Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 307
Filter
1.
J Cell Physiol ; 237(11): 4132-4156, 2022 11.
Article in English | MEDLINE | ID: mdl-36181695

ABSTRACT

Breast cancer is one of the leading causes contributing to the global cancer burden. The triple negative breast cancer (TNBC) molecular subtype accounts for the most aggressive type. Despite progression in therapeutic options and prognosis in breast cancer treatment options, there remains a high rate of distant relapse. With advancements in understanding the role of zinc and zinc carriers in the prognosis and treatment of the disease, the scope of precision treatment/targeted therapy has been expanded. Zinc levels and zinc transporters play a vital role in maintaining cellular homeostasis, tumor surveillance, apoptosis, and immune function. This review focuses on the zinc transporter, LIV1, as an essential target for breast cancer prognosis and emerging treatment options. Previous studies give an insight into the role of LIV1 in fulfilling the most important hallmarks of cancer such as apoptosis, metastasis, invasion, and evading the immune system. Normal tissue expression of LIV1 is limited. Higher expression of LIV1 has been linked to Epithelial-Mesenchymal Transition, histological grade of cancer, and early node metastasis. LIV1 was found to be one of the attractive targets in the therapeutic hunt for TNBCs. TNBCs are an immunogenic breast cancer subtype. As zinc transporters are known to serve as the metabolic gatekeepers of immune cells, this review bridges tumor infiltrating lymphocytes, TNBC and LIV1. In addition, the suitability of LIV1 as an antibody-drug conjugate (Seattle genetics [SGN]-LIV1A) target in TNBC, represents a promising strategy for patients. Early clinical trial results reveal that this novel agent reduces tumor burden by inducing mitotic arrest, immunomodulation, and immunogenic cell death, warranting further investigation of SGN-LIV1A in combination with immuno-oncology agents. Priming the patient's immune response in combination with SGN-LIV1A could eventually change the landscape for the TNBC patient population.


Subject(s)
Cation Transport Proteins , Triple Negative Breast Neoplasms , Humans , Biomarkers, Tumor/therapeutic use , Carrier Proteins , Neoplasm Recurrence, Local , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Zinc/metabolism , Cation Transport Proteins/antagonists & inhibitors
2.
Cancer Res ; 81(23): 5991-6003, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34706862

ABSTRACT

Melanoma and most other cancers occur more frequently and have worse prognosis in males compared with females. Although sex steroids are thought to be involved, classical androgen and estrogen receptors are not detectable in most melanomas. Here we show that testosterone promotes melanoma proliferation by activating ZIP9 (SLC39A9), a zinc transporter that is widely expressed in human melanoma but not intentionally targeted by available therapeutics. This testosterone activity required an influx of zinc, activation of MAPK, and nuclear translocation of YAP. FDA-approved inhibitors of the classical androgen receptor also inhibited ZIP9, thereby antagonizing the protumorigenic effects of testosterone in melanoma. In male mice, androgen receptor inhibitors suppressed growth of ZIP9-expressing melanomas but had no effect on isogenic melanomas lacking ZIP9 or on melanomas in females. These data suggest that ZIP9 might be effectively targeted in melanoma and other cancers by repurposing androgen receptor inhibitors that are currently approved only for prostate cancer. SIGNIFICANCE: Testosterone signaling through ZIP9 mediates some of the sex differences in melanoma, and drugs that target AR can be repurposed to block ZIP9 and inhibit melanoma in males.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Cation Transport Proteins/antagonists & inhibitors , Melanoma/drug therapy , Receptors, Androgen/chemistry , Testosterone/pharmacology , Androgens/pharmacology , Animals , Apoptosis , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Movement , Cell Proliferation , Humans , Male , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, SCID , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Sex Factors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Nat Commun ; 12(1): 5311, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493724

ABSTRACT

Although some effective therapies have been available for cancer, it still poses a great threat to human health and life due to its drug resistance and low response in patients. Here, we develop a ferroptosis-based therapy by combining iron nanoparticles and cancer-specific gene interference. The expression of two iron metabolic genes (FPN and LCN2) was selectively knocked down in cancer cells by Cas13a or microRNA controlled by a NF-κB-specific promoter. Cells were simultaneously treated by iron nanoparticles. As a result, a significant ferroptosis was induced in a wide variety of cancer cells. However, the same treatment had little effect on normal cells. By transferring genes with adeno-associated virus and iron nanoparticles, the significant tumor growth inhibition and durable cure were obtained in mice with the therapy. In this work, we thus show a cancer therapy based on gene interference-enhanced ferroptosis.


