Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 709: 149825, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38537599

ABSTRACT

SARS-Cov-2, the virus causing COVID-19, penetrates host target cells via the receptor of angiotensin-converting enzyme 2 (ACE2). Disrupting the virus interaction with ACE2 affords a plausible mechanism for prevention of cell penetration and inhibiting dissemination of the virus. Our studies demonstrate that ACE2 interaction with the receptor binding domain of SARS-Cov-2 spike protein (RBD) can be impaired by modulating the α7 nicotinic acetylcholine receptor (α7 nAChR) contiguous with ACE2. U373 cells of human astrocytoma origin were shown to bind both ACE2-specific antibody and recombinant RBD in Cell-ELISA. ACE2 was found to interact with α7 nAChR in U373 cell lysates studied by Sandwich ELISA. Our studies demonstrate that inhibition of RBD binding to ACE2-expressing U373 cells were defined with α7 nAChR agonists choline and PNU282987, but not a competitive antagonist methyllicaconitine (MLA). Additionally, the type 2 positive allosteric modulator (PAM2) PNU120596 and hydroxyurea (HU) also inhibited the binding. Our studies demonstrate that activation of α7 AChRs has efficacy in inhibiting the SARS-Cov-2 interaction with the ACE2 receptor and in such a way can prevent virus target cell penetration. These studies also help to clarify the consistent efficacy and positive outcomes for utilizing HU in treating COVID-19.


Subject(s)
Receptors, Nicotinic , alpha7 Nicotinic Acetylcholine Receptor , Humans , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Angiotensin-Converting Enzyme 2/metabolism , COVID-19 , Protein Binding , Receptors, Nicotinic/metabolism , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/chemistry
2.
J Neuroimmunol ; 385: 578244, 2023 12 15.
Article in English | MEDLINE | ID: mdl-38016403

ABSTRACT

In this paper the authors provide evidence that hydroxyurea (hydroxycarbamide) interacts with α7 nicotinic acetylcholine receptor, exerts anti-inflammatory and pro-survival effect, prevents α7 nicotinic receptor interaction with angiotensin-converting enzyme-2 and stimulates IgM to IgG class switch upon immunization with SARS spike protein fragment 674-685. Hydroxyurea shifts immunoglobulin glycosylation profile to anti-inflammatory phenotype and prevents the appearance of anti-idiotypic α7(179-190)-specific antibodies, as well as memory impairment. According to these results, interaction with α7 nicotinic acetylcholine receptor may underlie positive therapeutic effects of hydroxyurea upon SARS-Cov-2 infection by interfering with virus penetration into the cell and providing anti-inflammatory and immunomodulatory effects.


Subject(s)
COVID-19 , Receptors, Nicotinic , Humans , alpha7 Nicotinic Acetylcholine Receptor/genetics , Hydroxyurea/pharmacology , Hydroxyurea/therapeutic use , SARS-CoV-2/metabolism , Anti-Inflammatory Agents/therapeutic use
3.
Biochem Biophys Res Commun ; 657: 35-42, 2023 05 21.
Article in English | MEDLINE | ID: mdl-36972659

ABSTRACT

Nicotinic acetylcholine receptors (nAChRs), initially characterized as ligand-gated ion channels mediating fast synaptic transmission, are now found in many non-excitable cells and mitochondria where they function in ion-independent manner and regulate vital cellular processes like apoptosis, proliferation, cytokine secretion. Here we show that the nAChRs of α7 subtype are present in the nuclei of liver cells and astrocytoma U373 cell line. As shown by lectin ELISA, the nuclear α7 nAChRs are mature glycoproteins that follow the standard rout of post-translational modifications in Golgi; however, their glycosylation profile is non-identical to that of mitochondrial nAChRs. They are exposed on the outer nuclear membrane and are found in combination with lamin B1. The nuclear α7 nAChRs are up-regulated in liver within 1 h after partial hepatectomy and in H2O2-treated U373 cells. As shown both in silico and experimentally, the α7 nAChR interacts with hypoxia-inducible factor HIF-1α and this interaction is impaired by α7-selective agonists PNU282987 and choline or type 2 positive allosteric modulator PNU120596, which prevent HIF-1α accumulation in the nuclei. Similarly, HIF-1α interacts with mitochondrial α7 nAChRs in U373 cells treated with dimethyloxalylglycine. It is concluded that functional α7 nAChRs influence HIF-1α translocation into the nucleus and mitochondria upon hypoxia.


