Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Physiol ; 599(19): 4415-4426, 2021 10.
Article in English | MEDLINE | ID: mdl-34411300

ABSTRACT

Ryanodine receptors (RyRs) are intracellular Ca2+ release channels ubiquitously expressed in various cell types. RyRs were extensively studied in striated muscle cells due to their crucial role in muscle contraction. In contrast, the role of RyRs in Ca2+ signalling and functions in non-excitable cells, such as T lymphocytes, remains poorly understood. Expression of different isoforms of RyRs was shown in primary T cells and T cell lines. In T cells, RyRs co-localize with the plasmalemmal store-operated Ca2+ channels of the Orai family and endoplasmic reticulum Ca2+ sensing Stim family proteins and are activated by store-operated Ca2+ entry and pyridine nucleotide metabolites, the intracellular second messengers generated upon stimulation of T cell receptors. Experimental data indicate that together with d-myo-inositol 1,4,5-trisphosphate receptors, RyRs regulate intercellular Ca2+ dynamics by controlling Ca2+ concentration within the lumen of the endoplasmic reticulum and, consequently, store-operated Ca2+ entry. Gain-of-function mutations, genetic deletion or pharmacological inhibition of RyRs alters T cell Ca2+ signalling and effector functions. The picture emerging from the collective data shows that RyRs are the essential regulators of T cell Ca2+ signalling and can be potentially used as molecular targets for immunomodulation or T cell-based diagnostics of the disorders associated with RyRs dysregulation.


Subject(s)
Calcium , Ryanodine Receptor Calcium Release Channel , Calcium/metabolism , Calcium Signaling , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Receptors, Antigen, T-Cell , Ryanodine , Ryanodine Receptor Calcium Release Channel/metabolism , T-Lymphocytes
2.
Channels (Austin) ; 15(1): 67-78, 2021 12.
Article in English | MEDLINE | ID: mdl-33356832

ABSTRACT

In the last 5 years inhibitors of the potassium channel KV1.3 have been shown to reduce neuroinflammation in rodent models of ischemic stroke, Alzheimer's disease, Parkinson's disease and traumatic brain injury. At the systemic level these beneficial actions are mediated by a reduction in microglia activation and a suppression of pro-inflammatory cytokine and nitric oxide production. However, the molecular mechanisms for the suppressive action of KV1.3 blockers on pro-inflammatory microglia functions was not known until our group recently demonstrated that KV1.3 channels not only regulate membrane potential, as would be expected of a voltage-gated potassium channel, but also play a crucial role in enabling microglia to resist depolarizations produced by the danger signal ATP thus regulating calcium influx through P2X4 receptors. We here review the role of KV1.3 in microglial signaling and show that, similarly to their role in T cells, KV1.3 channels also regulated store-operated calcium influx in microglia.


Subject(s)
Microglia , Alzheimer Disease , Calcium Signaling , Membrane Potentials , Neuroinflammatory Diseases , Potassium Channels, Voltage-Gated
3.
Front Physiol ; 12: 770820, 2021.
Article in English | MEDLINE | ID: mdl-35027891

ABSTRACT

Ryanodine receptors (RyRs), the intracellular Ca2+ release channels, are expressed in T lymphocytes and other types of immune cells. Modulation of RyRs has been shown to affect T cell functions in vitro and immune responses in vivo. The effects of modulation of RyRs on the development of autoimmune diseases have not been investigated. Here we studied how modulation of RyRs through administration of RyR inhibitor dantrolene or introducing a gain-of-function RYR1-p.R163C mutation affects clinical progression of experimental autoimmune encephalomyelitis (EAE) in mice, a T cell-mediated autoimmune neuroinflammatory disease. We found that daily intraperitoneal administration of 5 or 10 mg/kg dantrolene beginning at the time of EAE induction significantly reduced the severity of EAE clinical symptoms and dampened inflammation in the spinal cord. The protective effect of dantrolene on EAE was reversible. Dantrolene administration elicited dose-dependent skeletal muscle weakness: mice that received 10 mg/kg dose developed a waddling gait, while 5 mg/kg dantrolene dose administration produced a reduction in four-limb holding impulse values. Mice bearing the gain-of-function RYR1-p.R163C mutation developed the EAE clinical symptoms faster and more severely than wild-type mice. This study demonstrates that RyRs play a significant role in EAE pathogenesis and suggests that inhibition of RyRs with low doses of dantrolene may have a protective effect against autoimmunity and inflammation in humans.