Subject(s)
Cation Transport Proteins/antagonists & inhibitors , Ferroptosis/genetics , Iron/metabolism , Lipocalin-2/antagonists & inhibitors , Neoplasms/therapy , RNA Interference , Reactive Oxygen Species/agonists , Animals , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line, Tumor , Dependovirus/genetics , Dependovirus/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lipocalin-2/genetics , Lipocalin-2/metabolism , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Promoter Regions, Genetic , Reactive Oxygen Species/metabolism , Signal Transduction , Spleen/metabolism , Spleen/pathology , Survival Analysis , Tumor Burden , Xenograft Model Antitumor Assays
4.
ChemMedChem ; 16(21): 3306-3314, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34309203

ABSTRACT

Solute carrier proteins (SLCs) are membrane proteins controlling fluxes across biological membranes and represent an emerging class of drug targets. Here we searched for inhibitors of divalent metal transporters in a library of 1,676 commercially available 3D-shaped fragment-like molecules from the generated database GDB-17, which lists all possible organic molecules up to 17 atoms of C, N, O, S and halogen following simple criteria for chemical stability and synthetic feasibility. While screening against DMT1 (SLC11A2), an iron transporter associated with hemochromatosis and for which only very few inhibitors are known, only yielded two weak inhibitors, our approach led to the discovery of the first inhibitor of ZIP8 (SLC39A8), a zinc transporter associated with manganese homeostasis and osteoarthritis but with no previously reported pharmacology, demonstrating that this target is druggable.


Subject(s)
Carbazoles/pharmacology , Carboxylic Acids/pharmacology , Cation Transport Proteins/antagonists & inhibitors , Sulfones/pharmacology , Carbazoles/chemistry , Carboxylic Acids/chemistry , Cation Transport Proteins/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Molecular Structure , Structure-Activity Relationship , Sulfones/chemistry
5.
Free Radic Res ; 55(7): 821-830, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34181495

ABSTRACT

Ferroptosis is a newly described regulated form of cell death that contributes to the progression of non-small cell lung cancers (NSCLCs). MicroRNA-302a-3p (miR-302a-3p) plays critical roles in the tumorigenicity of different cancers; however, its function and underlying mechanism in ferroptosis and NSCLCs remain unclear. Human NSCLCs cells were incubated with miR-302a-3pmimic or inhibitor in the presence or absence of erastin or RSL3. Cell viability, colony numbers, lactate dehydrogenase (LDH) releases, lipid peroxidation and intracellular iron level were measured. Besides, the synergistic effects of cisplatin and paclitaxel with miR-302a-3p were determined. miR-302a-3p level was reduced in human NSCLCs cells and tissues. ThemiR-302a-3p mimic induced lipid peroxidation, iron overload and ferroptosis, thereby inhibiting cell growth and colony formation of NSCLCs cells. Conversely, the miR-302a-3p inhibitor block ederastin- or RSL3-related ferroptosis and tumor suppression. Additionally, we found that miR-302a-3p directly bound to the 3'-untranslational region of ferroportin to decrease its protein expression, and that ferroportin overexpression significantly prevented miR-302a-3p mimic-induced ferroptosis and tumor inhibition. Moreover, the miR-302a-3p mimic sensitized NSCLCs cells to cisplatin and paclitaxel chemotherapy. miR-302a-3p functions as a tumor inhibitor, at least partly, via targeting ferroportin to induce ferroptosis of NSCLCs.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cation Transport Proteins/antagonists & inhibitors , Ferroptosis , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , MicroRNAs/genetics , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Proliferation , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Tumor Cells, Cultured
6.
Expert Opin Ther Pat ; 31(7): 585-595, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33975503

ABSTRACT

INTRODUCTION: Iron is a crucial element necessary for blood formation in the body and its normal growth. However, irregular metabolism of iron due to absence of an elimination mechanism may deposit excess iron in the organs (iron overload) leading to metabolic disorders. Interactions between the iron regulatory peptide hormone, hepcidin and the iron exporter ferroportin plays major role in regulating the iron metabolism. Mutations in the ferroportin encoding genes, and dysregulation of hepsidin production often results in iron overload resulting in conditions like hemochromatosis, ß-thalassemia, and sickle cell anemia. Until today, there is no efficacious treatment available for managing iron overload targeting ferroportin inhibition via oral administration. AREAS COVERED: Novel salts of substituted benzoimidazole compounds useful for the prophylaxis and/or treatment of iron overload are claimed. These compounds act as hepcidin mimetic and inhibit the ferroportin thereby preventing iron overload. The claimed actives are useful in the treatment of disease conditions such as neurodegenerative and cardiac diseases triggered by iron overload. Preclinical studies of these salts on mouse model are also discussed. EXPERT OPINION: Prevention and/or treatment of iron overload is critical. The claimed compounds are the first oral drug candidate to treat iron overload and reach the pre-clinical development stage.