Subject(s)
Cell Nucleus , Hypoxia-Inducible Factor 1 , Hypoxia , Mitochondria , alpha7 Nicotinic Acetylcholine Receptor , Humans , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Cell Nucleus/metabolism , Hydrogen Peroxide/metabolism , Hypoxia/metabolism , Mitochondria/metabolism , Hypoxia-Inducible Factor 1/metabolism
4.
Biochem Biophys Res Commun ; 622: 57-63, 2022 09 24.
Article in English | MEDLINE | ID: mdl-35843095

ABSTRACT

COVID-19 is accompanied by strong inflammatory reaction and is often followed by long-term cognitive disorders. The fragment 674-685 of SARS-Cov-2 spike protein was shown to interact with α7 nicotinic acetylcholine receptor involved in regulating both inflammatory reactions and cognitive functions. Here we show that mice immunized with the peptide corresponding to 674-685 fragment of SARS-Cov-2 spike protein conjugated to hemocyanin (KLH-674-685) demonstrate decreased level of α7 nicotinic acetylcholine receptors, increased levels of IL-1ß and TNFα in the brain and impairment of episodic memory. Choline injections prevented α7 nicotinic receptor decline and memory loss. Mice injected with immunoglobulins obtained from the blood of (KLH-674-685)-immunized mice also demonstrated episodic memory decline. These data allow suggesting that post-COVID memory impairment in humans is related to SARS-Cov-2 spike protein-specific immune reaction. The mechanisms of such effect are being discussed.


Subject(s)
COVID-19 , Memory, Episodic , Animals , Humans , Immunization , Inflammation , Memory Disorders/etiology , Memory Disorders/metabolism , Mice , Neuroinflammatory Diseases , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/adverse effects , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism
5.
Regen Med ; 17(8): 533-546, 2022 08.
Article in English | MEDLINE | ID: mdl-35638401

ABSTRACT

Background: The present research has been undertaken to study the therapeutic potential of mesenchymal stem cells (MSCs) for the treatment of neuroinflammation-induced cognitive disorders. Methods: Either umbilical cord or adipose MSCs were injected into mice treated with lipopolysaccharide. The mice were studied in behavioral tests, and their brains were examined by means of immunohistochemistry, electron microscopy and sandwich ELISA. Results: MSCs, introduced either intravenously or intraperitoneally, restored episodic memory of mice disturbed by inflammation, normalized nAChR and Aß1-42 levels and stimulated proliferation of neural progenitor cells in the brain. The effect of MSCs was observed for months, whereas that of MSC-conditioned medium was transient and stimulated an immune reaction. SDF-1α potentiated the effects of MSCs on the brain and memory. Conclusion: MSCs of different origins provide a long-term therapeutic effect in the treatment of neuroinflammation-induced episodic memory impairment.


Subject(s)
Cognitive Dysfunction , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Cognitive Dysfunction/therapy , Mice , Neuroinflammatory Diseases , Umbilical Cord
6.
Biochem Biophys Res Commun ; 561: 14-18, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34000512

ABSTRACT

In spite of numerous studies, many details of SARS-Cov-2 interaction with human cells are still poorly understood. The 674-685 fragment of SARS-Cov-2 spike protein is homologous to the fragment of α-cobratoxin underlying its interaction with α7 nicotinic acetylcholine receptors (nAChRs). The interaction of 674-685 peptide with α7 nAChR has been predicted in silico. In the present paper we confirm this prediction experimentally and investigate the effect of SARS-Cov-2 spike protein peptide on mitochondria, which express α7 nAChRs to regulate apoptosis-related events. We demonstrate that SARS-Cov-2 spike protein peptide 674-685 competes with the antibody against 179-190 fragment of α7 nAChR subunit for the binding to α7-expressing cells and mitochondria and prevents the release of cytochrome c from isolated mitochondria in response to 0.5 mM H2O2 but does not protect intact U373 cells against apoptogenic effect of H2O2. Our data suggest that the α7 nAChR-binding portion of SARS-Cov-2 spike protein prevents mitochondria-driven apoptosis when the virus is uncoated inside the cell and, therefore, supports the infected cell viability before the virus replication cycle is complete.


