Dox, promoting double-stranded DNA breaks and apoptosis. CDT proved safe to administer in vivo, and when incorporated into standard frontline chemoimmunotherapy in place of Dox, it improved overall survival by controlling patient-derived xenograft tumors with greatly reduced host toxicities. Nanocarrier-mediated Dox delivery to cell-surface TFR1, therefore, warrants optimization as a potential new therapeutic option in DLBCL.Significance:Targeted nanoparticle delivery of doxorubicin chemotherapy via the TRF1 receptor presents a new opportunity against high-risk DLBCL tumors using potency and precision.
- Published
- 2023
- Full Text
- View/download PDF
96. Supplementary Figures + Legend from Optimized Doxorubicin Chemotherapy for Diffuse Large B-cell Lymphoma Exploits Nanocarrier Delivery to Transferrin Receptors
- Author
-
Jonathan H. Schatz, Roger M. Leblanc, Daniel Bilbao, Francisco Vega, Melissa W. Taggart, Austin D. Newsam, Yuguang Ban, Zhen Gao, Braulio C.L.B. Ferreira, Lingxiao Li, Evan R. Roberts, Asaad Trabolsi, Piumi Y. Liyanage, and Artavazd Arumov
- Abstract
Supplemental Figures 1-9. Figure S1. Carbon-Nitride Dot Synthesis. Figure S2. Characterization of CND-Dox-TF. Figure S3. CND cytotoxicity. Figure S4. Cell surface TFR1 expression and receptor recycling. Figure S5. Rapid nuclear entry by Dox after CND-Dox-TF treatment in DLBCL. Figure S6. HEK293 is a model cell line for elucidating CND-Dox-TF mechanism. Figure S7. CND-Dox-TF working dose identified. Figure S8. CND-Dox-TF has improved toxicity profile compared to Dox. Figure S9. R-nanoCHOP treatment has favorable toxicity profile in non-malignant organs.
- Published
- 2023
- Full Text
- View/download PDF
97. Transport of the Ruthenium Complex [Ru(GA)(dppe)2]PF6 into Triple-Negative Breast Cancer Cells Is Facilitated by Transferrin Receptors.
- Author
-
Naves, Marina A., Graminha, Angelica E., Vegas, Legna C., Luna-Dulcey, Liany, Honorato, João, Menezes, Antônio C. S., Batista, Alzir A., and Cominetti, Marcia R.
- Published
- 2019
- Full Text
- View/download PDF
98. Photo-Cross-Linking Mass Spectrometry and Integrative Modeling Enables Rapid Screening of Antigen Interactions Involving Bacterial Transferrin Receptors.
- Author
-
Ziemianowicz, Daniel S., Ng, Dixon, Schryvers, Anthony B., and Schriemer, David C.
- Published
- 2019
- Full Text
- View/download PDF
99. Gallium Uncouples Iron Metabolism to Enhance Glioblastoma Radiosensitivity.
- Author
-
Owusu, Stephenson B., Zaher, Amira, Ahenkorah, Stephen, Pandya, Darpah N., Wadas, Thaddeus J., and Petronek, Michael S.
- Subjects
IRON metabolism ,RIBONUCLEOSIDE diphosphate reductase ,CELL metabolism ,CHARGE exchange ,GALLIUM ,TRANSFERRIN ,TRANSFERRIN receptors ,NADH dehydrogenase - Abstract
Gallium-based therapy has been considered a potentially effective cancer therapy for decades and has recently re-emerged as a novel therapeutic strategy for the management of glioblastoma tumors. Gallium targets the iron-dependent phenotype associated with aggressive tumors by mimicking iron in circulation and gaining intracellular access through transferrin-receptor-mediated endocytosis. Mechanistically, it is believed that gallium inhibits critical iron-dependent enzymes like ribonucleotide reductase and NADH dehydrogenase (electron transport chain complex I) by replacing iron and removing the ability to transfer electrons through the protein secondary structure. However, information regarding the effects of gallium on cellular iron metabolism is limited. As mitochondrial iron metabolism serves as a central hub of the iron metabolic network, the goal of this study was to investigate the effects of gallium on mitochondrial iron metabolism in glioblastoma cells. Here, it has been discovered that gallium nitrate can induce mitochondrial iron depletion, which is associated with the induction of DNA damage. Moreover, the generation of gallium-resistant cell lines reveals a highly unstable phenotype characterized by impaired colony formation associated with a significant decrease in mitochondrial iron content and loss of the mitochondrial iron uptake transporter, mitoferrin-1. Moreover, gallium-resistant cell lines are significantly more sensitive to radiation and have an impaired ability to repair any sublethal damage and to survive potentially lethal radiation damage when left for 24 h following radiation. These results support the hypothesis that gallium can disrupt mitochondrial iron metabolism and serve as a potential radiosensitizer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
100. The Functions of SARS-CoV-2 Receptors in Diabetes-Related Severe COVID-19.
- Author
-
Drzymała, Adam
- Subjects
SARS-CoV-2 ,TYPE 2 diabetes ,ANGIOTENSIN converting enzyme ,TRANSFERRIN receptors ,VIMENTIN - Abstract
Angiotensin-converting enzyme 2 (ACE2) is considered a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor of high importance, but due to its non-ubiquitous expression, studies of other proteins that may participate in virus internalisation have been undertaken. To date, many alternative receptors have been discovered. Their functioning may provide an explanation for some of the events observed in severe COVID-19 that cannot be directly explained by the model in which ACE2 constitutes the central point of infection. Diabetes mellitus type 2 (T2D) can induce severe COVID-19 development. Although many mechanisms associated with ACE2 can lead to increased SARS-CoV-2 virulence in diabetes, proteins such as basigin (CD147), glucose-regulated protein 78 kDa (GRP78), cluster of differentiation 4 (CD4), transferrin receptor (TfR), integrins α
5 β1 /αv β3 , or ACE2 co-receptors neuropilin 2 (NRP2), vimentin, and even syalilated gangliosides may also be responsible for worsening the COVID-19 course. On the other hand, some others may play protective roles. Understanding how diabetes-associated mechanisms can induce severe COVID-19 via modification of virus receptor functioning needs further extensive studies. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.