Subject(s)
Benzimidazoles/pharmacology , Cation Transport Proteins/antagonists & inhibitors , Iron Overload/drug therapy , Administration, Oral , Animals , Benzimidazoles/chemistry , Cation Transport Proteins/metabolism , Disease Models, Animal , Drug Development , Hepcidins/metabolism , Humans , Iron/metabolism , Iron Overload/physiopathology , Mice , Patents as Topic , Salts
7.
Biomolecules ; 11(4)2021 04 16.
Article in English | MEDLINE | ID: mdl-33923411

ABSTRACT

Candidiasis is the wide-spread fungal infection caused by numerous strains of yeast, with the prevalence of Candida albicans. The current treatment of candidiasis is becoming rather ineffective and costly owing to the emergence of resistant strains; hence, the exploration of new possible drug targets is necessary. The most promising route is the development of novel antibiotics targeting this pathogen. In this review, we summarize such candidates found in C. albicans and those involved in the transport of (metal) cations, as the latter are essential for numerous processes within the cell; hence, disruption of their fluxes can be fatal for C. albicans.


Subject(s)
Antifungal Agents/pharmacology , Candidiasis/drug therapy , Cation Transport Proteins/metabolism , Fungal Proteins/metabolism , Animals , Antifungal Agents/therapeutic use , Candida albicans/drug effects , Candida albicans/metabolism , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/chemistry , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/chemistry , Humans
8.
Cell Death Dis ; 12(2): 198, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33608508

ABSTRACT

Ferroptosis is a newly described form of regulated cell death triggered by oxidative stresses and characterized by extensive lipid peroxidation and membrane damages. The name of ferroptosis indicates that the ferroptotic death process depends on iron, but not other metals, as one of its canonical features. Here, we reported that zinc is also essential for ferroptosis in breast and renal cancer cells. Zinc chelator suppressed ferroptosis, and zinc addition promoted ferroptosis, even during iron chelation. By interrogating zinc-related genes in a genome-wide RNAi screen of ferroptosis, we identified SLC39A7, encoding ZIP7 that controls zinc transport from endoplasmic reticulum (ER) to cytosol, as a novel genetic determinant of ferroptosis. Genetic and chemical inhibition of the ZIP7 protected cells against ferroptosis, and the ferroptosis protection upon ZIP7 knockdown can be abolished by zinc supplementation. We found that the genetic and chemical inhibition of ZIP7 triggered ER stresses, including the induction of the expression of HERPUD1 and ATF3. Importantly, the knockdown of HERPUD1 abolished the ferroptosis protection phenotypes of ZIP7 inhibition. Together, we have uncovered an unexpected role of ZIP7 in ferroptosis by maintaining ER homeostasis. These findings may have therapeutic implications for human diseases involving ferroptosis and zinc dysregulations.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Renal Cell/metabolism , Cation Transport Proteins/metabolism , Endoplasmic Reticulum/metabolism , Ferroptosis , Kidney Neoplasms/metabolism , Zinc/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/genetics , Cell Line, Tumor , Chelating Agents/pharmacology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Female , Ferroptosis/drug effects , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism
9.
Eur J Pharmacol ; 898: 173986, 2021 May 05.
Article in English | MEDLINE | ID: mdl-33640406

ABSTRACT

The high-affinity choline transporter CHT1 mediates choline uptake, the rate-limiting and regulatory step in acetylcholine synthesis at cholinergic presynaptic terminals. CHT1-medated choline uptake is specifically inhibited by hemicholinium-3, which is a type of choline analog that acts as a competitive inhibitor. Although the substrate choline and the inhibitor hemicholinium-3 are well-established ligands of CHT1, few potent ligands other than choline analogs have been reported. Here we show that tetrahydropyrimidine anthelmintics, known as nicotinic acetylcholine receptor agonists, act as competitive inhibitors of CHT1. A ligand-dependent trafficking assay in cell lines expressing human CHT1 was designed to search for CHT1 ligands from a collection of biologically active compounds. We found that morantel as well as other tetrahydropyrimidines, pyrantel and oxantel, potently inhibits the high-affinity choline uptake activity of CHT1 in a competitive manner similar to the inhibitor hemicholinium-3. They also inhibit the high-affinity choline transporter from the nematode Caenorhabditis elegans. Finally, tetrahydropyrimidines potently inhibit the high-affinity choline uptake in rat brain synaptosomes at a low micromolar level, resulting in the inhibition of acetylcholine synthesis. The rank order of potency in synaptosomes is as follows: morantel > pyarantel > oxantel (Ki = 1.3, 5.7, and 8.3 µM, respectively). Our results reveal that tetrahydropyrimidine anthelmintics are novel CHT1 ligands that inhibit the high-affinity choline uptake for acetylcholine synthesis in cholinergic neurons.