Subject(s)
Apoptosis , Cytochromes c/metabolism , Mitochondria/metabolism , Peptide Fragments/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Computer Simulation , Female , Hydrogen Peroxide/pharmacology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , SARS-CoV-2/drug effects , SARS-CoV-2/growth & development , alpha7 Nicotinic Acetylcholine Receptor/metabolism
7.
Int J Biochem Cell Biol ; 129: 105879, 2020 12.
Article in English | MEDLINE | ID: mdl-33147521

ABSTRACT

Nicotinic acetylcholine receptors (nAChRs) mediate fast synaptic transmission in muscles and autonomic ganglia and regulate cytokine and neurotransmitter release in the brain and non-excitable cells. The α7 nAChRs localized in the outer membrane of mitochondria regulate cytochrome c release stimulated by apoptosis-inducing agents. However, the mechanisms through which nAChRs influence mitochondrial permeability remain obscure. Here we put an aim to explore the interaction of nAChRs with voltage-dependent anion channels (VDAC1) and pro-apoptotic protein Bax in the course of apoptosis induction. By using molecular modeling in silico, it was shown that both Bax and VDAC1 can bind within the 4th transmembrane portion (M4) of nAChR subunits. Experimentally, α7 nAChR-Bax and α7 nAChR-VDAC1 complexes were identified by sandwich ELISA in mitochondria isolated from astrocytoma U373 cells. Stimulating apoptosis of U373 cells by H2O2 disrupted α7-VDAC complexes and favored formation of α7-Bax complexes accompanied by cytochrome c release from mitochondria. α7-selective agonist PNU282987 or type 2 positive allosteric modulator PNU120596 disrupted α7-Bax and returned α7 nAChR to complex with VDAC1 resulting in attenuation of cytochrome c release. It is concluded that mitochondrial nAChRs regulate apoptosis-induced mitochondrial channel formation by modulating the interplay of apoptosis-related proteins in mitochondria outer membrane.


Subject(s)
Apoptosis , Mitochondria/metabolism , Voltage-Dependent Anion Channel 1/metabolism , bcl-2-Associated X Protein/metabolism , Allosteric Regulation , Animals , Cell Death , Cell Line, Tumor , Humans , alpha7 Nicotinic Acetylcholine Receptor
9.
Int Immunopharmacol ; 82: 106373, 2020 Mar 09.
Article in English | MEDLINE | ID: mdl-32163855

ABSTRACT

This study demonstrates the presence of α7 nicotinic acetylcholine receptors (nAChR) in B lymphocyte-derived SP-2/0 cells by means of flow cytometry and immunocytochemistry. According to lectin and sandwich ELISA, the α7 subunits expressed in SP-2/0 cells are more glycosylated compared to those expressed in the brain or normal B lymphocytes and are combined with ß2 subunits. At zero and negative pipette potentials, either acetylcholine or α7-specific agonist PNU282987 stimulated the ion channel activity in SP-2/0 cells revealed by single channel patch-clamp recordings. The conductivity was within the range of 19 to 39 pS and reversal potential was between -17 mV and +28 mV, the currents were potentiated by α7-specific positive allosteric modulator PNU120596 and were partially blocked by α7-specific antagonist methyllicaconitine (MLA). However, they were oriented downwards suggesting that the channels mediated the cation outflux rather than influx. As shown by Ca2+ imaging studies, PNU282987 did not stimulate immediate Ca2+ influx into SP-2/0 cells. Instead, Ca2+ influx through Ca-release-activated channels (CRACs) was observed within minutes after either PNU282987 or MLA application. It is concluded that SP-2/0 express α7ß2 nAChRs, which mediate the cation outflux under negative pipette potentials applied, possibly, due to depolarized membrane or negative surface charge formed by carbohydrate residues. In addition, α7ß2 nAChRs may influence CRACs in ion-independent way.