4.
Cell Calcium ; 93: 102325, 2021 01.
Article in English | MEDLINE | ID: mdl-33310301

ABSTRACT

Gain-of-function RyR1-p.R163C mutation in ryanodine receptors type 1 (RyR1) deregulates Ca2+ signaling and mitochondrial function in skeletal muscle and causes malignant hyperthermia in humans and mice under triggering conditions. We investigated whether T lymphocytes from heterozygous RyR1-p.R163C knock-in mutant mice (HET T cells) display measurable aberrations in resting cytosolic Ca2+ concentration ([Ca2+]i), Ca2+ release from the store, store-operated Ca2+ entry (SOCE), and mitochondrial inner membrane potential (ΔΨm) compared with T lymphocytes from wild-type mice (WT T cells). We explored whether these variables can be used to distinguish between T cells with normal and altered RyR1 genotype. HET and WT T cells were isolated from spleen and lymph nodes and activated in vitro using phytohemagglutinin P. [Ca2+]i and ΔΨm dynamics were examined using Fura 2 and tetramethylrhodamine methyl ester fluorescent dyes, respectively. Activated HET T cells displayed elevated resting [Ca2+]i, diminished responses to Ca2+ mobilization with thapsigargin, and decreased rate of [Ca2+]i elevation in response to SOCE compared with WT T cells. Pretreatment of HET T cells with ryanodine or dantrolene sodium reduced disparities in the resting [Ca2+]i and ability of thapsigargin to mobilize Ca2+ between HET and WT T cells. While SOCE elicited dissipation of the ΔΨm in WT T cells, it produced ΔΨm hyperpolarization in HET T cells. When used as the classification variable, the amplitude of thapsigargin-induced Ca2+ transient showed the best promise in predicting the presence of RyR1-p.R163C mutation. Other significant variables identified by machine learning analysis were the ratio of resting cytosolic Ca2+ level to the amplitude of thapsigargin-induced Ca2+ transient and an integral of changes in ΔΨm in response to SOCE. Our study demonstrated that gain-of-function mutation in RyR1 significantly affects Ca2+ signaling and mitochondrial fiction in T lymphocytes, which suggests that this mutation may cause altered immune responses in its carrier. Our data link the RyR1-p.R163C mutation, which causes inherited skeletal muscle diseases, to deregulation of Ca2+ signaling and mitochondrial function in immune T cells and establish proof-of-principle for in vitro T cell-based diagnostic assay for hereditary RyR1 hyperfunction.


Subject(s)
Calcium Signaling , Intracellular Space/metabolism , Malignant Hyperthermia/immunology , Mitochondria/metabolism , T-Lymphocytes/immunology , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cell Proliferation/drug effects , Genotype , Lymphocyte Activation/drug effects , Machine Learning , Malignant Hyperthermia/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mutant Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Thapsigargin/pharmacology
5.
Cell Calcium ; 87: 102184, 2020 05.
Article in English | MEDLINE | ID: mdl-32151786

ABSTRACT

Neuroendocrine adrenal chromaffin cells release neurohormones catecholamines in response to Ca2+ entry via voltage-gated Ca2+ channels (VGCCs). Adrenal chromaffin cells also express non-voltage-gated channels, which may conduct Ca2+ at negative membrane potentials, whose role in regulation of exocytosis is poorly understood. We explored how modulation of Ca2+ influx at negative membrane potentials affects basal cytosolic Ca2+ concentration ([Ca2+]i) and exocytosis in metabolically intact voltage-clamped bovine adrenal chromaffin cells. We found that in these cells, Ca2+ entry at negative membrane potentials is balanced by Ca2+ extrusion by the Na+/Ca2+ exchanger and that this balance can be altered by membrane hyperpolarization or stimulation with an inflammatory hormone bradykinin. Membrane hyperpolarization or application of bradykinin augmented Ca2+-carrying current at negative membrane potentials, elevated basal [Ca2+]i, and facilitated synchronous exocytosis evoked by the small amounts of Ca2+ injected into the cell via VGCCs (up to 20 pC). Exocytotic responses evoked by the injections of the larger amounts of Ca2+ via VGCCs (> 20 pC) were suppressed by preceding hyperpolarization. In the absence of Ca2+ entry via VGCCs and Ca2+ extrusion via the Na+/Ca2+ exchanger, membrane hyperpolarization induced a significant elevation in [Ca2+]i and asynchronous exocytosis. Our results indicate that physiological interferences, such as membrane hyperpolarization and/or activation of non-voltage-gated Ca2+ channels, modulate basal [Ca2+]i and, consequently, segregation of exocytotic vesicles and their readiness to be released spontaneously and in response to Ca2+ entry via VGCCs. These mechanisms may play role in homeostatic plasticity of neuronal and endocrine cells.