Subject(s)
Anthelmintics/pharmacology , Brain/drug effects , Cation Transport Proteins/antagonists & inhibitors , Choline/metabolism , Pyrimidines/pharmacology , Symporters/antagonists & inhibitors , Animals , Anthelmintics/metabolism , Binding, Competitive , Biological Transport , Brain/metabolism , Cation Transport Proteins/metabolism , Female , HEK293 Cells , Humans , Ligands , Mice , Morantel/metabolism , Morantel/pharmacology , Protein Binding , Protein Transport , Pyrantel/analogs & derivatives , Pyrantel/metabolism , Pyrantel/pharmacology , Pyrimidines/metabolism , Symporters/genetics , Symporters/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism
10.
Autophagy ; 17(11): 3361-3374, 2021 11.
Article in English | MEDLINE | ID: mdl-33404288

ABSTRACT

Macroautophagy (hereafter referred to as "autophagy") is a lysosome-mediated degradation process that plays a complex role in cellular stress, either promoting survival or triggering death. Early studies suggest that ferroptosis, an iron-dependent form of regulated cell death, is not related to autophagy. Conversely, recent evidence indicates that the molecular machinery of autophagy facilitates ferroptosis through the selective degradation of anti-ferroptosis regulators. However, the mechanism of autophagy-dependent ferroptosis remains incompletely understood. Here, we examine the early dynamic change in protein expression of autophagic (e.g., MAP1LC3B and SQSTM1) or ferroptotic (e.g., SLC7A11 and GPX4) regulators in 60 human cancer cell lines in response to two classical ferroptosis activators (erastin and RSL3) in the absence or presence of the lysosomal inhibitor chloroquine. Compared to erastin, RSL3 exhibits wider and stronger activity in the upregulation of MAP1LC3B-II or downregulation of SQSTM1 in 80% (48/60) or 63% (38/60) of cell lines, respectively. Both RSL3 and erastin failed to affect SLC7A11 expression, but they led to GPX4 downregulation in 12% (7/60) and 3% (2/60) of cell lines, respectively. Additionally, the intracellular iron exporter SLC40A1/ferroportin-1 was identified as a new substrate for autophagic elimination, and its degradation by SQSTM1 promoted ferroptosis in vitro and in xenograft tumor mouse models. Together, these findings show tumor heterogeneity in autophagy-dependent ferroptosis, which might have different biological behaviors with regard to the dynamic characteristics of cell death.Abbreviations: ATG: Autophagy-related; CQ: Chloroquine; GPX4: Glutathione peroxidase 4; MAP1LC3B/LC3: Microtubule-associated protein 1 light chain 3 beta: NCOA4: Nuclear Receptor Coactivator 4; ROS: Reactive Oxygen Species; SLC40A1/ferroportin-1: Solute Carrier family 40 Member 1; SLC7A11: Solute Carrier Family 7 Member 11; SQSTM1/p62: Sequestosome 1.


Subject(s)
Autophagy/physiology , Ferroptosis/physiology , Neoplasms/pathology , Neoplasms/physiopathology , Animals , Autophagy/genetics , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line, Tumor , Ferroptosis/genetics , Gene Knockdown Techniques , Heterografts , Humans , Mice , Mice, Nude , Microtubule-Associated Proteins/metabolism , Neoplasms/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Sequestosome-1 Protein/metabolism
11.
Int J Mol Sci ; 22(2)2021 Jan 16.
Article in English | MEDLINE | ID: mdl-33467196

ABSTRACT

In ß-thalassemia, ineffective erythropoiesis leads to anemia and systemic iron overload. The management of iron overload by chelation therapy is a standard of care. However, iron chelation does not improve the ineffective erythropoiesis. We recently showed that the oral ferroportin inhibitor VIT-2763 ameliorates anemia and erythropoiesis in the Hbbth3/+ mouse model of ß-thalassemia. In this study, we investigated whether concurrent use of the iron chelator deferasirox (DFX) and the ferroportin inhibitor VIT-2763 causes any pharmacodynamic interactions in the Hbbth3/+ mouse model of ß-thalassemia. Mice were treated with VIT-2763 or DFX alone or with the combination of both drugs once daily for three weeks. VIT-2763 alone or in combination with DFX improved anemia and erythropoiesis. VIT-2763 alone decreased serum iron and transferrin saturation (TSAT) but was not able to reduce the liver iron concentration. While DFX alone had no effect on TSAT and erythropoiesis, it significantly reduced the liver iron concentration alone and in the presence of VIT-2763. Our results clearly show that VIT-2763 does not interfere with the iron chelation efficacy of DFX. Furthermore, VIT-2763 retains its beneficial effects on improving ineffective erythropoiesis when combined with DFX in the Hbbth3/+ mouse model. In conclusion, co-administration of the oral ferroportin inhibitor VIT-2763 and the iron chelator DFX is feasible and might offer an opportunity to improve both ineffective erythropoiesis and iron overload in ß-thalassemia.