10.
Biomolecules ; 10(2)2020 02 03.
Article in English | MEDLINE | ID: mdl-32028688

ABSTRACT

Nicotinic acetylcholine receptors of α7 subtype (α7 nAChRs) are involved in regulating neuroinflammation and cognitive functions. Correspondingly, α7-/- mice demonstrate pro-inflammatory phenotype and impaired episodic memory. In addition, nAChRs expressed in mitochondria regulate the release of pro-apoptotic factors like cytochrome c. Here we studied whether the cognitive deficiency of α7-/- mice can be cured by oral consumption of either nicotine or N-stearoylethanolamine (NSE), a lipid possessing anti-inflammatory, cannabimimetic and membrane-stabilizing activity. Mice were examined in Novel Object Recognition behavioral test, their blood, brains and brain mitochondria were tested for the levels of interleukin-6, various nAChR subtypes and cytochrome c released by ELISA. The data presented demonstrate that both substances stimulated the raise of interleukin-6 in the blood and improved episodic memory of α7-/- mice. However, NSE improved, while nicotine worsened the brain mitochondria sustainability to apoptogenic stimuli, as shown by either decreased or increased amounts of cytochrome c released. Both nicotine and NSE up-regulated α4ß2 nAChRs in the brain; NSE up-regulated, while nicotine down-regulated α9-containing nAChRs in the brain mitochondria. It is concluded that the level of alternative nAChR subtypes in the brain is critically important for memory and mitochondria sustainability in the absence of α7 nAChRs.


Subject(s)
Brain/drug effects , Ethanolamines/pharmacology , Memory, Episodic , Mitochondria/drug effects , Nicotine/pharmacology , Stearic Acids/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/genetics , Animals , Behavior, Animal , Brain/metabolism , Cytochromes c/metabolism , Enzyme-Linked Immunosorbent Assay , Interleukin-6/metabolism , Lipids/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism
11.
Neuroscience ; 413: 31-44, 2019 08 10.
Article in English | MEDLINE | ID: mdl-31202708

ABSTRACT

Nicotinic acetylcholine receptors of α7 subtype (α7 nAChRs) are involved in regulating cognition, inflammation and cell survival. Neuroinflammation is accompanied by the decrease of α7 nAChRs in the brain and impairment of memory. We show here that α7-/- mice possess pro-inflammatory phenotype and demonstrate worse episodic memory compared to wild-type mice. Previously we reported that mesenchymal stem cells (MSCs) restored episodic memory of lipopolysaccharide-treated wild-type mice. The aim of this study was to examine if MSCs or their soluble factors improve memory of α7-/- mice. The α7-specific signal (ELISA) and α7+ cells (IHC) were found in the brain of α7-/- mice on days 7 and 14 after intravenous injection of α7+ MSCs from either human umbilical cord (hMSCs) or mouse placenta (mMSCs). The intravenously injected MSCs or intraperitoneally injected hMSCs-conditioned medium transiently improved episodic memory of α7-/- mice and decreased cytochrome c release from their brain mitochondria under the effect of Ca2+. Either MSCs or conditioned medium stimulated an IL-6 increase in the brain, which coincided with the improvement of episodic memory. Injections of recombinant IL-6 also improved episodic memory of α7-/- mice accompanied by the up-regulation of α3, α4, ß2 and ß4 nAChR subunits in the brain. It is concluded that MSCs, injected intravenously, penetrate the brain of α7-/- mice and persist there for at least 2 weeks. They improve episodic memory of mice and make their mitochondria more resistant to apoptogenic influence. One of the soluble factors responsible for the memory improvement is IL-6.


Subject(s)
Interleukin-6/pharmacology , Memory Disorders/therapy , Memory, Episodic , Mesenchymal Stem Cell Transplantation , Nootropic Agents/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/deficiency , Animals , Brain/metabolism , Female , Humans , Memory Disorders/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics
12.
Front Aging Neurosci ; 11: 359, 2019.
Article in English | MEDLINE | ID: mdl-31998114

ABSTRACT

Neuroinflammation accompanies or even precedes the development of cognitive changes in many brain pathologies, including Alzheimer's disease. Therefore, dampening inflammatory reactions within the brain is a promising strategy for supporting cognitive functions in elderly people and for preventing the development of neurodegenerative disorders. Nicotinic acetylcholine receptors containing α7 subunits (α7 nAChRs) are involved in regulating cell survival, inflammation, and memory. The aim of our study was to evaluate the efficiency of α7-specific therapy at different stages of inflammation and to compare the effects of orthosteric agonist PNU282987 and type 2 positive allosteric modulator (PAM) PNU120596 in mice after a single injection of lipopolysaccharide (LPS). The data presented demonstrate that PNU282987 protected mice from LPS-induced impairment of episodic memory by decreasing IL-6 levels in the blood, stabilizing the brain mitochondria and up-regulating the brain α7-, α3-, and α4-containing nAChRs. Such treatment was efficient when given simultaneously with LPS or a week after LPS injection and was not efficient if LPS had been injected 2 months before. PNU120596 also decreased IL-6, stabilized mitochondria and up-regulated the brain nAChRs. However, its memory-improving effect was transient and disappeared after the end of the injection cycle. Moreover, cessation of PNU120596 treatment resulted in a sharp increase in IL-1ß and IL-6 levels in the blood. It is concluded that activating α7 nAChRs protects the mouse brain from the pathogenic effect of LPS in the early stages of inflammation but is not efficient when irreversible changes have already occurred. The use of a PAM does not improve the effect of the agonist, possibly potentiates the effect of endogenous agonists, and results in undesirable effects after treatment cessation.

14.
J Neuroimmunol ; 318: 8-14, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29395323

ABSTRACT

This study demonstrates that α7 nicotinic acetylcholine receptors (nAChRs) regulate mouse B lymphocyte proliferation and IgM production in ion-independent manner. The high α7 nAChR levels were found in CD5+ and Foxp3+ B cells; induction of Foxp3+ cells in vitro was attenuated in the absence or upon inhibition of α7 nAChRs. The adoptively transferred B lymphocytes, stimulated in presence of methyllicaconitine, decreased the IgM response and abolished the IgG response in the host. The data obtained demonstrate the importance of cholinergic regulation for the antibody immune response and immunosuppression.


Subject(s)
Antibody Formation/immunology , B-Lymphocytes, Regulatory/immunology , Lymphocyte Activation/immunology , alpha7 Nicotinic Acetylcholine Receptor/immunology , Animals , Cell Proliferation/physiology , Female , Immunoglobulin Class Switching/immunology , Mice , Mice, Inbred C57BL
15.
Front Mol Neurosci ; 9: 19, 2016.
Article in English | MEDLINE | ID: mdl-27013966

ABSTRACT

Neuro-inflammation, one of the pathogenic causes of neurodegenerative diseases, is regulated through the cholinergic anti-inflammatory pathway via the α7 nicotinic acetylcholine receptor (α7 nAChR). We previously showed that either bacterial lipopolysaccharide (LPS) or immunization with the α7(1-208) nAChR fragment decrease α7 nAChRs density in the mouse brain, exacerbating chronic inflammation, beta-amyloid accumulation and episodic memory decline, which mimic the early stages of Alzheimer's disease (AD). To study the molecular mechanisms underlying the LPS and antibody effects in the brain, we employed an in vivo model of acute LPS-induced inflammation and an in vitro model of cultured glioblastoma U373 cells. Here, we report that LPS challenge decreased the levels of α7 nAChR RNA and protein and of acetylcholinesterase (AChE) RNA and activity in distinct mouse brain regions, sensitized brain mitochondria to the apoptogenic effect of Ca(2+) and modified brain microRNA profiles, including the cholinergic-regulatory CholinomiRs-132/212, in favor of anti-inflammatory and pro-apoptotic ones. Adding α7(1-208)-specific antibodies to the LPS challenge prevented elevation of both the anti-inflammatory and pro-apoptotic miRNAs while supporting the resistance of brain mitochondria to Ca(2+) and maintaining α7 nAChR/AChE decreases. In U373 cells, α7-specific antibodies and LPS both stimulated interleukin-6 production through the p38/Src-dependent pathway. Our findings demonstrate that acute LPS-induced inflammation induces the cholinergic anti-inflammatory pathway in the brain, that α7 nAChR down-regulation limits this pathway, and that α7-specific antibodies aggravate neuroinflammation by inducing the pro-inflammatory interleukin-6 and dampening anti-inflammatory miRNAs; however, these antibodies may protect brain mitochondria and decrease the levels of pro-apoptotic miRNAs, preventing LPS-induced neurodegeneration.

16.
Int Immunopharmacol ; 23(2): 475-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25281899

ABSTRACT

α7 Nicotinic acetylcholine receptors (α7 nAChRs) are involved in regulating inflammatory cytokine production in macrophages and astrocytes. In the present paper, it is shown that α7-specific agonists PNU282987 (130nM) or choline (1.6mM) attenuated the interleukin-6 (IL-6) production stimulated by bacterial lipopolysaccharide in monocyte-derived U937 and astrocyte-derived U373 cell lines. In contrast, α7(179-190)-specific antibody, which bound to and was internalized by U373 cells, stimulated IL-6 production in p38 kinase-dependent manner in the absence of lipopolysaccharide. The antibody effect was not due to its Fc-fragment because similar capacity was found for recombinant single-chain (scFv) α7(179-190)-specific antibody selected from the gene library of healthy human subject. The data obtained allow suggesting that α7-specific antibody can provoke neuroinflammation within the brain by inducing IL-6 production in astrocytes.


Subject(s)
Antibodies, Monoclonal/pharmacology , Astrocytes/drug effects , Interleukin-6/metabolism , alpha7 Nicotinic Acetylcholine Receptor/immunology , p38 Mitogen-Activated Protein Kinases/metabolism , Antibodies, Monoclonal/immunology , Astrocytes/metabolism , Cell Line , Humans , Interleukin-6/genetics , Macrophages/drug effects , Macrophages/metabolism , Protein Subunits , alpha7 Nicotinic Acetylcholine Receptor/chemistry
17.
Mol Immunol ; 54(3-4): 319-26, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23352962

ABSTRACT

Receptor-specific antibodies can both prevent ligand-receptor interaction and initiate receptor signaling. Previously we generated monoclonal antibody 8E8 (mAb 8E8) against protease-activated receptor type 3 (PAR3) which inhibited proliferation of B cell hybridoma. Here we used mAb 8E8 and PAR1-specific polyclonal antibody to reveal the functions and cooperating partners of PAR3 in endothelial cells and in B lymphocytes. MAb 8E8 or PAR1 agonist peptide stimulated IL-6 and IL-8 production and VCAM-1 expression in HPMEC-ST1.6R cells. PAR1 antibody stimulated only VCAM-1 expression, while ICAM-1 expression was stimulated with mAB 8E8 or PAR3 peptide. MAb 8E8 stimulated weak mitogenic response, while PAR1 antibody inhibited it in normal but not in malignant B lymphocytes. Sandwich ELISA assay demonstrated the interaction of PAR3 with PAR1 in malignant cell lines and with IgM in normal B lymphocytes. It is concluded that PAR3 cooperates with PAR1 to mediate the effect of thrombin on cytokine production and VCAM-1 expression in endothelial cells and on cell proliferation in malignant B cells. ICAM-1 expression in endothelial cells requires PAR3 without PAR1. The inhibitory effect of thrombin in normal B lymphocytes is mediated by PAR1 alone, while mitogenic and pro-survival signaling in B lymphocytes is provided through PAR3 in cooperation with BCR.


Subject(s)
Antibodies, Monoclonal/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Receptors, Thrombin/immunology , Receptors, Thrombin/metabolism , Animals , Antibody Specificity , Cell Line , Cell Proliferation , Humans , Interleukin-6/immunology , Interleukin-6/metabolism , Interleukin-8/immunology , Interleukin-8/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptor, PAR-1/immunology , Receptor, PAR-1/metabolism , Receptors, Thrombin/biosynthesis , Thrombin/immunology , Thrombin/metabolism , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism
18.
PLoS One ; 7(2): e31361, 2012.
Article in English | MEDLINE | ID: mdl-22359587

ABSTRACT

Nicotinic acetylcholine receptors (nAChRs) are ligand-gated ion channels that mediate synaptic transmission in the muscle and autonomic ganglia and regulate transmitter release in the brain. The nAChRs composed of α7 subunits are also expressed in non-excitable cells to regulate cell survival and proliferation. Up to now, functional α7 nAChRs were found exclusively on the cell plasma membrane. Here we show that they are expressed in mitochondria and regulate early pro-apoptotic events like cytochrome c release. The binding of α7-specific antibody with mouse liver mitochondria was revealed by electron microscopy. Outer membranes of mitochondria from the wild-type and ß2-/- but not α7-/- mice bound α7 nAChR-specific antibody and toxins: FITC-labeled α-cobratoxin or Alexa 555-labeled α-bungarotoxin. α7 nAChR agonists (1 µM acetylcholine, 10 µM choline or 30 nM PNU-282987) impaired intramitochondrial Ca(2+) accumulation and significantly decreased cytochrome c release stimulated with either 90 µM CaCl(2) or 0.5 mM H(2)O(2). α7-specific antagonist methyllicaconitine (50 nM) did not affect Ca(2+) accumulation in mitochondria but attenuated the effects of agonists on cytochrome c release. Inhibitor of voltage-dependent anion channel (VDAC) 4,4'-diisothio-cyano-2,2'-stilbene disulfonic acid (0.5 µM) decreased cytochrome c release stimulated with apoptogens similarly to α7 nAChR agonists, and VDAC was co-captured with the α7 nAChR from mitochondria outer membrane preparation in both direct and reverse sandwich ELISA. It is concluded that α7 nAChRs are expressed in mitochondria outer membrane to regulate the VDAC-mediated Ca(2+) transport and mitochondrial permeability transition.


Subject(s)
Calcium/metabolism , Cytochromes c/metabolism , Mitochondria/metabolism , Receptors, Nicotinic/biosynthesis , Animals , Mice , Mice, Knockout , Mitochondrial Membranes , Permeability , Receptors, Nicotinic/physiology , Voltage-Dependent Anion Channels , alpha7 Nicotinic Acetylcholine Receptor
19.
J Alzheimers Dis ; 25(4): 747-61, 2011.
Article in English | MEDLINE | ID: mdl-21593571

ABSTRACT

Alzheimer's disease (AD) is characterized by a loss of α4ß2 and α7 nicotinic acetylcholine receptors (nAChRs) in the brain and severe memory impairments. Previously, we found that antibodies elicited against extracellular domain of α7 nAChR subunit decreased the number of α7 nAChRs in the brains of mice and impaired episodic memory. Here we show that antibodies capable to bind α7(1-208) are present in the blood of both healthy humans and AD patients. In healthy individuals, their capacity to compete with [(125)-I]-α-bungarotoxin for the binding to α7(1-208) increased with age. The level of such antibodies was significantly elevated in children with severe form of obstructive bronchitis and in mice injected with Lewis lung carcinoma cells expressing both α4ß2 and α7 nAChRs. Elevated antibody levels were accompanied with decreased surface nAChRs on the blood lymphocytes of children and of mice immunized with α7(1-208). Among AD patients, the level of α7 nAChR-specific antibodies was significantly larger in people 62.5 ± 1.5 years old with moderate or severe AD stages (15.2 ± 1.3 MMSE scores) compared to those of 76 ± 1.5 years old with the mild (22.7 ± 0.1 MMSE scores) AD stage. We concluded that α7(1-208) nAChR-specific antibodies found in the human blood are formed as a result of common infections accompanied with the destruction of respiratory epithelium. Elevated blood plasma levels of α7(1-208) nAChR-specific antibodies are characteristic for the early-onset AD and, therefore, are suggested as one of the risk factors for the development of this form of the disease.


Subject(s)
Alzheimer Disease/immunology , Autoantibodies/blood , Receptors, Nicotinic/immunology , Aged , Aging/metabolism , Animals , Antibody Specificity , Brain/metabolism , Bronchitis/immunology , Bungarotoxins/metabolism , Carcinoma, Lewis Lung/metabolism , Cell Line, Tumor , Cell Transplantation , Child , Enzyme-Linked Immunosorbent Assay , Epitopes , Female , Flow Cytometry , Humans , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Radioimmunoassay , Receptors, Cell Surface/metabolism , Recombinant Proteins/immunology , alpha7 Nicotinic Acetylcholine Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...