Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 726
Filtrer
1.
Sheng Li Xue Bao ; 76(3): 487-495, 2024 Jun 25.
Article de Chinois | MEDLINE | ID: mdl-38939942

RÉSUMÉ

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.


Sujet(s)
Cuivre , Homéostasie , Rein , Homéostasie/physiologie , Humains , Cuivre/métabolisme , Rein/métabolisme , Rein/physiologie , Animaux , Transporteurs de cations/métabolisme , Transporteurs de cations/physiologie , Maladies du rein/métabolisme , Adenosine triphosphatases/métabolisme , Adenosine triphosphatases/physiologie , Copper-transporting ATPases/métabolisme , Copper-transporting ATPases/génétique , Transporteur-1 du cuivre/métabolisme
2.
Nat Commun ; 14(1): 516, 2023 01 31.
Article de Anglais | MEDLINE | ID: mdl-36720859

RÉSUMÉ

High-voltage-activated R-type CaV2.3 channel plays pivotal roles in many physiological activities and is implicated in epilepsy, convulsions, and other neurodevelopmental impairments. Here, we determine the high-resolution cryo-electron microscopy (cryo-EM) structure of human CaV2.3 in complex with the α2δ1 and ß1 subunits. The VSDII is stabilized in the resting state. Electrophysiological experiments elucidate that the VSDII is not required for channel activation, whereas the other VSDs are essential for channel opening. The intracellular gate is blocked by the W-helix. A pre-W-helix adjacent to the W-helix can significantly regulate closed-state inactivation (CSI) by modulating the association and dissociation of the W-helix with the gate. Electrostatic interactions formed between the negatively charged domain on S6II, which is exclusively conserved in the CaV2 family, and nearby regions at the alpha-interacting domain (AID) and S4-S5II helix are identified. Further functional analyses indicate that these interactions are critical for the open-state inactivation (OSI) of CaV2 channels.


Sujet(s)
Canaux calciques de type R , Transporteurs de cations , Humains , Cryomicroscopie électronique , Canaux calciques de type R/physiologie , Transporteurs de cations/physiologie
3.
Reprod Biol Endocrinol ; 20(1): 17, 2022 Jan 22.
Article de Anglais | MEDLINE | ID: mdl-35065654

RÉSUMÉ

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.


Sujet(s)
Transporteurs de cations/physiologie , Spermatogonies/physiologie , Zinc/métabolisme , Animaux , Cellules cultivées , Cytoprotection/génétique , Homéostasie/génétique , Infertilité masculine/génétique , Infertilité masculine/métabolisme , Mâle , Souris , Souris de lignée C57BL , Stress oxydatif/génétique , Spermatogenèse/génétique , Testicule/métabolisme
4.
J Pharmacol Sci ; 148(1): 125-133, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-34924116

RÉSUMÉ

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.


Sujet(s)
Transporteurs de cations/physiologie , Manganèse/métabolisme , Animaux , Transporteurs de cations/génétique , Transporteurs de cations/métabolisme , Régulation de l'expression des gènes , Homéostasie , Humains , Mammifères , Manganèse/effets indésirables , Mutation , Syndromes parkinsoniens/étiologie , Phénotype
5.
J Pharmacol Sci ; 148(1): 14-18, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-34924118

RÉSUMÉ

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.


Sujet(s)
Transporteurs de cations/génétique , Transporteurs de cations/physiologie , Étude d'association pangénomique , Magnésium/métabolisme , Thérapie moléculaire ciblée , Tumeurs/génétique , Amélogenèse imparfaite/génétique , Amélogenèse imparfaite/thérapie , Animaux , Transporteurs de cations/métabolisme , Dystrophies des cônes et des batonnets/génétique , Dystrophies des cônes et des batonnets/thérapie , Homéostasie/génétique , Humains , Hypercalciurie/génétique , Hypercalciurie/thérapie , Hypertension artérielle/génétique , Hypertension artérielle/thérapie , Rein/métabolisme , Souris , Mutation , Tumeurs/thérapie , Néphrocalcinose/génétique , Néphrocalcinose/thérapie , Liaison aux protéines , Protein Tyrosine Phosphatases/métabolisme , Erreurs innées du transport tubulaire rénal/génétique , Erreurs innées du transport tubulaire rénal/thérapie , Schizophrénie/génétique , Schizophrénie/thérapie
6.
PLoS Biol ; 19(12): e3001496, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34928937

RÉSUMÉ

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.