Subject(s)
Benzimidazoles/pharmacology , Erythropoiesis/drug effects , Oxazoles/pharmacology , Pyridines/pharmacology , beta-Thalassemia/drug therapy , Administration, Oral , Animals , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , Benzimidazoles/therapeutic use , Cation Transport Proteins/antagonists & inhibitors , Cells, Cultured , Deferasirox/administration & dosage , Deferasirox/pharmacology , Deferasirox/therapeutic use , Drug Combinations , Drug Interactions , Female , Iron/blood , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Male , Mice , Mice, Inbred C57BL , Oxazoles/administration & dosage , Oxazoles/adverse effects , Oxazoles/therapeutic use , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/therapeutic use , Transferrin/metabolism
12.
J Cereb Blood Flow Metab ; 41(3): 641-655, 2021 03.
Article in English | MEDLINE | ID: mdl-32501158

ABSTRACT

The mechanism of early blood-brain barrier (BBB) disruption after stroke has been intensively studied but still not fully understood. Here, we report that microRNA-30a (miR-30a) could mediate BBB damage using both cellular and animal models of ischemic stroke. In the experiments in vitro, inhibition of miR-30a decreased BBB permeability, prevented the degradation of tight junction proteins, and reduced intracellular free zinc in endothelial cells. We found that the zinc transporter ZnT4 was a direct target of negative regulation by miR-30a, and ZnT4/zinc signaling pathway contributed significantly to miR-30a-mediated BBB damage. Consistent with these in vitro findings, treatment with miR-30a inhibitor reduced zinc accumulation, increased the expression of ZnT4, and prevented the loss of tight junction proteins in microvessels of ischemic animals. Furthermore, inhibition of miR-30a, even at 90 min post onset of middle cerebral artery occlusion, prevented BBB damage, reduced infarct volume, and ameliorated neurological deficits. Together, our findings provide novel insights into the mechanisms of cerebral ischemia-induced BBB disruption and indicate miR-30a as a regulator of BBB function that can be an effective therapeutic target for ischemic stroke.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Ischemia/pathology , Cation Transport Proteins/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Antagomirs/pharmacology , Antagomirs/therapeutic use , Blood-Brain Barrier/drug effects , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/genetics , Cell Line , Cell Survival , Claudin-5/metabolism , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Microvessels/metabolism , Occludin/metabolism , Permeability/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Zinc/metabolism
13.
J Integr Plant Biol ; 63(3): 528-542, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32877013

ABSTRACT

Type 2C protein phosphatases (PP2Cs) are the largest protein phosphatase family. PP2Cs dephosphorylate substrates for signaling in Arabidopsis, but the functions of most PP2Cs remain unknown. Here, we characterized PP2C49 (AT3G62260, a Group G PP2C), which regulates Na+ distribution under salt stress and is localized to the cytoplasm and nucleus. PP2C49 was highly expressed in root vascular tissues and its disruption enhanced plant tolerance to salt stress. Compared with wild type, the pp2c49 mutant contained more Na+ in roots but less Na+ in shoots and xylem sap, suggesting that PP2C49 regulates shoot Na+ extrusion. Reciprocal grafting revealed a root-based mechanism underlying the salt tolerance of pp2c49. Systemic Na+ distribution largely depends on AtHKT1;1 and loss of function of AtHKT1;1 in the pp2c49 background overrode the salt tolerance of pp2c49, resulting in salt sensitivity. Furthermore, compared with plants overexpressing PP2C49 in the wild-type background, plants overexpressing PP2C49 in the athtk1;1 mutant background were sensitive to salt, like the athtk1;1 mutants. Moreover, protein-protein interaction and two-voltage clamping assays demonstrated that PP2C49 physically interacts with AtHKT1;1 and inhibits the Na+ permeability of AtHKT1;1. This study reveals that PP2C49 negatively regulates AtHKT1;1 activity and thus determines systemic Na+ allocation during salt stress.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Arabidopsis/physiology , Cation Transport Proteins/antagonists & inhibitors , Protein Phosphatase 2C/metabolism , Salt Tolerance/physiology , Symporters/antagonists & inhibitors , Abscisic Acid/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/antagonists & inhibitors , Arabidopsis Proteins/genetics , Cation Transport Proteins/metabolism , Gene Expression Regulation, Plant/drug effects , Mutation/genetics , Phenotype , Plant Roots/drug effects , Plant Roots/metabolism , Plant Shoots/drug effects , Plant Shoots/metabolism , Protein Binding/drug effects , Protein Phosphatase 2C/genetics , Signal Transduction/drug effects , Sodium/metabolism , Sodium Chloride/pharmacology , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Symporters/metabolism , Xylem/metabolism
14.
Biochem Biophys Res Commun ; 533(4): 1004-1011, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33012507

