Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 726
Filter
Add more filters

Publication year range
1.
Genes Dev ; 32(1): 79-92, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29437726

ABSTRACT

Phosphorus is an essential element assimilated largely as orthophosphate (Pi). Cells respond to Pi starvation by importing Pi from their surroundings. We now report that impaired protein synthesis alone triggers a Pi starvation response even when Pi is plentiful in the extracellular milieu. In the bacterium Salmonella enterica serovar Typhimurium, this response entails phosphorylation of the regulatory protein PhoB and transcription of PhoB-dependent Pi transporter genes and is eliminated upon stimulation of adenosine triphosphate (ATP) hydrolysis. When protein synthesis is impaired due to low cytoplasmic magnesium (Mg2+), Salmonella triggers the Pi starvation response because ribosomes are destabilized, which reduces ATP consumption and thus free cytoplasmic Pi. This response is transient because low cytoplasmic Mg2+ promotes an uptake in Mg2+ and a decrease in ATP levels, which stabilizes ribosomes, resulting in ATP consumption and Pi increase, thus ending the response. Notably, pharmacological inhibition of protein synthesis also elicited a Pi starvation response in the bacterium Escherichia coli and the yeast Saccharomyces cerevisiae Our findings identify a regulatory connection between protein synthesis and Pi homeostasis that is widespread in nature.


Subject(s)
Bacterial Proteins/metabolism , Phosphates/metabolism , Protein Biosynthesis , Adenosine Triphosphatases/physiology , Bacterial Proteins/physiology , Cation Transport Proteins/physiology , Escherichia coli/drug effects , Escherichia coli/metabolism , Homeostasis , Magnesium/metabolism , Membrane Transport Proteins/physiology , Protein Synthesis Inhibitors/pharmacology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Transcription, Genetic
2.
PLoS Biol ; 19(12): e3001496, 2021 12.
Article in English | MEDLINE | ID: mdl-34928937

ABSTRACT

Magnesium is essential for cellular life, but how it is homeostatically controlled still remains poorly understood. Here, we report that members of CNNM family, which have been controversially implicated in both cellular Mg2+ influx and efflux, selectively bind to the TRPM7 channel to stimulate divalent cation entry into cells. Coexpression of CNNMs with the channel markedly increased uptake of divalent cations, which is prevented by an inactivating mutation to the channel's pore. Knockout (KO) of TRPM7 in cells or application of the TRPM7 channel inhibitor NS8593 also interfered with CNNM-stimulated divalent cation uptake. Conversely, KO of CNNM3 and CNNM4 in HEK-293 cells significantly reduced TRPM7-mediated divalent cation entry, without affecting TRPM7 protein expression or its cell surface levels. Furthermore, we found that cellular overexpression of phosphatases of regenerating liver (PRLs), known CNNMs binding partners, stimulated TRPM7-dependent divalent cation entry and that CNNMs were required for this activity. Whole-cell electrophysiological recordings demonstrated that deletion of CNNM3 and CNNM4 from HEK-293 cells interfered with heterologously expressed and native TRPM7 channel function. We conclude that CNNMs employ the TRPM7 channel to mediate divalent cation influx and that CNNMs also possess separate TRPM7-independent Mg2+ efflux activities that contribute to CNNMs' control of cellular Mg2+ homeostasis.


Subject(s)
Cation Transport Proteins/metabolism , Cyclins/metabolism , Protein Serine-Threonine Kinases/metabolism , TRPM Cation Channels/metabolism , Cation Transport Proteins/physiology , Cations, Divalent/metabolism , Cell Line, Tumor , Cyclins/physiology , HEK293 Cells , Humans , Magnesium/metabolism , Patch-Clamp Techniques , Protein Serine-Threonine Kinases/physiology , TRPM Cation Channels/genetics , TRPM Cation Channels/physiology
3.
Sheng Li Xue Bao ; 76(3): 487-495, 2024 Jun 25.
Article in Zh | MEDLINE | ID: mdl-38939942

ABSTRACT

Copper is a vital trace metal element necessary for the functioning of living organisms. It serves as a co-factor or structural component in numerous enzymes, participating in crucial biological metabolic processes. Disruptions in copper homeostasis, whether inherited or acquired, such as copper overload, deficiency, or uneven distribution, can contribute to or exacerbate various diseases, including Menkes disease, Wilson's disease, neurodegenerative disorders, anemia, cardiovascular diseases, kidney diseases and cancer. Recent research has highlighted the close correlation between chronic kidney disease and intracellular copper overload. Therefore, renal cells must establish a well-organized and efficient copper regulation network to maintain intracellular copper homeostasis. This review summarizes the processes of copper uptake, intracellular trafficking, storage, and excretion in renal cells, and elucidates the underlying mechanisms involved, aiming to provide a theoretical foundation and potential therapeutic targets for the fundamental investigation and clinical management of kidney-related diseases.