Sujet(s)
Transporteurs de cations/métabolisme , Cyclines/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Canaux cationiques TRPM/métabolisme , Transporteurs de cations/physiologie , Cations divalents/métabolisme , Lignée cellulaire tumorale , Cyclines/physiologie , Cellules HEK293 , Humains , Magnésium/métabolisme , Techniques de patch-clamp , Protein-Serine-Threonine Kinases/physiologie , Canaux cationiques TRPM/génétique , Canaux cationiques TRPM/physiologie
7.
J Genet ; 1002021.
Article de Anglais | MEDLINE | ID: mdl-34825662

RÉSUMÉ

Arabis paniculata has been reported as a hyperaccumulator and functions in cadmium (Cd) tolerance and accumulation. However, the genes involved in Cd stress resistance in A. paniculata are still unknown. In this work, genes of the natural resistanceassociated macrophage proteins (NRAMPs) were characterized in A. paniculata, and their evolutionary relationship and expression patterns were analysed. Expression profiles indicated that ApNRAMPs showed large differences in response to Cd stress. It was highly induced by Cd in root and shoot tissues. To investigate the function of ApNRAMP4 under Cd stress, ApNRAMP4 was cloned and expressed in yeast and Arabidopsis. The results indicated that yeast and Arabidopsis expressing ApNRAMP4 showed normal growth under Cd stress. In addition, transgenic yeast and Arabidopsis showed the ability to concentrate Cd. Under 20 µM CdCl2, Cd concentrations in wild type (WT) and transgenic yeast were 3.11 and 5.92 mg/kg, respectively. Cd concentrations in root tissues of WTand transgenic Arabidopsis were 0.18 and 0.54 mg/kg, respectively. In shoot tissues of WT and transgenic Arabidopsis, Cd concentrations were 0.13 and 0.49 mg/kg, respectively. This report provides genomic information on hyperaccumulator A. paniculata. In addition, the present work identified key NRAMP genes that may serve as resources for heavy metal phytoremediation.


Sujet(s)
Protéines d'Arabidopsis/génétique , Arabidopsis/génétique , Arabis/effets des médicaments et des substances chimiques , Arabis/génétique , Cadmium/toxicité , Transporteurs de cations/génétique , Protéines d'Arabidopsis/physiologie , Arabis/métabolisme , Cadmium/métabolisme , Transporteurs de cations/physiologie , Évolution moléculaire , Végétaux génétiquement modifiés/effets des médicaments et des substances chimiques , Végétaux génétiquement modifiés/génétique , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/physiologie , Transcriptome
8.
Gene ; 799: 145824, 2021 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-34252531

RÉSUMÉ

The SLC39A12 gene encodes the zinc transporter protein ZIP12, which is expressed across many tissues and is highly abundant in the vertebrate nervous system. As a zinc transporter, ZIP12 functions to transport zinc across cellular membranes, including cellular zinc influx across the plasma membrane. Genome-wide association and exome sequencing studies have shown that brain susceptibility-weighted magnetic resonance imaging (MRI) intensity is associated with ZIP12 polymorphisms and rare mutations. ZIP12 is required for neural tube closure and embryonic development in Xenopus tropicalis. Frog embryos depleted of ZIP12 by antisense morpholinos develop an anterior neural tube defect and lack viability. ZIP12 is also necessary for neurite outgrowth and mitochondrial function in mouse neural cells. ZIP12 mRNA is increased in brain regions of schizophrenic patients. Outside of the nervous system, hypoxia induces ZIP12 expression in multiple mammalian species, including humans, which leads to endothelial and smooth muscle thickening in the lung and contributes towards pulmonary hypertension. Other studies have associated ZIP12 with other diseases such as cancer. Given that ZIP12 is highly expressed in the brain and that susceptibility-weighted MRI is associated with brain metal content, ZIP12 may affect neurological diseases and psychiatric illnesses such as Parkinson's disease, Alzheimer's disease, and schizophrenia. Furthermore, the induction of ZIP12 and resultant zinc uptake under pathophysiological conditions may be a critical component of disease pathology, such as in pulmonary hypertension. Drug compounds that bind metals like zinc may be able to treat diseases associated with impaired zinc homeostasis and altered ZIP12 function.