Subject(s)
Calcium/metabolism , Exocytosis , Membrane Potentials , Neuroendocrine Cells/metabolism , Animals , Bradykinin/pharmacology , Cattle , Exocytosis/drug effects , Membrane Potentials/drug effects , Models, Biological , Neuroendocrine Cells/drug effects , Receptors, Bradykinin/metabolism , Solutions
6.
J Biol Chem ; 287(44): 37233-44, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22948152

ABSTRACT

The expression and functional significance of ryanodine receptors (RyR) were investigated in resting and activated primary human T cells. RyR1, RyR2, and RyR3 transcripts were detected in human T cells. RyR1/2 transcript levels increased, whereas those of RyR3 decreased after T cell activation. RyR1/2 protein immunoreactivity was detected in activated but not in resting T cells. The RyR agonist caffeine evoked Ca(2+) release from the intracellular store in activated T cells but not in resting T cells, indicating that RyR are functionally up-regulated in activated T cells compared with resting T cells. In the presence of store-operated Ca(2+) entry (SOCE) via plasmalemmal Ca(2+) release-activated Ca(2+) (CRAC) channels, RyR blockers reduced the Ca(2+) leak from the endoplasmic reticulum (ER) and the magnitude of SOCE, suggesting that a positive feedback relationship exists between RyR and CRAC channels. Overexpression of fluorescently tagged RyR2 and stromal interaction molecule 1 (STIM1), an ER Ca(2+) sensor gating CRAC channels, in HEK293 cells revealed that RyR are co-localized with STIM1 in the puncta formed after store depletion. These data indicate that in primary human T cells, the RyR are coupled to CRAC channel machinery such that SOCE activates RyR via a Ca(2+)-induced Ca(2+) release mechanism, which in turn reduces the Ca(2+) concentration within the ER lumen in the vicinity of STIM1, thus facilitating SOCE by reducing store-dependent CRAC channel inactivation. Treatment with RyR blockers suppressed activated T cell expansion, demonstrating the functional importance of RyR in T cells.


Subject(s)
Calcium Signaling , Ryanodine Receptor Calcium Release Channel/metabolism , T-Lymphocytes/metabolism , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Cell Proliferation , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Gene Expression , HEK293 Cells , Humans , Jurkat Cells , Lymphocyte Activation , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Stromal Interaction Molecule 1 , T-Lymphocytes/physiology , Up-Regulation
7.
Channels (Austin) ; 5(6): 510-7, 2011.
Article in English | MEDLINE | ID: mdl-22172731

ABSTRACT

CRAC channel-mediated Ca(2+) entry plays a crucial role in T lymphocyte activation. Activated T cells display enhanced Ca(2+) signaling compared with resting T cells; this is partially attributed to activation-induced upregulation of CRAC channel expression. Orai and Stim family genes encode CRAC channel structural elements and regulatory proteins, respectively, but studies of their expression in T cells have led to controversial results. We re-examined Orai and Stim gene expression in resting, activated, and Jurkat T cells. Levels of Orai1 transcripts, encoding the human T cell CRAC channel subunit, were not significantly different between resting T and activated T cells. The total amount of all Orai transcripts was 2-fold higher in activated T cells than in resting T cells. Orai1 and total Orai transcript levels were significantly higher in Jurkat T cells than those in resting T cells. Stim expression did not vary significantly among cell types. Maximal whole-cell CRAC current amplitudes were 1.4-fold and 2.3-fold higher in activated and Jurkat T cells, respectively, than in resting T cells. Due to the small size of resting T cells, the surface CRAC channel density was 2.5-fold and 1.6-fold higher in resting T cells than in activated and Jurkat T cells, respectively. Predicted the rates of cytosolic Ca(2+) elevation calculated using the average values of CRAC channel currents and cell volumes showed that < 2-fold increase in the functional CRAC channel expression level cannot account for the enhanced rate of store-operated Ca(2+) entry in activated T cells compared with resting T cells.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Gene Expression Regulation/physiology , Lymphocyte Activation/physiology , T-Lymphocytes/metabolism , Female , Humans , Jurkat Cells , Male , ORAI1 Protein , T-Lymphocytes/cytology
8.
J Vis Exp ; (46)2010 Dec 21.
Article in English | MEDLINE | ID: mdl-21389932