Subject(s)
Copper , Homeostasis , Kidney , Homeostasis/physiology , Humans , Copper/metabolism , Kidney/metabolism , Kidney/physiology , Animals , Cation Transport Proteins/metabolism , Cation Transport Proteins/physiology , Kidney Diseases/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/physiology , Copper-Transporting ATPases/metabolism , Copper-Transporting ATPases/genetics , Copper Transporter 1/metabolism
4.
Proc Natl Acad Sci U S A ; 117(35): 21731-21739, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32801213

ABSTRACT

Ca2+ uptake by mitochondria regulates bioenergetics, apoptosis, and Ca2+ signaling. The primary pathway for mitochondrial Ca2+ uptake is the mitochondrial calcium uniporter (MCU), a Ca2+-selective ion channel in the inner mitochondrial membrane. MCU-mediated Ca2+ uptake is driven by the sizable inner-membrane potential generated by the electron-transport chain. Despite the large thermodynamic driving force, mitochondrial Ca2+ uptake is tightly regulated to maintain low matrix [Ca2+] and prevent opening of the permeability transition pore and cell death, while meeting dynamic cellular energy demands. How this is accomplished is controversial. Here we define a regulatory mechanism of MCU-channel activity in which cytoplasmic Ca2+ regulation of intermembrane space-localized MICU1/2 is controlled by Ca2+-regulatory mechanisms localized across the membrane in the mitochondrial matrix. Ca2+ that permeates through the channel pore regulates Ca2+ affinities of coupled inhibitory and activating sensors in the matrix. Ca2+ binding to the inhibitory sensor within the MCU amino terminus closes the channel despite Ca2+ binding to MICU1/2. Conversely, disruption of the interaction of MICU1/2 with the MCU complex disables matrix Ca2+ regulation of channel activity. Our results demonstrate how Ca2+ influx into mitochondria is tuned by coupled Ca2+-regulatory mechanisms on both sides of the inner mitochondrial membrane.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Mitochondria/metabolism , Apoptosis , Biological Transport , Calcium/physiology , Calcium Channels/physiology , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/physiology , Cation Transport Proteins/metabolism , Cation Transport Proteins/physiology , Cytoplasm/metabolism , Cytosol/metabolism , HEK293 Cells , HeLa Cells , Humans , Membrane Potential, Mitochondrial/physiology , Mitochondria/physiology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/physiology , Oxidation-Reduction , Protein Multimerization , Signal Transduction
5.
Mol Microbiol ; 115(4): 554-573, 2021 04.
Article in English | MEDLINE | ID: mdl-33034093

ABSTRACT

S. aureus USA300 isolates utilize the copBL and copAZ gene products to prevent Cu intoxication. We created and examined a ΔcopAZ ΔcopBL mutant strain (cop-). The cop- strain was sensitive to Cu and accumulated intracellular Cu. We screened a transposon (Tn) mutant library in the cop- background and isolated strains with Tn insertions in the mntABC operon that permitted growth in the presence of Cu. The mutations were in mntA and they were recessive. Under the growth conditions utilized, MntABC functioned in manganese (Mn) import. When cultured with Cu, strains containing a mntA::Tn accumulated less Cu than the parent strain. Mn(II) supplementation improved growth when cop- was cultured with Cu and this phenotype was dependent upon the presence of MntR, which is a repressor of mntABC transcription. A ΔmntR strain had an increased Cu load and decreased growth in the presence of Cu, which was abrogated by the introduction of mntA::Tn. Over-expression of mntABC increased cellular Cu load and sensitivity to Cu. The presence of a mntA::Tn mutation protected iron-sulfur (FeS) enzymes from inactivation by Cu. The data presented are consistent with a model wherein defective MntABC results in decreased cellular Cu accumulation and protection to FeS enzymes from Cu poisoning.


Subject(s)
Cation Transport Proteins/physiology , Copper/metabolism , Copper/pharmacology , Gene Expression Regulation, Bacterial , Manganese/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/physiology , DNA, Bacterial , Humans , Iron/metabolism , Iron-Sulfur Proteins/metabolism , Membrane Transport Proteins/physiology , Mutagenesis, Insertional , Operon , RNA, Bacterial , Repressor Proteins/physiology , Staphylococcal Infections/microbiology
6.
Reprod Biol Endocrinol ; 20(1): 17, 2022 Jan 22.
Article in English | MEDLINE | ID: mdl-35065654