ABSTRACT

Zinc is an essential trace element and participates in a variety of biological processes. ZnT (SLC30) family members are generally responsible for zinc efflux across the membrane regulating zinc homeostasis. In mammals, the only predominantly plasma membrane resident ZnT has been reported to be ZnT1, and ZnT1-/ZnT1- mice die at the embryonic stage. In Drosophila, knock down of ZnT1 homologue (dZnT1//ZnT63C/CG17723) results in growth arrest under zinc-limiting conditions. To investigate the essentiality of dZnT1 for zinc homeostasis, as well as its role in dietary zinc uptake especially under normal physiological conditions, we generated dZnT1 mutants by the CRISPER/Cas9 method. Homozygous mutant dZnT1 is lethal, with substantial zinc accumulation in the iron cell region, posterior midgut as well as gastric caeca. Expression of human ZnT1 (hZnT1), in the whole body or in the entire midgut, fully rescued the dZnT1 mutant lethality, whereas tissue-specific expression of hZnT1 in the iron cell region and posterior midgut partially rescued the developmental defect of the dZnT1 mutant. Supplementation of zinc together with clioquinol or hinokitiol conferred a limited but observable rescue upon dZnT1 loss. Our work demonstrated the absolute requirement of dZnT1 in Drosophila survival and indicated that the most essential role of dZnT1 is in the gut.


Subject(s)
Cation Transport Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Zinc/metabolism , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/genetics , Diet , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Female , Gene Knockdown Techniques , Genes, Insect , Humans , Intestinal Absorption/genetics , Intestinal Absorption/physiology , Male , Mutation , Phylogeny , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Trace Elements/administration & dosage , Trace Elements/metabolism , Trace Elements/pharmacokinetics , Zinc/administration & dosage , Zinc/pharmacokinetics
15.
Int J Mol Sci ; 21(20)2020 Oct 10.
Article in English | MEDLINE | ID: mdl-33050492

ABSTRACT

A brand new approach to the understanding of breast cancer (BC) is urgently needed. In this contribution, the etiology, pathogenesis, and treatment of this disease is approached from the new pH-centric anticancer paradigm. Only this unitarian perspective, based upon the hydrogen ion (H+) dynamics of cancer, allows for the understanding and integration of the many dualisms, confusions, and paradoxes of the disease. The new H+-related, wide-ranging model can embrace, from a unique perspective, the many aspects of the disease and, at the same time, therapeutically interfere with most, if not all, of the hallmarks of cancer known to date. The pH-related armamentarium available for the treatment of BC reviewed here may be beneficial for all types and stages of the disease. In this vein, we have attempted a megasynthesis of traditional and new knowledge in the different areas of breast cancer research and treatment based upon the wide-ranging approach afforded by the hydrogen ion dynamics of cancer. The concerted utilization of the pH-related drugs that are available nowadays for the treatment of breast cancer is advanced.


Subject(s)
Breast Neoplasms/metabolism , Hydrogen/metabolism , Protons , Animals , Antineoplastic Agents , Biomarkers, Tumor , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms/etiology , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/metabolism , Cell Respiration/drug effects , Clinical Studies as Topic , Combined Modality Therapy , Disease Management , Drug Resistance, Neoplasm , Female , Humans , Hydrogen-Ion Concentration , Intracellular Space , Molecular Targeted Therapy , Proton Pump Inhibitors/pharmacology , Proton Pump Inhibitors/therapeutic use , Proton Pumps/metabolism , Sodium-Hydrogen Exchangers/metabolism , Translational Research, Biomedical , Treatment Outcome , Tumor Microenvironment
16.
Toxicology ; 440: 152489, 2020 07.
Article in English | MEDLINE | ID: mdl-32416107

ABSTRACT

Busulfan is commonly used for cancer chemotherapy, nevertheless it cause male infertility via damaging the germ cells. Therefore, the underlying mechanism should be explored. In the present study, we demonstrated for the first time that ferroptosis was involved in busulfan-induced oligospermia in mice. Mice were given testicular injection of busulfan on both sides at the dose of 4 mg/kg body weight to establish the model of oligospermia. Four weeks later, the results showed that busulfan-treated mice exhibited decreased sperm concentration and motility, along with features of typical ferroptosis in testis, such as increased malondialdehyde (MDA) content and prostaglandin-endoperoxide synthase (PTGS2) mRNA expression, and decreased NADPH content. Inhibition of ferroptosis by ferrostatin-1 (Fer-1) or deferoxamine (DFO) partially alleviated busulfan-induced oligospermia in mice. Additionally, we also revealed that busulfan treatment induced spermatogenic cells ferroptosis by down-regulating nuclear factor-E2-related factor 2 (Nrf2) and glutathione peroxidase 4 (GPX4) expressions, and decreasing iron efflux through reduction of ferroportin 1 (FPN1) expression. Fer-1 or DFO obviously reversed busulfan-induced ferroptosis by increasing Nrf2, GPX4 and FPN1 expressions. Furthermore, after activation of Nrf2 by sulforaphane, sperm concentration and motility in busulfan-treated mice increased, accompanied by enhanced expressions of GPX4 and FPN1. These findings imply that busulfan-induced ferroptosis might be mediated via inhibition of Nrf2-GPX4 (FPN1) signaling pathway, and highlight that targeting ferroptosis serves as a potential strategy for prevention of busulfan-induced damage and male infertility.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Busulfan/toxicity , Ferroptosis/drug effects , Oligospermia/chemically induced , Oligospermia/prevention & control , Animals , Cation Transport Proteins/antagonists & inhibitors , Cyclohexylamines/pharmacology , Cyclooxygenase 2/drug effects , Deferoxamine/pharmacology , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred ICR , NF-E2-Related Factor 2/antagonists & inhibitors , Oligospermia/pathology , Phenylenediamines/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Sperm Motility/drug effects , Testis/drug effects , Testis/pathology
17.
Transfusion ; 60(3): 513-523, 2020 03.
Article in English | MEDLINE | ID: mdl-32064619

ABSTRACT

BACKGROUND: Red blood cell (RBC) transfusions result in the sequestration and metabolism of storage-damaged RBCs within the spleen and liver. These events are followed by increased plasma iron concentrations that can contribute to oxidant stress and cellular injury. We hypothesized that administration of a ferroportin inhibitor (FPN-INH) immediately after acute RBC exchange transfusion could attenuate posttransfusion circulatory compartment iron exposure, by retaining iron in spleen and hepatic macrophages. STUDY DESIGN AND METHODS: Donor guinea pig blood was leukoreduced, and RBCs were preserved at 4°C. Recipient guinea pigs (n = 5/group) were exchange transfused with donor RBCs after refrigerator preservation and dosed intravenously with a small-molecule FPN-INH. Groups included transfusion with vehicle (saline), 5 mg/kg or 25 mg/kg FPN-INH. A time course of RBC morphology, plasma non-transferrin-bound iron (NTBI) and plasma hemoglobin (Hb) were evaluated. End-study spleen, liver, and kidney organ iron levels, as well as renal tissue oxidation and injury, were measured acutely (24-hr after transfusion). RESULTS: RBC transfusion increased plasma NTBI, with maximal concentrations occurring 8 hours after transfusion. Posttransfusion iron accumulation resulted in tubule oxidation and acute kidney injury. FPN inhibition increased spleen and liver parenchymal/macrophage iron accumulation, but attenuated plasma NTBI, and subsequent renal tissue oxidation/injury. CONCLUSION: In situations of acute RBC transfusion, minimizing circulatory NTBI exposure by FPN inhibition may attenuate organ-specific adverse consequences of iron exposure.


Subject(s)
Cation Transport Proteins/metabolism , Iron/blood , Animals , Blood Preservation , Cation Transport Proteins/antagonists & inhibitors , Erythrocyte Transfusion/methods , Guinea Pigs , Humans , Male , Oxidative Stress/physiology
18.
Exp Cell Res ; 386(2): 111740, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31756312

ABSTRACT

Advanced gastric cancer (GC) is aggressive with a high mortality rate. Rhesus family, C glycoprotein (RhCG) participates in tumor progression in many cancers, however its function in GC is still unknown. Here, we showed that RhCG was overexpressed in GC tissues at mRNA (P = 0.036) and protein levels (P < 0.05) compared with normal tissues. High RhCG level was correlated with poor differentiation (P = 0.037), TNM stage (P < 0.001), high HER-2 level (P = 0.018) and worse prognosis (P < 0.001). Cox proportional hazard model indicated that RhCG level was an independent prognostic biomarker. RhCG knockdown significantly decreased pHi and impeded tumor cellular proliferation, migration and invasion and repressed ß-catenin and c-myc expression in GC cells. Moreover, GC cells with high RhCG level had reduced oxaliplatin efficacy suggesting a role for RhCG as a therapeutic target for GC. Our findings revealed a function of RhCG in cancer pathogenesis, invasion and metastasis in human GC. We suggest that RhCG act may as a novel prognostic indicator and a therapeutic target for gastric adenocarcinoma.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Cation Transport Proteins/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Membrane Glycoproteins/genetics , Receptor, ErbB-2/genetics , Aged , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/mortality , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Hydrogen-Ion Concentration , Liver Neoplasms/diagnosis , Liver Neoplasms/drug therapy , Liver Neoplasms/mortality , Lymphatic Metastasis , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Middle Aged , Neoplasm Staging , Oxaliplatin/therapeutic use , Prognosis , Proportional Hazards Models , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , beta Catenin/genetics , beta Catenin/metabolism
19.
Am J Hematol ; 95(1): 68-77, 2020 01.
Article in English | MEDLINE | ID: mdl-31674058

ABSTRACT

Restriction of iron availability by ferroportin inhibition is a novel approach to treating non-transfusion-dependent thalassemia (ß-thalassemia intermedia). This first-in-human, Phase I study (https://www.clinicaltrialsregister.eu; EudraCT no. 2017-003395-31) assessed the safety, tolerability, pharmacokinetics and pharmacodynamics of single- and multiple-ascending doses (SAD and MAD) of the oral ferroportin inhibitor, VIT-2763, in healthy volunteers. Participants received VIT-2763 5/15/60/120/240 mg or placebo in the SAD phase and VIT-2763 60/120 mg once daily, VIT-2763 60/120 mg twice daily, or placebo for 7 days in the MAD phase. Seventy-two participants completed treatment. VIT-2763 was well tolerated and demonstrated a similar safety profile to the placebo. There were no serious or severe adverse events, or discontinuations due to adverse events. VIT-2763 absorption was relatively fast, with detectable levels 15 to 30 minutes post-dose. Following multiple dosing there was no apparent change in absorption and accumulation was minimal. Mean elimination half-life was 1.9 to 5.3 hours following single dosing, and 2.1 to 3.8 hours on Day 1 and 2.6 to 5.3 hours on Day 7, following repeated dosing. There was a temporary decrease in mean serum iron levels with VIT-2763 single doses ≥60 mg and all multiple doses; mean calculated transferrin saturation (only assessed following multiple dosing) also temporarily decreased. A shift in mean serum hepcidin peaks followed administration of all iron-lowering doses of VIT-2763. This effect was less pronounced after 7 days of multiple dosing (aside from with 120 mg once daily). These results support the initiation of clinical studies in patients with non-transfusion-dependent thalassemia and documented iron overload due to ineffective erythropoiesis.


Subject(s)
Benzimidazoles/therapeutic use , Cation Transport Proteins/antagonists & inhibitors , Oxazoles/therapeutic use , Pyridines/therapeutic use , Thalassemia/drug therapy , Administration, Oral , Benzimidazoles/pharmacology , Dose-Response Relationship, Drug , Drug Administration Schedule , Healthy Volunteers , Hepcidins/blood , Humans , Iron/blood , Iron Overload/drug therapy , Oxazoles/pharmacology , Pyridines/pharmacology
20.
J Clin Invest ; 130(1): 491-506, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31638596

ABSTRACT

ß-Thalassemia is a genetic anemia caused by partial or complete loss of ß-globin synthesis, leading to ineffective erythropoiesis and RBCs with a short life span. Currently, there is no efficacious oral medication modifying anemia for patients with ß-thalassemia. The inappropriately low levels of the iron regulatory hormone hepcidin enable excessive iron absorption by ferroportin, the unique cellular iron exporter in mammals, leading to organ iron overload and associated morbidities. Correction of unbalanced iron absorption and recycling by induction of hepcidin synthesis or treatment with hepcidin mimetics ameliorates ß-thalassemia. However, hepcidin modulation or replacement strategies currently in clinical development all require parenteral drug administration. We identified oral ferroportin inhibitors by screening a library of small molecular weight compounds for modulators of ferroportin internalization. Restricting iron availability by VIT-2763, the first clinical stage oral ferroportin inhibitor, ameliorated anemia and the dysregulated iron homeostasis in the Hbbth3/+ mouse model of ß-thalassemia intermedia. VIT-2763 not only improved erythropoiesis but also corrected the proportions of myeloid precursors in spleens of Hbbth3/+ mice. VIT-2763 is currently being developed as an oral drug targeting ferroportin for the treatment of ß-thalassemia.


Subject(s)
Cation Transport Proteins/antagonists & inhibitors , Ferric Compounds/administration & dosage , Maltose/analogs & derivatives , beta-Thalassemia/drug therapy , Administration, Oral , Animals , Cation Transport Proteins/metabolism , Cell Line , Disease Models, Animal , Dogs , Drug Evaluation, Preclinical , Erythropoiesis/drug effects , Erythropoiesis/genetics , Female , Hepcidins/metabolism , Humans , Iron/blood , Iron/metabolism , Madin Darby Canine Kidney Cells , Male , Maltose/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Proteolysis/drug effects , Rats, Sprague-Dawley , Ubiquitination/drug effects , beta-Globins/deficiency , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...