ABSTRACT

In T lymphocytes, depletion of Ca(2+) from the intracellular Ca(2+) store leads to activation of plasmalemmal Ca(2+) channels, called Calcium Release-Activated Calcium (CRAC) channels. CRAC channels play important role in regulation of T cell proliferation and gene expression. Abnormal CRAC channel function in T cells has been linked to severe combined immunodeficiency and autoimmune diseases. Studying CRAC channel function in human T cells may uncover new molecular mechanisms regulating normal immune responses and unravel the causes of related human diseases. Electrophysiological recordings of membrane currents provide the most accurate assessment of functional channel properties and their regulation. Electrophysiological assessment of CRAC channel currents in Jurkat T cells, a human leukemia T cell line, was first performed more than 20 years ago, however, CRAC current measurements in normal human T cells remains a challenging task. The difficulties in recording CRAC channel currents in normal T cells are compounded by the fact that blood-derived T lymphocytes are much smaller in size than Jurkat T cells and, therefore, the endogenous whole-cell CRAC currents are very low in amplitude. Here, we give a step-by-step procedure that we routinely use to record the Ca(2+) or Na(+) currents via CRAC channels in resting human T cells isolated from the peripheral blood of healthy volunteers. The method described here was adopted from the procedures used for recording the CRAC currents in Jurkat T cells and activated human T cells.


Subject(s)
Calcium Channels/physiology , Patch-Clamp Techniques/methods , T-Lymphocytes/physiology , Electrophysiological Phenomena , Humans , Jurkat Cells
9.
J Cell Physiol ; 221(1): 84-91, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19492415

ABSTRACT

A jumonji domain containing gene 6 (Jmjd6), previously referred to as phosphatidylserine receptor (PSR) gene, plays an important role in cell differentiation and development of multiple organs, although mechanisms of its action are not known. The Jmjd6 gene product was initially identified as a membrane protein that participates in phagocytosis. However, the later findings that recombinant Jmjd6 in expression systems was targeted to the nucleus challenged the role of Jmjd6 as a membrane receptor. Using immunocytochemistry approach we studied the subcellular distribution of endogenous Jmjd6 protein in THP-1 cells activated with phorbol 12-myristate 13 acetate (PMA). We found that treatment with PMA stimulated Jmjd6 expression in the cytosol of activated cells. Furthermore, Jmjd6 initially appeared at the cell surface of immature phagocytes (1-2 days after activation) but then translocated into the nucleus of differentiated macrophage-like cells (5-9 days after activation). Anti-Jmjd6 antibodies suppressed the engulfment of dead cell corpses by THP-1 cells expressing the Jmjd6 at the cell surface. These data indicate that Jmjd6 serves as a membrane-associated receptor that regulates phagocytosis in immature macrophages but is dispensable for phagocytosis and has other functions when it is expressed in the cytosol and nucleus of mature macrophage-like cells.


Subject(s)
Cell Differentiation , Cell Membrane/metabolism , Monocytes/cytology , Monocytes/metabolism , Phagocytosis , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Antibodies/pharmacology , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Membrane/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytosol/drug effects , Cytosol/metabolism , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Jumonji Domain-Containing Histone Demethylases , Monocytes/drug effects , Phagocytosis/drug effects , Protein Transport/drug effects , Recombinant Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tetradecanoylphorbol Acetate/pharmacology
10.
Am J Physiol Cell Physiol ; 296(5): C1098-104, 2009 May.
Article in English | MEDLINE | ID: mdl-19295174

ABSTRACT

In blood cells, changes in intracellular Ca(2+) concentration ([Ca(2+)](i)) are associated with multiple cellular events, including activation of cellular kinases and phosphatases, degranulation, regulation of cytoskeleton binding proteins, transcriptional control, and modulation of surface receptors. Although there is no doubt as to the significance of Ca(2+) signaling in blood cells, there is sparse knowledge about the molecular identities of the plasmalemmal Ca(2+) permeable channels that control Ca(2+) fluxes across the plasma membrane and mediate changes in [Ca(2+)](i) in blood cells. Using RNA expression analysis, we have shown that human leukemia K562 cells endogenously coexpress transient receptor potential vanilloid channels type 5 (TRPV5) and type 6 (TRPV6) mRNAs. Moreover, we demonstrated that TRPV5 and TRPV6 channel proteins are present in both the total lysates and the crude membrane preparations from leukemia cells. Immunoprecipitation revealed that a physical interaction between TRPV5 and TRPV6 may take place. Single-channel patch-clamp experiments demonstrated the presence of inwardly rectifying monovalent currents that displayed kinetic characteristics of unitary TRPV5 and/or TRPV6 currents and were blocked by extracellular Ca(2+) and ruthenium red. Taken together, our data strongly indicate that human myeloid leukemia cells coexpress functional TRPV5 and TRPV6 calcium channels that may interact with each other and contribute into intracellular Ca(2+) signaling.


Subject(s)
Calcium Channels/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology , TRPV Cation Channels/genetics , Calcium/pharmacokinetics , Calcium Channels/metabolism , Calcium Signaling/physiology , Humans , Indicators and Reagents/pharmacokinetics , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Membrane Potentials/physiology , Patch-Clamp Techniques , RNA, Messenger/metabolism , Ruthenium Red/pharmacokinetics , TRPV Cation Channels/metabolism
11.
Neural Dev ; 3: 22, 2008 Sep 11.
Article in English | MEDLINE | ID: mdl-18786248

ABSTRACT

BACKGROUND: Olfactory discrimination depends on the large numbers of odorant receptor genes and differential ligand-receptor signaling among neurons expressing different receptors. In this study, we describe an in vitro system that enables the expression of exogenous odorant receptors in cultured olfactory sensory neurons. Olfactory sensory neurons in the culture express characteristic signaling molecules and, therefore, provide a system to study receptor function within its intrinsic cellular environment. RESULTS: We demonstrate that cultured olfactory sensory neurons express endogenous odorant receptors. Lentiviral vector-mediated gene transfer enables successful ectopic expression of odorant receptors. We show that the ectopically expressed mouse I7 is functional in the cultured olfactory sensory neurons. When two different odorant receptors are ectopically expressed simultaneously, both receptor proteins co-localized in the same olfactory sensory neurons up to 10 days in vitro. CONCLUSION: This culture technique provided an efficient method to culture olfactory sensory neurons whose morphology, molecular characteristics and maturation progression resembled those observed in vivo. Using this system, regulation of odorant receptor expression and its ligand specificity can be studied in its intrinsic cellular environment.


Subject(s)
Gene Transfer Techniques , Lentivirus/genetics , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/physiology , Receptors, Odorant/genetics , Animals , Axons/physiology , Biomarkers , Cells, Cultured , Female , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Olfactory Receptor Neurons/ultrastructure , Pregnancy
12.
J Biol Chem ; 283(18): 12512-9, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18316371

ABSTRACT

The precise control of many T cell functions relies on cytosolic Ca(2+) dynamics that is shaped by the Ca(2+) release from the intracellular store and extracellular Ca(2+) influx. The Ca(2+) influx activated following T cell receptor (TCR)-mediated store depletion is considered to be a major mechanism for sustained elevation in cytosolic Ca(2+) concentration ([Ca(2+)](i)) necessary for T cell activation, whereas the role of intracellular Ca(2+) release channels is believed to be minor. We found, however, that in Jurkat T cells [Ca(2+)](i) elevation observed upon activation of the store-operated Ca(2+) entry (SOCE) by passive store depletion with cyclopiazonic acid, a reversible blocker of sarco-endoplasmic reticulum Ca(2+)-ATPase, inversely correlated with store refilling. This indicated that intracellular Ca(2+) release channels were activated in parallel with SOCE and contributed to global [Ca(2+)](i) elevation. Pretreating cells with (-)-xestospongin C (10 microM) or ryanodine (400 microM), the antagonists of inositol 1,4,5-trisphosphate receptor (IP3R) or ryanodine receptor (RyR), respectively, facilitated store refilling and significantly reduced [Ca(2+)](i) elevation evoked by the passive store depletion or TCR ligation. Although the Ca(2+) release from the IP3R can be activated by TCR stimulation, the Ca(2+) release from the RyR was not inducible via TCR engagement and was exclusively activated by the SOCE. We also established that inhibition of IP3R or RyR down-regulated T cell proliferation and T-cell growth factor interleukin 2 production. These studies revealed a new aspect of [Ca(2+)](i) signaling in T cells, that is SOCE-dependent Ca(2+) release via IP3R and/or RyR, and identified the IP3R and RyR as potential targets for manipulation of Ca(2+)-dependent functions of T lymphocytes.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Intracellular Space/metabolism , Lymphocyte Activation , T-Lymphocytes/cytology , Calcium Signaling/drug effects , Cell Line , Cell Proliferation/drug effects , Dantrolene/pharmacology , Down-Regulation/drug effects , Humans , Indoles/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Interleukin-2/biosynthesis , Intracellular Space/drug effects , Lymphocyte Activation/drug effects , Macrocyclic Compounds/pharmacology , Models, Immunological , Oxazoles/pharmacology , Receptors, Antigen, T-Cell/agonists , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , T-Lymphocytes/drug effects
13.
J Cell Physiol ; 212(1): 174-81, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17299798

ABSTRACT

Activated T lymphocytes release vesicles, termed exosomes, enriched in cholesterol and exposing phosphatidylserine (PS) at their outer membrane leaflet. Although CD4(+) activated T lymphocytes infiltrate an atherosclerotic plaque, the effects of T cell exosomes on the atheroma-associated cells are not known. We report here that exosomes isolated from the supernatants of activated human CD4(+) T cells enhance cholesterol accumulation in cultured human monocytes and THP-1 cells. Lipid droplets found in the cytosol of exosome-treated monocytes contained both cholesterol ester and free cholesterol. Anti-phosphatidylserine receptor antibodies recognized surface protein on the monocyte plasma membrane and prevented exosome-induced cholesterol accumulation, indicating that exosome internalization is mediated via endogenous phosphatidylserine receptor. The production of proinflammatory cytokine TNF-alpha enhanced in parallel with monocyte cholesterol accumulation. Our data strongly indicate that exosomes released by activated T cells may represent a powerful, previously unknown, atherogenic factor.


Subject(s)
Cholesterol/metabolism , Monocytes/metabolism , Receptors, Cell Surface/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Adult , Antibodies , Atherosclerosis/metabolism , Female , Humans , Male , Middle Aged , Tumor Necrosis Factor-alpha/metabolism
14.
J Cell Physiol ; 205(3): 372-8, 2005 Dec.
Article in English | MEDLINE | ID: mdl-15895364

ABSTRACT

The endogenous Mg(2+)-inhibited cation (MIC) current was recently described in different cells of hematopoietic lineage and was implicated in the regulation of Mg2+ homeostasis. Here we present a single channel study of endogenously expressed Mg(2+)-dependent cation channels in the human myeloid leukemia K562 cells. Inwardly directed unitary currents were activated in cell-attached experiments in the absence of Ca2+ and Mg2+ in the pipette solution. The current-voltage (I-V) relationships displayed strong inward rectification and yielded a single channel slope conductance of approximately 30 pS at negative potentials. The I-V relationships were not altered by patch excision into divalent-free solution. Channel open probability (P(o)) and mean closed time constant (tau(C)) were strongly voltage-dependent, indicating that gating mechanisms may underlie current inward rectification. Millimolar concentrations of Ca2+ or Mg2+ applied to the cytoplasmic side of the membrane produced slow irreversible inhibition of channel activity. The Mg(2+)-dependent cation channels described in this study differ from the MIC channels described in human T-cells, Jurkat, and rat basophilic leukemia (RBL) cells in their I-V relationships, kinetic parameters and dependence on intracellular divalent cations. Our results suggested that endogenously expressed Mg(2+)-dependent cation channels in K562 cells and the MIC channels in other hematopoietic cells might be formed by different channel proteins.


Subject(s)
Cations/metabolism , Ion Channels/metabolism , Leukemia, Myeloid/metabolism , Magnesium/metabolism , Calcium/administration & dosage , Calcium/pharmacology , Calcium Channels/genetics , Cell Line, Tumor , Cell Membrane/drug effects , Cytoplasm/drug effects , Dose-Response Relationship, Drug , Electric Conductivity , Humans , Ion Channels/antagonists & inhibitors , Leukemia, Myeloid/pathology , Leukemia, Myeloid/physiopathology , Magnesium/administration & dosage , Magnesium/pharmacology , Patch-Clamp Techniques , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , TRPV Cation Channels/genetics
15.
J Biol Chem ; 280(16): 16377-82, 2005 Apr 22.
Article in English | MEDLINE | ID: mdl-15710604

ABSTRACT

Stimulation of T cell receptor in lymphocytes enhances Ca(2+) signaling and accelerates membrane trafficking. The relationships between these processes are not well understood. We employed membrane-impermeable lipid marker FM1-43 to explore membrane trafficking upon mobilization of intracellular Ca(2+) in Jurkat T cells. We established that liberation of intracellular Ca(2+) with T cell receptor agonist phytohemagglutinin P or with Ca(2+)-mobilizing agents ionomycin or thapsigargin induced accumulation of FM1-43 within the lumen of the endoplasmic reticulum (ER), nuclear envelope (NE), and Golgi. FM1-43 loading into ER-NE and Golgi was not mediated via the cytosol because other organelles such as mitochondria and multivesicular bodies located in close proximity to the FM1-43-containing ER were free of dye. Intralumenal FM1-43 accumulation was observed even when Ca(2+) signaling in the cytosol was abolished by the removal of extracellular Ca(2+). Our findings strongly suggest that release of intracellular Ca(2+) may create continuity between the extracellular leaflet of the plasma membrane and the lumenal membrane leaflet of the ER by a mechanism that does not require global cytosolic Ca(2+) elevation.


Subject(s)
Calcium/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Pyridinium Compounds/metabolism , Quaternary Ammonium Compounds/metabolism , T-Lymphocytes/metabolism , Cell Membrane/ultrastructure , Humans , Jurkat Cells , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/ultrastructure
16.
Exp Cell Res ; 291(1): 150-66, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14597416

ABSTRACT

FM1-43, a fluorescent styryl dye that penetrates into and stains membranes, was used to investigate kinetics of constitutive endocytosis and to visualize the fate of endocytic organelles in resting and activated human T lymphocytes. The rate of dye accumulation was strongly temperature dependent and approximately 10-fold higher in activated than in resting T cells. Elevation of cytosolic free Ca2+ concentration with thapsigargin or ionomycin further accelerated the rate of FM1-43 accumulation associated with cytosolic actin polymerization. Direct modulation of actin polymerization affected membrane trafficking. Actin condensation beneath the plasma membrane with calyculin A abolished FM1-43 internalization, whereas actin depolymerization with cytochalasin D had no effect. Photoconversion of DAB by FM1-43 revealed altered endocytic compartment targeting associated with T cell activation. Internalized cargo was carried to lysosome-like compartments in resting T cells and to multivesicular bodies (MVB) in activated T cells. Externalization of exosomes from MVB occurred commonly in activated but not in resting T cells. T cell exosomes contained raft-associated CD3 proteins, GM1 glycosphingolipids, and phosphatidylserine at the outer membrane leaflet. The present study demonstrates the utility of FM1-43 as a marker of membrane trafficking in T cells and reveals possible mechanisms of its modulation during T cell activation.


Subject(s)
Cell Compartmentation/physiology , Cell Membrane/metabolism , Endocytosis/physiology , T-Lymphocytes/metabolism , Transport Vesicles/metabolism , Actins/drug effects , Actins/metabolism , Calcium/metabolism , Cell Compartmentation/drug effects , Cell Membrane/ultrastructure , Cells, Cultured , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Lymphocyte Activation/drug effects , Lymphocyte Activation/physiology , Lysosomes/drug effects , Lysosomes/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Microscopy, Electron , Organelles/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Pyridinium Compounds/pharmacokinetics , Quaternary Ammonium Compounds/pharmacokinetics , T-Lymphocytes/ultrastructure , Temperature , Transport Vesicles/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...