ABSTRACT

BACKGROUND: Overwhelming evidences suggest oxidative stress is a major cause of sperm dysfunction and male infertility. Zinc is an important non-enzymatic antioxidant with a wide range of biological functions and plays a significant role in preserving male fertility. Notably, zinc trafficking through the cellular and intracellular membrane is mediated by specific families of zinc transporters, i.e., SLC39s/ZIPs and SLC30s/ZnTs. However, their expression and function were rarely evaluated in the male germ cells. The aim of this study is to determine and characterize the crucial zinc transporter responsible for the maintenance of spermatogenesis. METHODS: The expression patterns of all 14 ZIP members were characterized in the mouse testis. qRT-PCR, immunoblot and immunohistochemistry analyses evaluated the ZIP12 gene and protein expression levels. The role of ZIP12 expression was evaluated in suppressing the sperm quality induced by exposure to an oxidative stress in a spermatogonia C18-4 cell line. Zip12 RNAi transfection was performed to determine if its downregulation altered cell viability and apoptosis in this cell line. An obese mouse model fed a high-fat-diet was employed to determine if there is a correlation between changes in the ZIP12 expression level and sperm quality. RESULTS: The ZIP12 mRNA and protein expression levels were higher than those of other ZIP family members in both the mouse testis and other tissues. Importantly, the ZIP12 expression levels were very significantly higher in both mice and human spermatogonia and spermatozoa. Moreover, the testicular ZIP12 expression levels significantly decreased in obese mice, which was associated with reduced sperm zinc content, excessive sperm ROS generation, poor sperm quality and male subfertility. Similarly, exposure to an oxidative stress induced significant declines in the ZIP12 expression level in C18-4 cells. Knockdown of ZIP12 expression mediated by transfection of a ZIP12 siRNA reduced both the zinc content and viability whereas apoptotic activity increased in the C18-4 cell line. CONCLUSIONS: The testicular zinc transporter ZIP12 expression levels especially in spermatogonia and spermatozoa are higher than in other tissues. ZIP12 may play a key role in maintaining intracellular zinc content at levels that reduce the inhibitory effects of rises in oxidative stress on spermatogonia and spermatozoa viability during spermatogenesis which help counteract declines in male fertility.


Subject(s)
Cation Transport Proteins/physiology , Spermatogonia/physiology , Zinc/metabolism , Animals , Cells, Cultured , Cytoprotection/genetics , Homeostasis/genetics , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/genetics , Spermatogenesis/genetics , Testis/metabolism
7.
J Pharmacol Sci ; 148(1): 125-133, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34924116

ABSTRACT

Manganese (Mn) is an essential trace element required for various biological processes. However, excess Mn causes serious side effects in humans, including parkinsonism. Thus, elucidation of Mn homeostasis at the systemic, cellular, and molecular levels is important. Many metal transporters and channels can be involved in the transport and homeostasis of Mn, and an increasing body of evidence shows that several zinc (Zn) transporters belonging to the ZIP and ZNT families, specifically, ZNT10, ZIP8, and ZIP14, play pivotal roles in Mn metabolism. Mutations in the genes encoding these transporter proteins are associated with congenital disorders related to dysregulated Mn homeostasis in humans. Moreover, single nucleotide polymorphisms of ZIP8 are associated with multiple clinical phenotypes. In this review, we discuss the recent literature on the structural and biochemical features of ZNT10, ZIP8, and ZIP14, including transport mechanisms, regulation of expression, and pathophysiological functions. Because a disturbance in Mn homeostasis is closely associated with a variety of phenotypes and risk of human diseases, these transporters constitute a significant target for drug development. An understanding of the roles of these key transporters in Mn metabolism should provide new insights into pharmacological applications of their inhibitors and enhancers in human diseases.


Subject(s)
Cation Transport Proteins/physiology , Manganese/metabolism , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Gene Expression Regulation , Homeostasis , Humans , Mammals , Manganese/adverse effects , Mutation , Parkinsonian Disorders/etiology , Phenotype
8.
J Pharmacol Sci ; 148(1): 14-18, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34924118

ABSTRACT

Cyclin M (CNNM) and its prokaryotic ortholog CorC belong to a family of proteins that function as Mg2+-extruding transporters by stimulating Na+/Mg2+ exchange, and thereby control intracellular Mg2+ levels. The Mg2+-extruding function of CNNM is inhibited by the direct binding of an oncogenic protein, phosphatase of regenerating liver (PRL), and this inhibition is responsible for the PRL-driven malignant progression of cancers. Studies with mouse strains deficient for the CNNM gene family revealed the importance of CNNM4 and CNNM2 in maintaining organismal Mg2+ homeostasis by participating in intestinal Mg2+ absorption and renal reabsorption, respectively. Moreover, CNNM proteins are involved in various diseases, and gene mutations in CNNM2 and CNNM4 cause dominant familial hypomagnesemia and Jalili syndrome, respectively. Genome wide association studies have also revealed the importance of CNNM2 in multiple major diseases, such as hypertension and schizophrenia. Collectively, the molecular and biological characterizations of CNNM/CorC show that they are an intriguing therapeutic target; the current status of drug development targeting these proteins is also discussed.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/physiology , Genome-Wide Association Study , Magnesium/metabolism , Molecular Targeted Therapy , Neoplasms/genetics , Amelogenesis Imperfecta/genetics , Amelogenesis Imperfecta/therapy , Animals , Cation Transport Proteins/metabolism , Cone-Rod Dystrophies/genetics , Cone-Rod Dystrophies/therapy , Homeostasis/genetics , Humans , Hypercalciuria/genetics , Hypercalciuria/therapy , Hypertension/genetics , Hypertension/therapy , Kidney/metabolism , Mice , Mutation , Neoplasms/therapy , Nephrocalcinosis/genetics , Nephrocalcinosis/therapy , Protein Binding , Protein Tyrosine Phosphatases/metabolism , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/therapy , Schizophrenia/genetics , Schizophrenia/therapy
9.
J Mol Cell Cardiol ; 152: 69-79, 2021 03.
Article in English | MEDLINE | ID: mdl-33307093