Sujet(s)
Transporteurs de cations/physiologie , Phénomènes physiologiques du système nerveux , Protéines de Xénope/physiologie , Zinc/métabolisme , Animaux , Trouble autistique/métabolisme , Biobanques , Régulation de l'expression des gènes au cours du développement , Humains , Poumon/physiopathologie , Famille multigénique , Maladies neurodégénératives/étiologie , Stress oxydatif/physiologie , Royaume-Uni , Vertébrés/génétique
9.
J Cell Mol Med ; 25(17): 8432-8441, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34302427

RÉSUMÉ

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.


Sujet(s)
Transporteurs de cations/physiologie , Matrice extracellulaire/métabolisme , Myopie/métabolisme , Protéines Smad/métabolisme , Zinc/métabolisme , Cellules HEK293 , Humains , Mutation
10.
Biochem Biophys Res Commun ; 560: 7-13, 2021 06 30.
Article de Anglais | MEDLINE | ID: mdl-33964505

RÉSUMÉ

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.


Sujet(s)
Protéines d'Arabidopsis/physiologie , Transporteurs de cations/physiologie , Fer/métabolisme , Zinc/métabolisme , Arabidopsis/effets des médicaments et des substances chimiques , Arabidopsis/génétique , Arabidopsis/croissance et développement , Arabidopsis/métabolisme , Protéines d'Arabidopsis/biosynthèse , Protéines d'Arabidopsis/génétique , Protéines d'Arabidopsis/métabolisme , Transporteurs de cations/biosynthèse , Transporteurs de cations/génétique , Homéostasie , Fer/physiologie , Fer/toxicité , Mutation , Phénotype , Plant/métabolisme , Stress physiologique/génétique , Zinc/toxicité
11.
Yakugaku Zasshi ; 141(5): 695-703, 2021.
Article de Japonais | MEDLINE | ID: mdl-33952754

RÉSUMÉ

Cellular transport systems for both essential and toxic trace elements remain elusive. In our studies on the transport systems for cadmium (Cd), we found that the cellular uptake of Cd is mediated by the transporter for manganese (Mn). We identified ZIP8 and ZIP14, members of the ZIP zinc (Zn) transporter family, as transporters having high affinities for both Cd and Mn. Notably, the uptake of Cd into rice root from soil is mediated by a transporter for Mn as well. We found that ZIP8 is highly expressed at the S3 segment of the kidney proximal tubule and can transport glomerulus-filtered Cd and Mn ions in the lumen into epithelial cells of the proximal tubule, suggesting that ZIP8 has an important role in the renal reabsorption of both toxic Cd and essential Mn. Mutations in ZIP8 and ZIP14 genes were found in humans having congenital disorders associated with the disturbed transport of Mn, although ZIP8 mutation causes whole-body Mn deficiency while ZIP14 mutation causes Mn accumulation in the brain. Mutations in ZnT10, a Zn transporter responsible for Mn excretion, also cause hyperaccumulation of Mn in the brain. Results of genome-wide association studies have indicated that ZIP8 SNPs are involved in a variety of common diseases. Thus, ZIP8, ZIP14, and ZnT10 play crucial roles in the transport of Mn and thereby control Mn- and Cd-related biological events in the body.


Sujet(s)
Cadmium/métabolisme , Cadmium/toxicité , Protéines de transport/physiologie , Manganèse/métabolisme , Manganèse/toxicité , Animaux , Encéphale/métabolisme , Protéines de transport/métabolisme , Transporteurs de cations/génétique , Transporteurs de cations/métabolisme , Transporteurs de cations/physiologie , Étude d'association pangénomique , Humains , Tubules contournés proximaux/métabolisme , Mutation , Oryza/métabolisme , Racines de plante/métabolisme , Réabsorption rénale
12.
SLAS Discov ; 26(6): 798-810, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-33825579

RÉSUMÉ

Membrane proteins are involved in different physiological functions and are the target of pharmaceutical and abuse drugs. Xenopus laevis oocytes provide a powerful heterologous expression system for functional studies of these proteins. Typical experiments investigate transport using electrophysiology and radiolabeled uptake. A two-electrode voltage clamp is suitable only for electrogenic proteins, and uptake measurements require the existence of radiolabeled substrates and adequate laboratory facilities.Recently, Dictyostelium discoideum Nramp1 and NrampB were characterized using multidisciplinary approaches. NrampB showed no measurable electrogenic activity, and it was investigated in Xenopus oocytes by acquiring confocal images of the quenching of injected fluorophore calcein.This method is adequate to measure the variation in emitted fluorescence, and thus transporter activity indirectly, but requires long experimental procedures to collect statistically consistent data. Considering that optimal expression of heterologous proteins lasts for 48-72 h, a slow acquiring process requires the use of more than one batch of oocytes to complete the experiments. Here, a novel approach to measure substrate uptake is reported. Upon injection of a fluorophore, oocytes were incubated with the substrate and the transport activity measured, evaluating fluorescence quenching in a microplate reader. The technique permits the testing of tens of oocytes in different experimental conditions simultaneously, and thus the collection of significant statistical data for each batch, saving time and animals.The method was tested with different metal transporters (SLC11), DMT1, DdNramp1, and DdNrampB, and verified with the peptide transporter PepT1 (SLC15). Comparison with traditional methods (uptake, two-electrode voltage clamp) and with quenching images acquired by fluorescence microscopy confirmed its efficacy.


Sujet(s)
Phénomènes électrophysiologiques , Protéines de transport membranaire/métabolisme , Techniques de patch-clamp/méthodes , Animaux , Transport biologique , Transporteurs de cations/métabolisme , Transporteurs de cations/physiologie , Dictyostelium/métabolisme , Femelle , Fluorescéines/pharmacocinétique , Colorants fluorescents/pharmacocinétique , Potentiels de membrane , Microscopie de fluorescence , Ovocytes/composition chimique , Ovocytes/métabolisme , Xenopus laevis
13.
Am J Hematol ; 96(6): 659-670, 2021 06 01.
Article de Anglais | MEDLINE | ID: mdl-33684239

RÉSUMÉ

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.


Sujet(s)
Transporteurs de cations/physiologie , Carences en fer , Fer alimentaire/administration et posologie , Foie/métabolisme , Complications de la grossesse/métabolisme , Rate/métabolisme , Animaux , Protéines de transport/biosynthèse , Protéines de transport/génétique , Transporteurs de cations/biosynthèse , Transporteurs de cations/génétique , Cytokines/sang , Duodénum/métabolisme , Vieillissement érythrocytaire , Index érythrocytaires , Femelle , Foetus/métabolisme , Hémoglobines/métabolisme , Hepcidines/biosynthèse , Hepcidines/génétique , Fer/métabolisme , Foie/embryologie , Macrophages/métabolisme , Échange foetomaternel , Protéines membranaires/biosynthèse , Protéines membranaires/génétique , Souris , Souris de souche-129 , Protéines du muscle/sang , Protéines de tissu nerveux/biosynthèse , Protéines de tissu nerveux/génétique , Spécificité d'organe , Phagocytose , Placenta/métabolisme , Grossesse , Régulation positive
14.
J Orthop Surg Res ; 16(1): 170, 2021 Mar 03.
Article de Anglais | MEDLINE | ID: mdl-33658057

RÉSUMÉ

BACKGROUND: The dysregulation of circular RNAs (circRNAs) has been identified in various human diseases, including osteoarthritis (OA). The purpose of this study was to identify the role and mechanism of circ_SLC39A8 in regulating the progression of OA. METHODS: The expression levels of circ_SLC39A8, miR-591, and its potential target gene, interleukin-1-receptor-associated kinase 3 (IRAK3), were identified by quantitative real-time polymerase chain reaction (qRT-PCR). Cell viability and apoptosis were determined by Cell Counting Kit-8 (CCK-8) assay and flow cytometry, respectively. The relationship between miR-591 and circ_SLC39A8 or IRAK3 was predicted by bioinformatics tools and verified by dual-luciferase reporter. RESULTS: Circ_SLC39A8 and IRAK3 were upregulated and miR-591 was downregulated in OA cartilage tissues. Knockdown of circ_SLC39A8 inhibited apoptosis and inflammation in OA chondrocytes, while these effects were reversed by downregulating miR-591. Promotion cell viability effects of miR-591 were partially reversed by IRAK3 overexpression. CONCLUSION: Our findings indicated that knockdown of circ_SLC39A8 delayed the progression of OA via modulating the miR-591-IRAK3 axis, providing new insight into the molecular mechanisms of OA pathogenesis.


Sujet(s)
Transporteurs de cations/génétique , Transporteurs de cations/physiologie , Chondrocytes/métabolisme , Techniques de knock-down de gènes , Interleukin-1 Receptor-Associated Kinases/métabolisme , microARN/métabolisme , Arthrose/génétique , Sujet âgé , Apoptose/génétique , Survie cellulaire/génétique , Cellules cultivées , Chondrocytes/physiologie , Évolution de la maladie , Régulation négative/génétique , Femelle , Humains , Interleukin-1 Receptor-Associated Kinases/génétique , Mâle , microARN/génétique , Adulte d'âge moyen , Arthrose/métabolisme
15.
J Mol Cell Cardiol ; 152: 69-79, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33307093

RÉSUMÉ

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.


Sujet(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/métabolisme , Calcium/métabolisme , Transporteurs de cations/physiologie , Mitochondries du myocarde/anatomopathologie , Lésion de reperfusion myocardique/anatomopathologie , Myocytes cardiaques/anatomopathologie , Animaux , Calcium-Calmodulin-Dependent Protein Kinase Type 2/génétique , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Mitochondries du myocarde/métabolisme , Lésion de reperfusion myocardique/étiologie , Lésion de reperfusion myocardique/métabolisme , Myocytes cardiaques/métabolisme , Phosphorylation , Transduction du signal
16.
Mol Microbiol ; 115(4): 554-573, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33034093

RÉSUMÉ

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.


Sujet(s)
Transporteurs de cations/physiologie , Cuivre/métabolisme , Cuivre/pharmacologie , Régulation de l'expression des gènes bactériens , Manganèse/métabolisme , Staphylococcus aureus/effets des médicaments et des substances chimiques , Staphylococcus aureus/physiologie , Antibactériens/métabolisme , Antibactériens/pharmacologie , Protéines bactériennes/physiologie , ADN bactérien , Humains , Fer/métabolisme , Ferrosulfoprotéines/métabolisme , Protéines de transport membranaire/physiologie , Mutagenèse par insertion , Opéron , ARN bactérien , Protéines de répression/physiologie , Infections à staphylocoques/microbiologie
17.
Cell Rep ; 33(10): 108486, 2020 12 08.
Article de Anglais | MEDLINE | ID: mdl-33296646

RÉSUMÉ

The mitochondrial calcium uniporter is a multi-subunit Ca2+-activated Ca2+ channel, made up of the pore-forming MCU protein, a metazoan-specific EMRE subunit, and MICU1/MICU2, which mediate Ca2+ activation. It has been established that metazoan MCU requires EMRE binding to conduct Ca2+, but how EMRE promotes MCU opening remains unclear. Here, we demonstrate that EMRE controls MCU activity via its transmembrane helix, while using an N-terminal PKP motif to strengthen binding with MCU. Opening of MCU requires hydrophobic interactions mediated by MCU residues near the pore's luminal end. Enhancing these interactions by single mutation allows human MCU to transport Ca2+ without EMRE. We further show that EMRE may facilitate MCU opening by stabilizing the open state in a conserved MCU gating mechanism, present also in non-metazoan MCU homologs. These results provide insights into the evolution of the uniporter machinery and elucidate the mechanism underlying the physiologically crucial EMRE-dependent MCU activation process.


Sujet(s)
Canaux calciques/métabolisme , Protéines de liaison au calcium/métabolisme , Transporteurs de cations/métabolisme , Protéines de transport de la membrane mitochondriale/métabolisme , Calcium/métabolisme , Canaux calciques/physiologie , Canaux calciques/ultrastructure , Protéines de liaison au calcium/physiologie , Protéines de liaison au calcium/ultrastructure , Transporteurs de cations/physiologie , Transporteurs de cations/ultrastructure , Cellules HEK293 , Humains , Mitochondries/métabolisme , Protéines de transport de la membrane mitochondriale/physiologie , Protéines de transport de la membrane mitochondriale/ultrastructure , Membranes mitochondriales/métabolisme
18.
Int J Mol Sci ; 21(21)2020 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-33167316

RÉSUMÉ

Our recent study demonstrated altered expression of Notch ligands, receptors, and effector genes in testes of pubertal rats following reduced androgen production or signaling. Herein we aimed to explore the role of nuclear androgen receptor (AR) and membrane androgen receptor (Zrt- and Irt-like protein 9; ZIP9) in the regulation of Notch pathway activation in rodent Sertoli cells. Experiments were performed using TM4 and 15P-1 Sertoli cell lines and rat primary Sertoli cells (PSC). We found that testosterone (10-8 M-10-6 M) increased the expression of Notch1 receptor, its active form Notch1 intracellular domain (N1ICD) (p < 0.05, p < 0.01, p < 0.001), and the effector genes Hey1 (p < 0.05, p < 0.01, p < 0.001) and Hes1 (p < 0.05, p < 0.001) in Sertoli cells. Knockdown of AR or ZIP9 as well as antiandrogen exposure experiments revealed that (i) action of androgens via both AR and ZIP9 controls Notch1/N1ICD expression and transcriptional activity of recombination signal binding protein (RBP-J), (ii) AR-dependent signaling regulates Hey1 expression, (iii) ZIP9-dependent pathway regulates Hes1 expression. Our findings indicate a crosstalk between androgen and Notch signaling in Sertoli cells and point to cooperation of classical and non-classical androgen signaling pathways in controlling Sertoli cell function.


Sujet(s)
Androgènes/métabolisme , Transporteurs de cations/physiologie , Récepteurs Notch/métabolisme , Cellules de Sertoli/métabolisme , Androgènes/pharmacologie , Androgènes/physiologie , Animaux , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Cellules cultivées , Mâle , Souris , Rats , Interactions entre récepteurs/effets des médicaments et des substances chimiques , Interactions entre récepteurs/physiologie , Récepteurs aux androgènes/génétique , Récepteurs aux androgènes/métabolisme , Protéines de répression/génétique , Protéines de répression/métabolisme , Cellules de Sertoli/effets des médicaments et des substances chimiques , Cellules de Sertoli/physiologie , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/physiologie , Facteur de transcription HES-1/génétique , Facteur de transcription HES-1/métabolisme
19.
Sci Rep ; 10(1): 15993, 2020 10 02.
Article de Anglais | MEDLINE | ID: mdl-33009476

RÉSUMÉ

The mating of 77 heterozygous pairs (Cav3.2[+|-] x Cav3.2[+|-]) revealed a significant deviation of genotype distribution from Mendelian inheritance in weaned pups. The mating of 14 pairs (Cav3.2[-|-] female x Cav3.2[+|-] male) and 8 pairs (Cav3.2[+|-] female x Cav3.2[-|-] male) confirmed the significant reduction of deficient homozygous Cav3.2[-|-] pups, leading to the conclusion that prenatal lethality may occur, when one or both alleles, encoding the Cav3.2T-type Ca2+ channel, are missing. Also, the mating of 63 heterozygous pairs (Cav2.3[+|-] x Cav2.3[+|-]) revealed a significant deviation of genotype distribution from Mendelian inheritance in weaned pups, but only for heterozygous male mice, leading to the conclusion that compensation may only occur for Cav2.3[-|-] male mice lacking both alleles of the R-type Ca2+ channel. During the mating of heterozygous parents, the number of female mice within the weaned population does not deviate from the expected Mendelian inheritance. During prenatal development, both, T- and R-type Ca2+ currents are higher expressed in some tissues than postnatally. It will be discussed that the function of voltage-gated Ca2+ channels during prenatal development must be investigated in more detail, not least to understand devastative diseases like developmental epileptic encephalopathies (DEE).


Sujet(s)
Canaux calciques de type R/physiologie , Canaux calciques de type T/physiologie , Transporteurs de cations/physiologie , Chromosomes/génétique , Instabilité du génome , Croisement consanguin/méthodes , Locus de caractère quantitatif , Animaux , Femelle , Mâle , Souris , Souris de lignée C57BL , Souris knockout
20.
Planta ; 252(3): 35, 2020 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-32767128

RÉSUMÉ

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+.


Sujet(s)
Transport biologique/physiologie , Transporteurs de cations/génétique , Transporteurs de cations/physiologie , Noyau de la cellule/physiologie , Cytosol/physiologie , Vitis/génétique , Vitis/physiologie , Fruit/physiologie , Régulation de l'expression des gènes végétaux , Feuilles de plante/physiologie , Protéines végétales/physiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE