Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(19)2020 Sep 24.
Article in English | MEDLINE | ID: mdl-32987913

ABSTRACT

Ranunculus glacialis grows and reproduces successfully, although the snow-free time period is short (2-3 months) and night frosts are frequent. At a nival site (3185 m a.s.l.), we disentangled the interplay between the atmospheric temperature, leaf temperatures, and leaf freezing frequency to assess the actual strain. For a comprehensive understanding, the freezing behavior from the whole plant to the leaf and cellular level and its physiological after-effects as well as cell wall chemistry were studied. The atmospheric temperatures did not mirror the leaf temperatures, which could be 9.3 °C lower. Leaf freezing occurred even when the air temperature was above 0 °C. Ice nucleation at on average -2.6 °C started usually independently in each leaf, as the shoot is deep-seated in unfrozen soil. All the mesophyll cells were subjected to freezing cytorrhysis. Huge ice masses formed in the intercellular spaces of the spongy parenchyma. After thawing, photosynthesis was unaffected regardless of whether ice had formed. The cell walls were pectin-rich and triglycerides occurred, particularly in the spongy parenchyma. At high elevations, atmospheric temperatures fail to predict plant freezing. Shoot burial prevents ice spreading, specific tissue architecture enables ice management, and the flexibility of cell walls allows recurrent freezing cytorrhysis. The peculiar patterning of triglycerides close to ice rewards further investigation.


Subject(s)
Cell Wall/physiology , Cold-Shock Response , Mesophyll Cells , Ranunculus/physiology , Freezing , Ice , Mesophyll Cells/cytology , Mesophyll Cells/physiology , Photosynthesis
2.
J Exp Med ; 217(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-31914175

ABSTRACT

The gene IL6ST encodes GP130, the common signal transducer of the IL-6 cytokine family consisting of 10 cytokines. Previous studies have identified cytokine-selective IL6ST defects that preserve LIF signaling. We describe three unrelated families with at least five affected individuals who presented with lethal Stüve-Wiedemann-like syndrome characterized by skeletal dysplasia and neonatal lung dysfunction with additional features such as congenital thrombocytopenia, eczematoid dermatitis, renal abnormalities, and defective acute-phase response. We identified essential loss-of-function variants in IL6ST (a homozygous nonsense variant and a homozygous intronic splice variant with exon skipping). Functional tests showed absent cellular responses to GP130-dependent cytokines including IL-6, IL-11, IL-27, oncostatin M (OSM), and leukemia inhibitory factor (LIF). Genetic reconstitution of GP130 by lentiviral transduction in patient-derived cells reversed the signaling defect. This study identifies a new genetic syndrome caused by the complete lack of signaling of a whole family of GP130-dependent cytokines in humans and highlights the importance of the LIF signaling pathway in pre- and perinatal development.


Subject(s)
Cytokine Receptor gp130/metabolism , Exostoses, Multiple Hereditary/metabolism , Osteochondrodysplasias/metabolism , Signal Transduction/physiology , Antigens, CD/metabolism , Cells, Cultured , HEK293 Cells , Humans , Interleukin-11/metabolism , Interleukin-6/metabolism , Leukemia Inhibitory Factor/metabolism , Oncostatin M/metabolism , Receptors, Cytokine/metabolism
3.
Nat Genet ; 50(7): 944-950, 2018 07.
Article in English | MEDLINE | ID: mdl-29867221

ABSTRACT

A major challenge to personalized oncology is that driver mutations vary among cancer cells inhabiting the same tumor. Whether this reflects principally disparate patterns of Darwinian evolution in different tumor regions has remained unexplored1-5. We mapped the prevalence of genetically distinct clones over 250 regions in 54 childhood cancers. This showed that primary tumors can simultaneously follow up to four evolutionary trajectories over different anatomic areas. The most common pattern consists of subclones with very few mutations confined to a single tumor region. The second most common is a stable coexistence, over vast areas, of clones characterized by changes in chromosome numbers. This is contrasted by a third, less frequent, pattern where a clone with driver mutations or structural chromosome rearrangements emerges through a clonal sweep to dominate an anatomical region. The fourth and rarest pattern is the local emergence of a myriad of clones with TP53 inactivation. Death from disease was limited to tumors exhibiting the two last, most dynamic patterns.


Subject(s)
Mutation/genetics , Neoplasms/genetics , Child , Chromosomes/genetics , Evolution, Molecular , Gene Rearrangement/genetics , Humans , Tumor Suppressor Protein p53/genetics
4.
Pediatr Blood Cancer ; 64(8)2017 Aug.
Article in English | MEDLINE | ID: mdl-28074537

ABSTRACT

OBJECTIVE: To assess the effect of neuroblastoma (NB) on the intestinal microbiome, metabolism, and inflammatory parameters in a murine model. MATERIALS AND METHODS: Athymic Hsd:Fox1nu mice received subperitoneal implantation of human NB cells (MHH-NB11) (tumor group, TG) or culture medium (sham group). Following 10 weeks of tumor growth, all animals were sacrificed to collect total white adipose tissue (WAT). Luminex assays were performed for gut hormone and inflammation marker analysis. Bile acids were measured by high-performance liquid chromatography-mass spectrometry in feces and serum. The microbiome of the ileal content was determined by 16S rDNA next-generation sequencing. RESULTS: At 10 weeks, tumors masses in the TG reached a mean weight of 1.10 g (interquartile range 3.45 g) associated with a significant reduction in WAT. Furthermore, in the TG, there was a marked reduction in leptin and an increase in glucagon-like peptide 1 serum levels. Moreover, the TG mice displayed a pro-inflammatory profile, with significant increases in monocyte chemotactic protein 1, tumor necrosis factor alpha, and interleukin-10. Lithocholic acid, deoxycholic acid, and ursodeoxycholic acid were significantly decreased in the stool of TG mice. Significant alterations of the intestinal microbiome were found in the ileal contents of the TG. CONCLUSIONS: The present study provides a first glimpse that human NB in a murine model induces tumor cachexia associated with alterations in metabolic and inflammatory parameters, as well as changes in the intestinal microbiota. Since the intestinal microbiome is known to contribute to the host's ability to harvest energy, a favorable modulation of the intestinal microbiome in tumor patients could potentially represent a novel therapeutic target to prevent tumor-associated cachexia.


Subject(s)
Bile Acids and Salts/metabolism , Cytokines/metabolism , Gastrointestinal Microbiome , Neuroblastoma/pathology , Animals , Cell Line, Tumor , Chromatography, High Pressure Liquid , Disease Models, Animal , Heterografts , Humans , Inflammation/pathology , Male , Mass Spectrometry , Mice , Mice, Nude
5.
Case Rep Pediatr ; 2016: 4103734, 2016.
Article in English | MEDLINE | ID: mdl-27803831

ABSTRACT

Preterm infants are highly susceptible to injuries following necessary and often life-saving medical interventions. Esophageal perforation is a rare, yet serious complication that can be caused by aerodigestive tract suction, endotracheal intubation, or nasogastric tube placement. We present the case of a neonate born at 23 weeks plus three days of gestation with chest radiography showing malposition of the nasogastric feeding tube and massive right-sided effusion of Iopamidol in the pleural cavity due to esophageal perforation. In addition, the article summarizes common signs and symptoms associated with esophageal perforation in infants and discusses diagnostic approaches.

6.
Oncotarget ; 7(23): 33832-40, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27213586

ABSTRACT

Metabolic reprogramming is a hallmark of cancer. Understanding cancer metabolism is instrumental to devise innovative therapeutic approaches. Anabolic metabolism, including the induction of lipogenic enzymes, is a key feature of proliferating cells. Here, we report a novel tumor suppressive function for adipose triglyceride lipase (ATGL), the rate limiting enzyme in the triglyceride hydrolysis cascade.In immunohistochemical analysis, non-small cell lung cancers, pancreatic adenocarcinoma as well as leiomyosarcoma showed significantly reduced levels of ATGL protein compared to corresponding normal tissues. The ATGL gene was frequently deleted in various forms of cancers. Low levels of ATGL mRNA correlated with significantly reduced survival in patients with ovarian, breast, gastric and non-small cell lung cancers. Remarkably, pulmonary neoplasia including invasive adenocarcinoma developed spontaneously in mice lacking ATGL pointing to an important role for this lipase in controlling tumor development.Loss of ATGL, as detected in several forms of human cancer, induces spontaneous development of pulmonary neoplasia in a mouse model. Our results, therefore, suggest a novel tumor suppressor function for ATGL and contribute to the understanding of cancer metabolism. We propose to evaluate loss of ATGL protein expression for the diagnosis of malignant tumors. Finally, modulation of the lipolytic pathway may represent a novel therapeutic approach in the treatment of human cancer.


Subject(s)
Adenocarcinoma/enzymology , Biomarkers, Tumor/analysis , Cell Transformation, Neoplastic/metabolism , Lipase/analysis , Lipase/deficiency , Lung Neoplasms/enzymology , Neoplasms/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Computational Biology , Data Mining , Databases, Genetic , Down-Regulation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Lipase/genetics , Lipolysis , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/genetics , Neoplasms/pathology , Phenotype
7.
Sci Signal ; 9(412): ra10, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26814231

ABSTRACT

STIM1 (stromal interaction molecule 1) and Orai proteins are the essential components of Ca(2+) release-activated Ca(2+) (CRAC) channels. We focused on the role of cholesterol in the regulation of STIM1-mediated Orai1 currents. Chemically induced cholesterol depletion enhanced store-operated Ca(2+) entry (SOCE) and Orai1 currents. Furthermore, cholesterol depletion in mucosal-type mast cells augmented endogenous CRAC currents, which were associated with increased degranulation, a process that requires calcium influx. Single point mutations in the Orai1 amino terminus that would be expected to abolish cholesterol binding enhanced SOCE to a similar extent as did cholesterol depletion. The increase in Orai1 activity in cells expressing these cholesterol-binding-deficient mutants occurred without affecting the amount in the plasma membrane or the coupling of STIM1 to Orai1. We detected cholesterol binding to an Orai1 amino-terminal fragment in vitro and to full-length Orai1 in cells. Thus, our data showed that Orai1 senses the amount of cholesterol in the plasma membrane and that the interaction of Orai1 with cholesterol inhibits its activity, thereby limiting SOCE.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Cholesterol/metabolism , Biotinylation , Cell Line , Cell Membrane/metabolism , Cholesterol Oxidase/metabolism , Circular Dichroism , Electrophysiological Phenomena , Fluorescence Resonance Energy Transfer , HEK293 Cells , Histamine/metabolism , Humans , Mast Cells/metabolism , Mutation , ORAI1 Protein , Peptides/metabolism , Point Mutation , Protein Structure, Tertiary , Signal Transduction , Spectrometry, Fluorescence
8.
Sci Signal ; 8(408): ra131, 2015 12 22.
Article in English | MEDLINE | ID: mdl-26696631

ABSTRACT

The Ca(2+) release-activated Ca(2+) channel mediates Ca(2+) influx in a plethora of cell types, thereby controlling diverse cellular functions. The channel complex is composed of stromal interaction molecule 1 (STIM1), an endoplasmic reticulum Ca(2+)-sensing protein, and Orai1, a plasma membrane Ca(2+) channel. Channels composed of STIM1 and Orai1 mediate Ca(2+) influx even at low extracellular Ca(2+) concentrations. We investigated whether the activity of Orai1 adapted to different environmental Ca(2+) concentrations. We used homology modeling and molecular dynamics simulations to predict the presence of an extracellular Ca(2+)-accumulating region (CAR) at the pore entrance of Orai1. Furthermore, simulations of Orai1 proteins with mutations in CAR, along with live-cell experiments, or simulations and electrophysiological recordings of the channel with transient, electrostatic loop3 interacting with loop1 (the site of CAR) determined that CAR enhanced Ca(2+) permeation most efficiently at low external Ca(2+) concentrations. Consistent with these results, cells expressing Orai1 CAR mutants exhibited impaired gene expression stimulated by the Ca(2+)-activated transcription factor nuclear factor of activated T cells (NFAT). We propose that the Orai1 channel architecture with a close proximity of CAR to the selectivity filter, which enables Ca(2+)-selective ion permeation, enhances the local extracellular Ca(2+) concentration to maintain Ca(2+)-dependent gene regulation even in environments with relatively low Ca(2+)concentrations.


Subject(s)
Calcium/metabolism , Cell Membrane Permeability/physiology , Drosophila Proteins , Membrane Proteins , Transcription, Genetic/physiology , Animals , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , HEK293 Cells , Humans , Ion Transport/physiology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , ORAI1 Protein , Protein Structure, Secondary , Stromal Interaction Molecule 1
9.
J Pediatr Hematol Oncol ; 37(8): e486-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26165406

ABSTRACT

A 4 ½-year-old female was diagnosed with ovarian juvenile granulosa cell tumor stage IA. After complete tumor resection she received 4 courses of chemotherapy due to unfavorable histopathologic features (high mitotic index, high microvessel density, blood vessel invasion). One year after diagnosis, she experienced paraaortic lymph node relapse treated with surgery, local radiotherapy, and conventional and high-dose chemotherapy. A second, paratracheal lymph node relapse 7 months later necessitated surgical removal and radiotherapy. Subsequently an adjuvant antiangiogenesis-based treatment including paclitaxel, bevacizumab, thalidomide, and pegylated interferon was initiated and continued for 2 years. The female is now in third complete remission 6 years after second relapse.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Granulosa Cell Tumor/secondary , Ovarian Neoplasms/drug therapy , Salvage Therapy , Child, Preschool , Female , Granulosa Cell Tumor/therapy , Humans , Lymph Node Excision , Lymphatic Irradiation , Lymphatic Metastasis/radiotherapy , Neoplasm Staging , Neovascularization, Pathologic/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/surgery , Ovariectomy , Remission Induction
10.
Semin Oncol ; 41(2): 281-95, 2014 04.
Article in English | MEDLINE | ID: mdl-24787299

ABSTRACT

In this review we introduce the concept of the tumor macroenvironment and explore it in the context of metabolism. Tumor cells interact with the tumor microenvironment including immune cells. Blood and lymph vessels are the critical components that deliver nutrients to the tumor and also connect the tumor to the macroenvironment. Several factors are then released from the tumor itself but potentially also from the tumor microenvironment, influencing the metabolism of distant tissues and organs. Amino acids, and distinct lipid and lipoprotein species can be essential for further tumor growth. The role of glucose in tumor metabolism has been studied extensively. Cancer-associated cachexia is the most important tumor-associated systemic syndrome and not only affects the quality of life of patients with various malignancies but is estimated to be the cause of death in 15%-20% of all cancer patients. On the other hand, systemic metabolic diseases such as obesity and diabetes are known to influence tumor development. Furthermore, the clinical implications of the tumor macroenvironment are explored in the context of the patient's outcome with special consideration for pediatric tumors. Finally, ways to target the tumor macroenvironment that will provide new approaches for therapeutic concepts are described.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Adipose Tissue/metabolism , Amino Acids/metabolism , Animals , Cachexia/metabolism , Diabetes Complications , Diabetes Mellitus/metabolism , Female , Humans , Inflammation , Lipid Metabolism , Lipids/blood , Male , Neovascularization, Pathologic , Obesity/complications , Proteins/metabolism , Stromal Cells/cytology , Tumor Microenvironment
12.
J Minim Invasive Gynecol ; 20(5): 714-6, 2013.
Article in English | MEDLINE | ID: mdl-23680516

ABSTRACT

Intrauterine ovarian torsion is a rare event, but it is a possible cause for unilateral ovarian aplasia. Most commonly the ovary undergoes autolysis after torsion so that no tissue or remnants can be discovered on the involved side. We report a rare case of unilateral intrauterine torsion followed by autoamputation and abdominal reimplantation resulting in an intra-abdominal complex cystic mass with a review of the literature.


Subject(s)
Abdomen/surgery , Ovarian Cysts/surgery , Torsion Abnormality/surgery , Abdomen/diagnostic imaging , Female , Humans , Infant , Ovarian Cysts/diagnostic imaging , Torsion Abnormality/diagnostic imaging , Treatment Outcome , Ultrasonography
13.
EMBO J ; 30(9): 1678-89, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21427704

ABSTRACT

Stromal interaction molecule (STIM1) and ORAI1 are key components of the Ca(2+) release-activated Ca(2+) (CRAC) current having an important role in T-cell activation and mast cell degranulation. CRAC channel activation occurs via physical interaction of ORAI1 with STIM1 when endoplasmic reticulum Ca(2+) stores are depleted. Here we show, utilizing a novel STIM1-derived Förster resonance energy transfer sensor, that the ORAI1 activating small fragment (OASF) undergoes a C-terminal, intramolecular transition into an extended conformation when activating ORAI1. The C-terminal rearrangement of STIM1 does not require a functional CRAC channel, suggesting interaction with ORAI1 as sufficient for this conformational switch. Extended conformations were also engineered by mutations within the first and third coiled-coil domains in the cytosolic portion of STIM1 revealing the involvement of hydrophobic residues in the intramolecular transition. Corresponding full-length STIM1 mutants exhibited enhanced interaction with ORAI1 inducing constitutive CRAC currents, even in the absence of store depletion. We suggest that these mutant STIM1 proteins imitate a physiological activated state, which mimics the intramolecular transition that occurs in native STIM1 upon store depletion.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/physiology , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Conformation , Blotting, Western , Chromatography, Gel , Cloning, Molecular , Electrophysiology , Fluorescence Resonance Energy Transfer , Humans , Membrane Proteins/genetics , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Neoplasm Proteins/genetics , ORAI1 Protein , Stromal Interaction Molecule 1 , Transfection
14.
J Biol Chem ; 286(10): 8577-8584, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21220423

ABSTRACT

Activation of immune cells is triggered by the Ca(2+) release-activated Ca(2+) current, which is mediated via channels of the Orai protein family. A key gating process of the three Orai channel isoforms to prevent Ca(2+) overload is fast inactivation, most pronounced in Orai3. A subsequent reactivation is a unique gating characteristic of Orai1 channels, whereas Orai2 and Orai3 currents display a second, slow inactivation phase. Employing a chimeric approach by sequential swapping of respective intra- and extracellular regions between Orai1 and Orai3, we show here that Orai1 specific proline/arginine-rich domains in the N terminus mediate reactivation, whereas the second, intracellular loop modulates fast and slow gating processes. Swapping C-terminal strands lacks a significant impact. However, simultaneous transfer of Orai3 N terminus and its second loop or C terminus in an Orai1 chimera substantially increases fast inactivation centered between wild-type channels. Concomitant swap of all three cytosolic strands from Orai3 onto Orai1 fully conveys Orai3-like gating characteristics, in a strongly cooperative manner. In conclusion, Orai subtype-specific gating requires a cooperative interplay of all three cytosolic domains.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Ion Channel Gating/physiology , Calcium Channels/genetics , Cell Line , Cytosol/metabolism , Humans , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
15.
J Biol Chem ; 285(52): 41135-42, 2010 Dec 24.
Article in English | MEDLINE | ID: mdl-20961852

ABSTRACT

Store-operated calcium entry is essential for many signaling processes in nonexcitable cells. The best studied store-operated calcium current is the calcium release-activated calcium (CRAC) current in T-cells and mast cells, with Orai1 representing the essential pore forming subunit. Although it is known that functional CRAC channels in store-depleted cells are composed of four Orai1 subunits, the stoichiometric composition in quiescent cells is still discussed controversially: both a tetrameric and a dimeric stoichiometry of resting state Orai1 have been reported. We obtained here robust and similar FRET values on labeled tandem repeat constructs of Orai1 before and after store depletion, suggesting an unchanged tetrameric stoichiometry. Moreover, we directly visualized the stoichiometry of mobile Orai1 channels in live cells using a new single molecule recording modality that combines single molecule tracking and brightness analysis. By alternating imaging and photobleaching pulses, we recorded trajectories of single, fluorescently labeled Orai1 channels, with each trajectory consisting of bright and dim segments, corresponding to higher and lower numbers of colocalized active GFP label. The according brightness values were used for global fitting and statistical analysis, yielding a tetrameric subunit composition of mobile Orai1 channels in resting cells.


Subject(s)
Calcium Channels/metabolism , Cell Membrane/metabolism , Protein Multimerization/physiology , Animals , CHO Cells , Calcium Channels/genetics , Cell Membrane/genetics , Cricetinae , Cricetulus , HEK293 Cells , Humans , ORAI1 Protein , Protein Structure, Quaternary
16.
Proc Natl Acad Sci U S A ; 106(46): 19623-8, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19887627

ABSTRACT

A general cellular response following depletion of intracellular calcium stores involves activation of store-operated channels (SOCs). While Orai1 forms the native Ca(2+) release-activated Ca(2+) (CRAC) channel in mast and T cells, the molecular architecture of less Ca(2+) selective SOCs is insufficiently defined. Here we present evidence that diminished Ca(2+) selectivity and robust Cs(+) permeation together with a reduced fast inactivation are characteristics of heteromeric Orai1 and Orai3 channels in contrast to their homomeric forms. The first extracellular loop of these Orai isoforms differs by two aspartates replacing glutamates that affect the selectivity. Co-expression of an Orai3 mutant that mimicked the first loop of Orai1 with either Orai1 or Orai3 recovered or decreased Ca(2+) selectivity, respectively. Heteromeric Orai1/3 protein assembly provides a concept for less Ca(2+)-selective SOCs.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Calcium Channels/genetics , Cell Line , Cesium/metabolism , Humans , ORAI1 Protein , Protein Multimerization
17.
J Biol Chem ; 284(37): 24933-8, 2009 Sep 11.
Article in English | MEDLINE | ID: mdl-19622747

ABSTRACT

STIM1 and ORAI1, the two limiting components in the Ca(2+) release-activated Ca(2+) (CRAC) signaling cascade, have been reported to interact upon store depletion, culminating in CRAC current activation. We have recently identified a modulatory domain between amino acids 474 and 485 in the cytosolic part of STIM1 that comprises 7 negatively charged residues. A STIM1 C-terminal fragment lacking this domain exhibits enhanced interaction with ORAI1 and 2-3-fold higher ORAI1/CRAC current densities. Here we focused on the role of this CRAC modulatory domain (CMD) in the fast inactivation of ORAI1/CRAC channels, utilizing the whole-cell patch clamp technique. STIM1 mutants either with C-terminal deletions including CMD or with 7 alanines replacing the negative amino acids within CMD gave rise to ORAI1 currents that displayed significantly reduced or even abolished inactivation when compared with STIM1 mutants with preserved CMD. Consistent results were obtained with cytosolic C-terminal fragments of STIM1, both in ORAI1-expressing HEK 293 cells and in RBL-2H3 mast cells containing endogenous CRAC channels. Inactivation of the latter, however, was much more pronounced than that of ORAI1. The extent of inactivation of ORAI3 channels, which is also considerably more prominent than that of ORAI1, was also substantially reduced by co-expression of STIM1 constructs missing CMD. Regarding the dependence of inactivation on Ca(2+), a decrease in intracellular Ca(2+) chelator concentrations promoted ORAI1 current fast inactivation, whereas Ba(2+) substitution for extracellular Ca(2+) completely abrogated it. In summary, CMD within the STIM1 cytosolic part provides a negative feedback signal to Ca(2+) entry by triggering fast Ca(2+)-dependent inactivation of ORAI/CRAC channels.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Calcium Signaling , Cell Line , Cloning, Molecular , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Humans , Models, Biological , Mutation , ORAI1 Protein , Patch-Clamp Techniques , Protein Structure, Tertiary , Stromal Interaction Molecule 1
SELECTION OF CITATIONS
SEARCH DETAIL
...