ABSTRACT

While Zn2+ dyshomeostasis is known to contribute to ischemia/reperfusion (I/R) injury, the roles of zinc transporters that are responsible for Zn2+ homeostasis in the pathogenesis of I/R injury remain to be addressed. This study reports that ZIP13 (SLC39A13), a zinc transporter, plays a role in myocardial I/R injury by modulating the Ca2+ signaling pathway rather than by regulating Zn2+ transport. ZIP13 is downregulated upon reperfusion in mouse hearts or in H9c2 cells at reoxygenation. Ca2+ but not Zn2+ was responsible for ZIP13 downregulation, implying that ZIP13 may play a role in I/R injury through the Ca2+ signaling pathway. In line with our assumption, knockout of ZIP13 resulted in phosphorylation (Thr287) of Ca2+-calmodulin-dependent protein kinase (CaMKII), indicating that downregulation of ZIP13 leads to CaMKII activation. Further studies showed that the heart-specific knockout of ZIP13 enhanced I/R-induced CaMKII phosphorylation in mouse hearts. In contrast, overexpression of ZIP13 suppressed I/R-induced CaMKII phosphorylation. Moreover, the heart-specific knockout of ZIP13 exacerbated myocardial infarction in mouse hearts subjected to I/R, whereas overexpression of ZIP13 reduced infarct size. In addition, knockout of ZIP13 induced increases of mitochondrial Ca2+, ROS, mitochondrial swelling, decrease in the mitochondrial respiration control rate (RCR), and dissipation of mitochondrial membrane potential (ΔΨm) in a CaMKII-dependent manner. These data suggest that downregulation of ZIP13 at reperfusion contributes to myocardial I/R injury through activation of CaMKII and the mitochondrial death pathway.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Cation Transport Proteins/physiology , Mitochondria, Heart/pathology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/metabolism , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Phosphorylation , Signal Transduction
10.
J Cell Mol Med ; 25(17): 8432-8441, 2021 09.
Article in English | MEDLINE | ID: mdl-34302427

ABSTRACT

High myopia is one of the leading causes of visual impairment worldwide with high heritability. We have previously identified the genetic contribution of SLC39A5 to nonsyndromic high myopia and demonstrated that disease-related mutations of SLC39A5 dysregulate the TGF-ß pathway. In this study, the mechanisms underlying SLC39A5 involvement in the pathogenesis of high myopia are determined. We observed the morphogenesis and migration abnormalities of the SLC39A5 knockout (KO) human embryonic kidney cells (HEK293) and found a significant injury of ECM constituents. RNA-seq and qRT-PCR revealed the transcription decrease in COL1A1, COL2A1, COL4A1, FN1 and LAMA1 in the KO cells. Further, we demonstrated that TGF-ß signalling, the regulator of ECM, was inhibited in SLC39A5 depletion situation, wherein the activation of receptor Smads (R-Smads) via phosphorylation was greatly blocked. SLC39A5 re-expression reversed the phenotype of TGF-ß signalling and ECM synthesis in the KO cells. The fact that TGF-ß signalling was zinc-regulated and that SLC39A5 was identified as a zinc transporter urged us to check the involvement of intracellular zinc in TGF-ß signalling impairment. Finally, we determined that insufficient zinc chelation destabilized Smad proteins, which naturally inhibited TGF-ß signalling. Overall, the SLC39A5 depletion-induced zinc deficiency destabilized Smad proteins, which inhibited the TGF-ß signalling and downstream ECM synthesis, thus contributing to the pathogenesis of high myopia. This discovery provides a deep insight into myopic development.


Subject(s)
Cation Transport Proteins/physiology , Extracellular Matrix/metabolism , Myopia/metabolism , Smad Proteins/metabolism , Zinc/metabolism , HEK293 Cells , Humans , Mutation
11.
J Biol Chem ; 295(32): 11002-11020, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32518166

ABSTRACT

Mitochondrial iron import is essential for iron-sulfur cluster formation and heme biosynthesis. Two nuclear-encoded vertebrate mitochondrial high-affinity iron importers, mitoferrin1 (Mfrn1) and Mfrn2, have been identified in mammals. In mice, the gene encoding Mfrn1, solute carrier family 25 member 37 (Slc25a37), is highly expressed in sites of erythropoiesis, and whole-body Slc25a37 deletion leads to lethality. Here, we report that mice with a deletion of Slc25a28 (encoding Mfrn2) are born at expected Mendelian ratios, but show decreased male fertility due to reduced sperm numbers and sperm motility. Mfrn2-/- mice placed on a low-iron diet exhibited reduced mitochondrial manganese, cobalt, and zinc levels, but not reduced iron. Hepatocyte-specific loss of Slc25a37 (encoding Mfrn1) in Mfrn2-/- mice did not affect animal viability, but resulted in a 40% reduction in mitochondrial iron and reduced levels of oxidative phosphorylation proteins. Placing animals on a low-iron diet exaggerated the reduction in mitochondrial iron observed in liver-specific Mfrn1/2-knockout animals. Mfrn1-/-/Mfrn2-/- bone marrow-derived macrophages or skin fibroblasts in vitro were unable to proliferate, and overexpression of Mfrn1-GFP or Mfrn2-GFP prevented this proliferation defect. Loss of both mitoferrins in hepatocytes dramatically reduced regeneration in the adult mouse liver, further supporting the notion that both mitoferrins transport iron and that their absence limits proliferative capacity of mammalian cells. We conclude that Mfrn1 and Mfrn2 contribute to mitochondrial iron homeostasis and are required for high-affinity iron import during active proliferation of mammalian cells.


Subject(s)
Cation Transport Proteins/physiology , Cell Proliferation/physiology , Liver Regeneration/physiology , Membrane Transport Proteins/physiology , Animals , Homeostasis , Iron/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria, Liver/metabolism
12.
Biochem Biophys Res Commun ; 560: 7-13, 2021 06 30.
Article in English | MEDLINE | ID: mdl-33964505

ABSTRACT

Zinc and iron are essential micronutrients for plant growth, and their homeostasis must be tightly regulated. Previously, it has been shown that Zinc-Induced Facilitator 1 (ZIF1) is involved in basal Zn tolerance by controlling the vacuolar storage of nicotianamine (NA). However, knowledge of the functional roles of two ZIF1 paralogs, ZIF-LIKE1 (ZIFL1) and ZIFL2, in metal homeostasis remains limited. Here, we functionally characterized the roles of ZIF1, ZIFL1, and ZIFL2 in Zn and Fe homeostasis. Expression of ZIF1 and ZIFL1 was induced by both excess Zn and Fe-deficiency, and their loss-of-function led to hypersensitivity under excess Zn and Fe-deficiency, suggesting functional overlap between ZIF1 and ZIFL1. By contrast, the disruption of ZIFL2 resulted in no obvious phenotypic alteration under both conditions. Additionally, the expression of ZIFL1, but not that of ZIFL2, in the zif1 mutant partially restored the phenotype under excess Zn, suggesting that ZIF1 and ZIFL1 perform functionally redundant roles in Zn homeostasis.


Subject(s)
Arabidopsis Proteins/physiology , Cation Transport Proteins/physiology , Iron/metabolism , Zinc/metabolism , Arabidopsis/drug effects , Arabidopsis/genetics , Arabidopsis/growth & development , Arabidopsis/metabolism , Arabidopsis Proteins/biosynthesis , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cation Transport Proteins/biosynthesis , Cation Transport Proteins/genetics , Homeostasis , Iron/physiology , Iron/toxicity , Mutation , Phenotype , Seedlings/metabolism , Stress, Physiological/genetics , Zinc/toxicity
13.
Nat Chem Biol ; 15(2): 179-188, 2019 02.
Article in English | MEDLINE | ID: mdl-30643281

ABSTRACT

The identification of activating mutations in NOTCH1 in 50% of T cell acute lymphoblastic leukemia has generated interest in elucidating how these mutations contribute to oncogenic transformation and in targeting the pathway. A phenotypic screen identified compounds that interfere with trafficking of Notch and induce apoptosis via an endoplasmic reticulum (ER) stress mechanism. Target identification approaches revealed a role for SLC39A7 (ZIP7), a zinc transport family member, in governing Notch trafficking and signaling. Generation and sequencing of a compound-resistant cell line identified a V430E mutation in ZIP7 that confers transferable resistance to the compound NVS-ZP7-4. NVS-ZP7-4 altered zinc in the ER, and an analog of the compound photoaffinity labeled ZIP7 in cells, suggesting a direct interaction between the compound and ZIP7. NVS-ZP7-4 is the first reported chemical tool to probe the impact of modulating ER zinc levels and investigate ZIP7 as a novel druggable node in the Notch pathway.


Subject(s)
Cation Transport Proteins/genetics , Endoplasmic Reticulum Stress/physiology , Receptor, Notch1/genetics , Animals , Apoptosis , Carrier Proteins/metabolism , Cation Transport Proteins/metabolism , Cation Transport Proteins/physiology , Cell Line , Cell Transformation, Neoplastic , Endoplasmic Reticulum/physiology , Humans , Mutation , Protein Transport , Receptor, Notch1/physiology , Signal Transduction , Zinc/metabolism
14.
PLoS Biol ; 16(6): e2006204, 2018 06.
Article in English | MEDLINE | ID: mdl-29879100

ABSTRACT

Zinc is an essential mineral, but our understanding of its uses in the body is limited. Capitalizing on approaches available in the model system Caenorhabditis elegans, Zhao and colleagues show that zinc transduces a signal that induces sperm to become motile. This is an enigmatic process because sperm in all sexually-reproducing animals are transcriptionally inactive. Zinc levels inside sperm are regulated by an evolutionarily conserved zinc transporter called Zrt- and Irt-like Protein Transporter 7.1 (ZIPT-7.1). This zinc transporter localizes to intracellular organelles, suggesting that it primarily controls zinc levels by releasing zinc into the cytoplasm from internal stores rather than importing it from the external environment. The zinc released within cells acts as a messenger in a signaling pathway to promote mobility acquisition. These studies reveal an important role for zinc as an intracellular second messenger that generates physiological changes vital for sperm motility and fertility.


Subject(s)
Spermatozoa/physiology , Zinc/physiology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Carrier Proteins/genetics , Carrier Proteins/physiology , Cation Transport Proteins/genetics , Cation Transport Proteins/physiology , Humans , Male , Models, Biological , Signal Transduction , Sperm Motility/genetics , Sperm Motility/physiology , Spermatogenesis/genetics , Spermatogenesis/physiology
15.
PLoS Biol ; 16(10): e2006024, 2018 10.
Article in English | MEDLINE | ID: mdl-30356235

ABSTRACT

In plants, nutrient provision of shoots depends on the uptake and transport of nutrients across the root tissue to the vascular system. Nutrient delivery to the vasculature is mediated via the apoplastic transport pathway (ATP), which uses the free space in the cell walls and is controlled by apoplastic barriers and nutrient transporters at the endodermis, or via the symplastic transport pathway (STP). However, the relative importance of these transport routes remains elusive. Here, we show that the STP, mediated by the epidermal ammonium transporter 1;3 (AMT1;3), dominates the radial movement of ammonium across the root tissue when external ammonium is low, whereas apoplastic transport controlled by AMT1;2 at the endodermis prevails at high external ammonium. Then, AMT1;2 favors nitrogen (N) allocation to the shoot, revealing a major importance of the ATP for nutrient partitioning to shoots. When an endodermal bypass was introduced by abolishing Casparian strip (CS) formation, apoplastic ammonium transport decreased. By contrast, symplastic transport was increased, indicating synergism between the STP and the endodermal bypass. We further establish that the formation of apoplastic barriers alters the cell type-specific localization of AMTs and determines STP and ATP contributions. These results show how radial transport pathways vary along the longitudinal gradient of the root axis and contribute to nutrient partitioning between roots and shoots.


Subject(s)
Ammonium Compounds/metabolism , Cation Transport Proteins/physiology , Plant Proteins/physiology , Plant Roots/metabolism , Arabidopsis/physiology , Arabidopsis Proteins/physiology , Biological Transport/physiology , Cation Transport Proteins/metabolism , Cell Wall , Gene Expression Regulation, Plant/genetics , Ion Transport/physiology , Membrane Transport Proteins/physiology , Nitrogen/metabolism , Plant Proteins/metabolism , Plant Roots/physiology
16.
Am J Hematol ; 96(6): 659-670, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33684239

ABSTRACT

The demand for iron is high in pregnancy to meet the increased requirements for erythropoiesis. Even pregnant females with initially iron-replete stores develop iron-deficiency anemia, due to inadequate iron absorption. In anemic females, the maternal iron supply is dedicated to maintaining iron metabolism in the fetus and placenta. Here, using a mouse model of iron deficiency in pregnancy, we show that iron recycled from senescent erythrocytes becomes a predominant source of this microelement that can be transferred to the placenta in females with depleted iron stores. Ferroportin is a key protein in the molecular machinery of cellular iron egress. We demonstrate that under iron deficiency in pregnancy, levels of ferroportin are greatly reduced in the duodenum, placenta and fetal liver, but not in maternal liver macrophages and in the spleen. Although low expression of both maternal and fetal hepcidin predicted ferroportin up-regulation in examined locations, its final expression level was very likely correlated with tissue iron status. Our results argue that iron released into the circulation of anemic females is taken up by the placenta, as evidenced by high expression of iron importers on syncytiotrophoblasts. Then, a substantial decrease in levels of ferroportin on the basolateral side of syncytiotrophoblasts, may be responsible for the reduced transfer of iron to the fetus. As attested by the lowest decrease in iron content among analyzed tissues, some part is retained in the placenta. These findings confirm the key role played by ferroportin in tuning iron turnover in iron-deficient pregnant mouse females and their fetuses.


Subject(s)
Cation Transport Proteins/physiology , Iron Deficiencies , Iron, Dietary/administration & dosage , Liver/metabolism , Pregnancy Complications/metabolism , Spleen/metabolism , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cation Transport Proteins/biosynthesis , Cation Transport Proteins/genetics , Cytokines/blood , Duodenum/metabolism , Erythrocyte Aging , Erythrocyte Indices , Female , Fetus/metabolism , Hemoglobins/metabolism , Hepcidins/biosynthesis , Hepcidins/genetics , Iron/metabolism , Liver/embryology , Macrophages/metabolism , Maternal-Fetal Exchange , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, 129 Strain , Muscle Proteins/blood , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Organ Specificity , Phagocytosis , Placenta/metabolism , Pregnancy , Up-Regulation
17.
Proc Natl Acad Sci U S A ; 115(52): E12443-E12452, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30530653

ABSTRACT

Stressors such as soil salinity and dehydration are major constraints on plant growth, causing worldwide crop losses. Compounding these insults, increasing climate volatility requires adaptation to fluctuating conditions. Salinity stress responses are relatively well understood in Arabidopsis thaliana, making this system suited for the rapid molecular dissection of evolutionary mechanisms. In a large-scale genomic analysis of Catalonian A. thaliana, we resequenced 77 individuals from multiple salinity gradients along the coast and integrated these data with 1,135 worldwide A. thaliana genomes for a detailed understanding of the demographic and evolutionary dynamics of naturally evolved salinity tolerance. This revealed that Catalonian varieties adapted to highly fluctuating soil salinity are not Iberian relicts but instead have immigrated to this region more recently. De novo genome assembly of three allelic variants of the high-affinity K+ transporter (HKT1;1) locus resolved structural variation between functionally distinct alleles undergoing fluctuating selection in response to seasonal changes in soil salinity. Plants harboring alleles responsible for low root expression of HKT1;1 and consequently high leaf sodium (HKT1;1HLS ) were migrants that have moved specifically into areas where soil sodium levels fluctuate widely due to geography and rainfall variation. We demonstrate that the proportion of plants harboring HKT1;1HLS alleles correlates with soil sodium level over time, HKT1;1HLS -harboring plants are better adapted to intermediate levels of salinity, and the HKT1;1HLS allele clusters with high-sodium accumulator accessions worldwide. Together, our evidence suggests that HKT1;1 is under fluctuating selection in response to climate volatility and is a worldwide determinant in adaptation to saline conditions.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Symporters/genetics , Symporters/metabolism , Adaptation, Biological/genetics , Adaptation, Physiological/genetics , Alleles , Arabidopsis/genetics , Arabidopsis/physiology , Arabidopsis Proteins/physiology , Cation Transport Proteins/physiology , Gene Frequency/genetics , Ion Transport , Salinity , Salt Tolerance , Sodium/metabolism , Sodium Chloride , Soil , Symporters/physiology
18.
Physiol Rev ; 93(4): 1721-41, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24137020

ABSTRACT

The iron hormone hepcidin and its receptor and cellular iron exporter ferroportin control the major fluxes of iron into blood plasma: intestinal iron absorption, the delivery of recycled iron from macrophages, and the release of stored iron from hepatocytes. Because iron losses are comparatively very small, iron absorption and its regulation by hepcidin and ferroportin determine total body iron content. Hepcidin is in turn feedback-regulated by plasma iron concentration and iron stores, and negatively regulated by the activity of erythrocyte precursors, the dominant consumers of iron. Hepcidin and ferroportin also play a role in host defense and inflammation, and hepcidin synthesis is induced by inflammatory signals including interleukin-6 and activin B. This review summarizes and discusses recent progress in molecular characterization of systemic iron homeostasis and its disorders, and identifies areas for further investigation.


Subject(s)
Hemostasis/physiology , Hepcidins/physiology , Iron/metabolism , Amino Acid Sequence , Cation Transport Proteins/analysis , Cation Transport Proteins/chemistry , Cation Transport Proteins/physiology , Hepatocytes/metabolism , Hepcidins/analysis , Hepcidins/chemistry , Humans , Macrophages/metabolism , Molecular Sequence Data
19.
Planta ; 252(3): 35, 2020 Aug 07.
Article in English | MEDLINE | ID: mdl-32767128

ABSTRACT

MAIN CONCLUSION: The nuclear-localized CAX-interacting protein VvCXIP4 is exported to the cytosol after a Ca2+ pulse, to activate the tonoplast-localized Ca2+/H+ exchanger VvCAX3. Vacuolar cation/H+ exchangers (CAXs) have long been recognized as 'housekeeping' components in cellular Ca2+ and trace metal homeostasis, being involved in a range of key cellular and physiological processes. However, the mechanisms that drive functional activation of the transporters are largely unknown. In the present study, we investigated the function of a putative grapevine CAX-interacting protein, VvCXIP4, by testing its ability to activate VvCAX3, previously characterized as a tonoplast-localized Ca2+/H+ exchanger. VvCAX3 contains an autoinhibitory domain that drives inactivation of the transporter and thus, is incapable of suppressing the Ca2+-hypersensitive phenotype of the S. cerevisiae mutant K667. In this study, the co-expression of VvCXIP4 and VvCAX3 in this strain efficiently rescued its growth defect at high Ca2+ levels. Flow cytometry experiments showed that yeast harboring both proteins effectively accumulated higher Ca2+ levels than cells expressing each of the proteins separately. Bimolecular fluorescence complementation (BiFC) assays allowed visualization of the direct interaction between the proteins in tobacco plants and in yeast, and also showed the self-interaction of VvCAX3 but not of VvCXIP4. Subcellular localization studies showed that, despite being primarily localized to the nucleus, VvCXIP4 is able to move to other cell compartments upon a Ca2+ stimulus, becoming prone to interaction with the tonoplast-localized VvCAX3. qPCR analysis showed that both genes are more expressed in grapevine stems and leaves, followed by the roots, and that the steady-state transcript levels were higher in the pulp than in the skin of grape berries. Also, both VvCXIP4 and VvCAX3 were upregulated by Ca2+ and Na+, indicating they share common regulatory mechanisms. However, VvCXIP4 was also upregulated by Li+, Cu2+ and Mn2+, and its expression increased steadily throughout grape berry development, contrary to VvCAX3, suggesting additional physiological roles for VvCXIP4, including the regulation of VvCAXs not yet functionally characterized. The main novelty of the present study was the demonstration of physical interaction between CXIP and CAX proteins from a woody plant model by BiFC assays, demonstrating the intracellular mobilization of CXIPs in response to Ca2+.


Subject(s)
Biological Transport/physiology , Cation Transport Proteins/genetics , Cation Transport Proteins/physiology , Cell Nucleus/physiology , Cytosol/physiology , Vitis/genetics , Vitis/physiology , Fruit/physiology , Gene Expression Regulation, Plant , Plant Leaves/physiology , Plant Proteins/physiology
20.
Plant Cell ; 29(2): 409-422, 2017 02.
Article in English | MEDLINE | ID: mdl-28188265

ABSTRACT

Ion transport in plants is not only strictly regulated on a transcriptional level, but it is also regulated posttranslationally. Enzyme modifications such as phosphorylation provide rapid regulation of many plant ion transporters and channels. Upon exposure to high ammonium concentrations in the rhizosphere, the high-affinity ammonium transporters (AMTs) in Arabidopsis thaliana are efficiently inactivated by phosphorylation to avoid toxic accumulation of cytoplasmic ammonium. External ammonium stimulates the phosphorylation of a conserved threonine in the cytosolic AMT1 C terminus, which allosterically inactivates AMT1 trimers. Using a genetic screen, we found that CALCINEURIN B-LIKE INTERACTING PROTEIN KINASE23 (CIPK23), a kinase that also regulates the most abundant NO3- transporter, NPF6;3, and activates the K+ channel AKT1, inhibits ammonium transport and modulates growth sensitivity to ammonium. Loss of CIPK23 increased root NH4+ uptake after ammonium shock and conferred hypersensitivity to ammonium and to the transport analog methylammonium. CIPK23 interacts with AMT1;1 and AMT1;2 in yeast, oocytes, and in planta. Inactivation of AMT1;2 by direct interaction with CIPK23 requires kinase activity and the calcineurin B-like binding protein CBL1. Since K+, NO3-, and NH4+ are major ions taken up by plants, CIPK23 appears to occupy a key position in controlling ion balance and ion homeostasis in the plant cell.


Subject(s)
Ammonium Compounds/metabolism , Arabidopsis Proteins/physiology , Arabidopsis/metabolism , Protein Serine-Threonine Kinases/physiology , Animals , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Biological Transport , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cation Transport Proteins/physiology , Gene Expression Regulation, Plant , Mutagenesis, Insertional , Mutation , Phenotype , Phosphorylation , Plant Proteins/genetics , Plant Proteins/metabolism , Plant Proteins/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL