Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
2.
J Physiol ; 594(24): 7229-7248, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27570189

ABSTRACT

KEY POINTS: Biotin, a vitamin whose main role is as a coenzyme for carboxylases, accumulates at unusually large amounts within cells of the carotid body (CB). In biotin-deficient rats biotin rapidly disappears from the blood; however, it remains at relatively high levels in CB glomus cells. The CB contains high levels of mRNA for SLC5a6, a biotin transporter, and SLC19a3, a thiamine transporter regulated by biotin. Animals with biotin deficiency exhibit pronounced metabolic lactic acidosis. Remarkably, glomus cells from these animals have normal electrical and neurochemical properties. However, they show a marked decrease in the size of quantal dopaminergic secretory events. Inhibitors of the vesicular monoamine transporter 2 (VMAT2) mimic the effect of biotin deficiency. In biotin-deficient animals, VMAT2 protein expression decreases in parallel with biotin depletion in CB cells. These data suggest that dopamine transport and/or storage in small secretory granules in glomus cells depend on biotin. ABSTRACT: Biotin is a water-soluble vitamin required for the function of carboxylases as well as for the regulation of gene expression. Here, we report that biotin accumulates in unusually large amounts in cells of arterial chemoreceptors, carotid body (CB) and adrenal medulla (AM). We show in a biotin-deficient rat model that the vitamin rapidly disappears from the blood and other tissues (including the AM), while remaining at relatively high levels in the CB. We have also observed that, in comparison with other peripheral neural tissues, CB cells contain high levels of SLC5a6, a biotin transporter, and SLC19a3, a thiamine transporter regulated by biotin. Biotin-deficient rats show a syndrome characterized by marked weight loss, metabolic lactic acidosis, aciduria and accelerated breathing with normal responsiveness to hypoxia. Remarkably, CB cells from biotin-deficient animals have normal electrophysiological and neurochemical (ATP levels and catecholamine synthesis) properties; however, they exhibit a marked decrease in the size of quantal catecholaminergic secretory events, which is not seen in AM cells. A similar differential secretory dysfunction is observed in CB cells treated with tetrabenazine, a selective inhibitor of the vesicular monoamine transporter 2 (VMAT2). VMAT2 is highly expressed in glomus cells (in comparison with VMAT1), and in biotin-deficient animals VMAT2 protein expression decreases in parallel with the decrease of biotin accumulated in CB cells. These data suggest that biotin has an essential role in the homeostasis of dopaminergic transmission modulating the transport and/or storage of transmitters within small secretory granules in glomus cells.


Subject(s)
Biotin/metabolism , Carotid Body/metabolism , Dopamine/metabolism , Adenosine Triphosphate/metabolism , Adrenal Medulla/metabolism , Animals , Arteries/metabolism , Biotin/blood , Biotin/deficiency , Chromaffin Cells/metabolism , Exocytosis , Hypoxia/physiopathology , Lactic Acid/blood , Rats, Wistar , Superior Cervical Ganglion/metabolism , Tetrabenazine/pharmacology , Vesicular Monoamine Transport Proteins/antagonists & inhibitors , Vesicular Monoamine Transport Proteins/metabolism
3.
Br J Haematol ; 173(6): 884-95, 2016 06.
Article in English | MEDLINE | ID: mdl-27071340

ABSTRACT

While proteasome inhibition is a validated therapeutic approach for multiple myeloma (MM), inhibition of individual constitutive proteasome (c20S) and immunoproteasome (i20S) subunits has not been fully explored owing to a lack of effective tools. We utilized the novel proteasome constitutive/immunoproteasome subunit enzyme-linked immunosorbent (ProCISE) assay to quantify proteasome subunit occupancy in samples from five phase I/II and II trials before and after treatment with the proteasome inhibitor carfilzomib. Following the first carfilzomib dose (15-56 mg/m(2) ), dose-dependent inhibition of c20S and i20S chymotrypsin-like active sites was observed [whole blood: ≥67%; peripheral blood mononuclear cells (PBMCs): ≥75%]. A similar inhibition profile was observed in bone marrow-derived CD138(+) tumour cells. Carfilzomib-induced proteasome inhibition was durable, with minimal recovery in PBMCs after 24 h but near-complete recovery between cycles. Importantly, the ProCISE assay can be used to quantify occupancy of individual c20S and i20S subunits. We observed a relationship between MM patient response (n = 29), carfilzomib dose and occupancy of multiple i20S subunits, where greater occupancy was associated with an increased likelihood of achieving a clinical response at higher doses. ProCISE represents a new tool for measuring proteasome inhibitor activity in clinical trials and relating drug action to patient outcomes.


Subject(s)
Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/drug effects , Antineoplastic Agents/therapeutic use , Bone Marrow/pathology , Dose-Response Relationship, Drug , Humans , Multiple Myeloma/drug therapy , Oligopeptides/therapeutic use , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Remission Induction , Tumor Cells, Cultured
4.
Sci Rep ; 3: 2937, 2013 Oct 14.
Article in English | MEDLINE | ID: mdl-24121824

ABSTRACT

X-gal staining is a common procedure used in the histochemical monitoring of gene expression by light microscopy. However, this procedure does not permit the direct confocal acquisition of images, thus preventing the identification of labelled cells on the depth (Z) axis of tissue sections and leading sometimes to erroneous conclusions in co-localization and gene expression studies. Here we report a technique, based on X-gal fluorescence emission and mathematically-based optical correction, to obtain high quality fluorescence confocal images. This method, combined with immunofluorescence, makes it possible to unequivocally identify X-gal-labelled cells in tissue sections, emerging as a valuable tool in gene expression and cell tracing analysis.


Subject(s)
Galactosides/metabolism , Indoles/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , beta-Galactosidase/metabolism , Animals , Fluorometry , Gene Expression , Genes, Reporter , Histocytological Preparation Techniques , Mice , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , beta-Galactosidase/genetics
5.
J Physiol ; 591(24): 6157-73, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24167224

ABSTRACT

The carotid body (CB) is the major peripheral arterial chemoreceptor in mammals that mediates the acute hyperventilatory response to hypoxia. The CB grows in response to sustained hypoxia and also participates in acclimatisation to chronic hypoxaemia. Knowledge of CB physiology at the cellular level has increased considerably in recent times thanks to studies performed on lower mammals, and rodents in particular. However, the functional characteristics of human CB cells remain practically unknown. Herein, we use tissue slices or enzymatically dispersed cells to determine the characteristics of human CB cells. The adult human CB parenchyma contains clusters of chemosensitive glomus (type I) and sustentacular (type II) cells as well as nestin-positive progenitor cells. This organ also expresses high levels of the dopaminotrophic glial cell line-derived neurotrophic factor (GDNF). We found that GDNF production and the number of progenitor and glomus cells were preserved in the CBs of human subjects of advanced age. Moreover, glomus cells exhibited voltage-dependent Na(+), Ca(2+) and K(+) currents that were qualitatively similar to those reported in lower mammals. These cells responded to hypoxia with an external Ca(2+)-dependent increase of cytosolic Ca(2+) and quantal catecholamine secretion, as reported for other mammalian species. Interestingly, human glomus cells are also responsive to hypoglycaemia and together these two stimuli can potentiate each other's effects. The chemosensory responses of glomus cells are also preserved at an advanced age. These new data on the cellular and molecular physiology of the CB pave the way for future pathophysiological studies involving this organ in humans.


Subject(s)
Action Potentials , Carotid Body/cytology , Adolescent , Adult , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Aged , Calcium/metabolism , Calcium Signaling , Carotid Body/metabolism , Carotid Body/physiology , Cell Hypoxia , Cells, Cultured , Child , Female , Glucose/metabolism , Humans , Ion Channels/metabolism , Male , Middle Aged , Nestin/genetics , Nestin/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Potassium/metabolism , Sodium/metabolism
6.
Am J Physiol Cell Physiol ; 302(3): C494-504, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22049210

ABSTRACT

Ion channels participate in cell homeostasis and are involved in the regulation of proliferation and differentiation in several cell types; however, their presence and function in embryonic stem (ES) cells are poorly studied. We have investigated the existence of voltage-dependent inward currents in mouse ES cells and their ability to modulate proliferation and self-renewal. Patch-clamped ES cells had inactivating tetrodotoxin (TTX)-sensitive Na(+) currents as well as transient Ca(2+) currents abolished by the external application of Ni(2+). Biophysical and pharmacological data indicated that the Ca(2+) current is predominantly mediated by T-type (Ca(v)3.2) channels. The number of cells expressing T-type channels and Ca(v)3.2 mRNA levels increased at the G1/S transition of the cell cycle. TTX had no effect on ES cell proliferation. However, blockade of T-type Ca(2+) currents with Ni(2+) induced a decrease in proliferation and alkaline phosphatase positive colonies as well as reduced expression of Oct3/4 and Nanog, all indicative of loss in self-renewal capacity. Decreased alkaline phosphatase and Oct3/4 expression were also observed in cells subjected to small interfering RNA-induced knockdown for T-type (Ca(v)3.2) Ca(2+) channels, thus partially recapitulating the pharmacological effects on self-renewal. These results indicate that Ca(v)3.2 channel expression in ES cells is modulated along the cell cycle being induced at late G1 phase. They also suggest that these channels are involved in the maintenance of the undifferentiated state of mouse ES cells. We propose that Ca(2+) entry mediated by Ca(v)3.2 channels might be one of the intracellular signals that participate in the complex network responsible for ES cell self-renewal.


Subject(s)
Calcium Channels, T-Type/metabolism , Cell Cycle/physiology , Embryonic Stem Cells/metabolism , Alkaline Phosphatase/biosynthesis , Animals , Calcium/metabolism , Calcium Channels, T-Type/biosynthesis , Calcium Channels, T-Type/genetics , Cell Cycle/drug effects , Cell Differentiation , Cell Line , Cell Proliferation/drug effects , Homeodomain Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Nanog Homeobox Protein , Nickel/pharmacology , Octamer Transcription Factor-3/biosynthesis , Patch-Clamp Techniques , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Voltage-Dependent Anion Channels
7.
J Gen Physiol ; 135(4): 379-92, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20351062

ABSTRACT

Background K(+) channels of the TASK family are believed to participate in sensory transduction by chemoreceptor (glomus) cells of the carotid body (CB). However, studies on the systemic CB-mediated ventilatory response to hypoxia and hypercapnia in TASK1- and/or TASK3-deficient mice have yielded conflicting results. We have characterized the glomus cell phenotype of TASK-null mice and studied the responses of individual cells to hypoxia and other chemical stimuli. CB morphology and glomus cell size were normal in wild-type as well as in TASK1(-/-) or double TASK1/3(-/-) mice. Patch-clamped TASK1/3-null glomus cells had significantly higher membrane resistance and less hyperpolarized resting potential than their wild-type counterpart. These electrical parameters were practically normal in TASK1(-/-) cells. Sensitivity of background currents to changes of extracellular pH was drastically diminished in TASK1/3-null cells. In contrast with these observations, responsiveness to hypoxia or hypercapnia of either TASK1(-/-) or double TASK1/3(-/-) cells, as estimated by the amperometric measurement of catecholamine release, was apparently normal. TASK1/3 knockout cells showed an enhanced secretory rate in basal (normoxic) conditions compatible with their increased excitability. Responsiveness to hypoxia of TASK1/3-null cells was maintained after pharmacological blockade of maxi-K(+) channels. These data in the TASK-null mouse model indicate that TASK3 channels contribute to the background K(+) current in glomus cells and to their sensitivity to external pH. They also suggest that, although TASK1 channels might be dispensable for O(2)/CO(2) sensing in mouse CB cells, TASK3 channels (or TASK1/3 heteromers) could mediate hypoxic depolarization of normal glomus cells. The ability of TASK1/3(-/-) glomus cells to maintain a powerful response to hypoxia even after blockade of maxi-K(+) channels, suggests the existence of multiple sensor and/or effector mechanisms, which could confer upon the cells a high adaptability to maintain their chemosensory function.


Subject(s)
Carotid Body/physiology , Chemoreceptor Cells/physiology , Nerve Tissue Proteins/metabolism , Potassium Channels/physiology , Animals , Cells, Cultured , Mice , Mice, Knockout
8.
Blood ; 114(16): 3439-47, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19671918

ABSTRACT

Carfilzomib is a proteasome inhibitor in clinical development that primarily targets the chymotrypsin-like (CT-L) subunits in both the constitutive proteasome (c20S) and the immunoproteasome (i20S). To investigate the impact of inhibiting the CT-L activity with carfilzomib, we set out to quantitate the levels of CT-L subunits beta5 from the c20S and LMP7 from the i20S in normal and malignant hematopoietic cells. We found that the i20S is a major form of the proteasome expressed in cells of hematopoietic origin, including multiple myeloma (MM) CD138+ tumor cells. Although specific inhibition of either LMP7 or beta5 alone was insufficient to produce an antitumor response, inhibition of all proteasome subunits was cytotoxic to both hematologic tumor cells and peripheral blood mononuclear cells. However, selective inhibition of both beta5 and LMP7 was sufficient to induce an antitumor effect in MM, non-Hodgkin lymphoma, and leukemia cells while minimizing the toxicity toward nontransformed cells. In MM tumor cells, CT-L inhibition alone was sufficient to induce proapoptotic sequelae, including proteasome substrate accumulation, Noxa and caspase 3/7 induction, and phospho-eIF2alpha suppression. These data support a hypothesis that hematologic tumor cells are uniquely sensitive to CT-L inhibition and provide a mechanistic understanding of the clinical safety profile and antitumor activity of proteasome inhibitors.


Subject(s)
Apoptosis/drug effects , Hematologic Neoplasms/drug therapy , Oligopeptides/pharmacology , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Caspase 3/metabolism , Caspase 7/metabolism , Catalytic Domain , Cell Line, Tumor , Chymotrypsin/antagonists & inhibitors , Chymotrypsin/metabolism , Drug Screening Assays, Antitumor/methods , Enzyme Induction/drug effects , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hematologic Neoplasms/enzymology , Humans , Oligopeptides/therapeutic use , Protease Inhibitors/therapeutic use , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
9.
J Physiol ; 587(Pt 9): 1917-29, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19273573

ABSTRACT

Neonatal chromaffin cells of the adrenal medulla (AM) are intrinsic chemoreceptors that secrete catecholamines in response to hypoxia, thus contributing to fetal adaptation to extrauterine life. In most mammals studied, oxygen sensitivity of AM cells disappears a few days after birth, possibly due to innervation of the adrenal gland by the cholinergic fibres of the splanchnic nerve (approximately postnatal day 7 in the rat). The mechanisms underlying these homeostatic changes in chromaffin cells are unknown. Low voltage-activated, T-type, Ca(2+) channels regulate cell excitability and their expression is up-regulated by hypoxia. Hence, we hypothesized that these channels contribute to the developmental changes in the chemoreceptive properties of AM chromaffin cells. Using electrophysiological, immunocytochemical and molecular biology methodologies we show here that neonatal AM chromaffin cells express T-type Ca(2+) channels (of alpha1H or Ca(v)3.2 sub-type) and that the function of these channels is necessary for catecholamine release in response to acute hypoxia. T-type Ca(2+) channel expression, as well as chromaffin cell responsiveness to hypoxia, decrease with postnatal maturation. Adult chromaffin cell sensitivity to hypoxia reappears after AM denervation in parallel with the recruitment of T-type Ca(2+) channels. These observations indicate that T-type Ca(2+) channels are essential for the acute response of chromaffin cells to hypoxia and help explain the disappearance of O(2) sensitivity in adult AM chromaffin cells. Our results may also be relevant for understanding the pathogenesis of disorders associated with chronic hypoxia or maternal nicotine consumption.


Subject(s)
Calcium Channels, T-Type/physiology , Chromaffin Cells/physiology , Ion Channel Gating/physiology , Oxygen Consumption/physiology , Oxygen/metabolism , Animals , Cell Hypoxia/physiology , Rats
10.
Hum Gene Ther ; 19(11): 1273-82, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19000019

ABSTRACT

Widespread adaptation of small molecule-regulated expression systems requires the development of selective inducer molecules that do not have any significant side effects on the endogenous receptors from which the regulated expression system is derived. Here we report the identification and in vitro validation of a novel inducer-receptor pair for the single-plasmid regulated expression system termed pBRES, which contains the ligand-binding domain from the human progesterone receptor (hPR). A small molecule inducer, BLX-913, has been identified as having a 30-fold lower IC(50) for the human progesterone receptor than mifepristone (MFP), the previously best characterized inducer for pBRES. Using modeling-guided protein engineering, compensatory mutations were installed at positions W755 and V729 (hPR numbering) in the ligand-binding pocket of the pBRES regulator protein (pBRES RP) to accommodate the new inducer and allow induction of transgene expression to levels previously seen with MFP. The improved inducer-pBRES RP complex was validated in vitro by monitoring the induction of luciferase, murine secreted alkaline phosphatase, and human interferon beta transgenes in mouse skeletal muscle cells. The engineered pBRES demonstrated low levels of transgene expression in the absence, and high expression levels in the presence, of the new BLX-913 inducer. Findings presented here allow induction of the pBRES-regulated gene expression system by a compound with markedly lower anti-hPR activity than MFP, the previously best characterized inducer.


Subject(s)
Breast Neoplasms/metabolism , Estrenes/pharmacology , Gene Expression Regulation, Neoplastic , Mifepristone/pharmacology , Oximes/pharmacology , Receptors, Progesterone/metabolism , Transgenes/physiology , Alkaline Phosphatase/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , DNA Primers/chemistry , Female , Genetic Vectors , Hormone Antagonists/pharmacology , Humans , Luciferases/metabolism , Mice , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Receptors, Progesterone/genetics , Transfection , Tumor Cells, Cultured
11.
Circulation ; 112(9): 1309-15, 2005 Aug 30.
Article in English | MEDLINE | ID: mdl-16116057

ABSTRACT

BACKGROUND: Hypertension, a major cause of cardiovascular morbidity and mortality, can result from chronic hypoxia; however, the pathogenesis of this disorder is unknown. We hypothesized that downregulation of the maxi-K+ channel beta1-subunit by hypoxia decreases the ability of these channels to hyperpolarize arterial smooth muscle cells, thus favoring vasoconstriction and hypertension. METHODS AND RESULTS: Lowering O2 tension produced a decrease of maxi-K+ beta1-subunit mRNA levels in rat (aortic and basilar) and human (mammary) arterial myocytes. This was paralleled by a reduction of the beta1-subunit protein level as determined by immunocytochemistry and flow cytometry. Exposure to hypoxia also produced a decrease of open probability, mean open time, and sensitivity to the xenoestrogen tamoxifen of single maxi-K+ channels recorded from patch-clamped dispersed myocytes. The number of channels per patch and the single-channel conductance were not altered. The vasorelaxing force of maxi-K+ channels was diminished in rat and human arterial rings exposed to low oxygen tension. CONCLUSIONS: These results indicate that a decrease of the maxi-K+ channel beta1-subunit expression in arterial myocytes is a key factor in the vasomotor alterations induced by hypoxia.


Subject(s)
Cell Hypoxia/physiology , Gene Expression Regulation , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Cells, Cultured , Down-Regulation , Humans , Hypertension/etiology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/physiology , Muscle, Smooth, Vascular/cytology , Phenylephrine/pharmacology , Rats , Reactive Oxygen Species , Vasodilation/drug effects
12.
Chembiochem ; 6(5): 890-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15832403

ABSTRACT

Investigations on the scope and utility of exo-mechanism proximity-accelerated reactions in engineered receptor-ligand systems are reported. We synthesized a series of electrophilic cyclosporin (CsA) derivatives by varying electrophiles and linker lengths, prepared a series of nucleophilic cysteine mutations on the surface of cyclophilin A (Cyp), and examined their reactivity and specificity in proximity-accelerated reactions. Acrylamide and epoxide electrophiles afforded useful reactivity and high specificity for alkylation of engineered receptors in Jurkat cell extracts. We found that remote cysteines (>17 A from the ligand) could be alkylated with useful rates under physiological conditions. The results from mutations of the receptor surface suggest that the dominant factors governing the rates of proximity-accelerated reactions are related to the local environment of the reactive group on the protein surface. This study defines several parameters affecting reactivity in exo-mechanism proximity-accelerated reactions and provides guidance for the design of experiments for biological investigations involving proximity-accelerated reactions.


Subject(s)
Cyclophilins/metabolism , Cyclosporins/chemistry , Cyclosporins/metabolism , Mutation/genetics , Protein Engineering , Alkylation , Cross-Linking Reagents , Cyclophilins/chemistry , Cyclophilins/genetics , Cyclosporins/chemical synthesis , Cysteine/genetics , Cysteine/metabolism , Hydrogen-Ion Concentration , Models, Molecular , Protein Structure, Tertiary , Static Electricity , Substrate Specificity
13.
J Appl Physiol (1985) ; 96(3): 1187-95; discussion 1170-2, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14766769

ABSTRACT

O(2) sensing is of critical importance for cell survival and adaptation of living organisms to changing environments or physiological conditions. O(2)-sensitive ion channels are major effectors of the cellular responses to hypoxia. These channels are preferentially found in excitable neurosecretory cells (glomus cells of the carotid body, cells in the neuroepithelial bodies of the lung, and neonatal adrenal chromaffin cells), which mediate fast cardiorespiratory adjustments to hypoxia. O(2)-sensitive channels are also expressed in the pulmonary and systemic arterial smooth muscle cells where they participate in the vasomotor responses to low O(2) tension (particularly in hypoxic pulmonary vasoconstriction). The mechanisms underlying O(2) sensing and how the O(2) sensors interact with the ion channels remain unknown. Recent advances in the field give different support to the various current hypotheses. Besides the participation of ion channels in acute O(2) sensing, they also contribute to the gene program developed under chronic hypoxia. Gene expression of T-type calcium channels is upregulated by hypoxia through the same hypoxia-inducible factor-dependent signaling pathway utilized by the classical O(2)-regulated genes. Alteration of acute or chronic O(2) sensing by ion channels could participate in the pathophysiology of human diseases, such as sudden infant death syndrome or primary pulmonary hypertension.


Subject(s)
Ion Channels/metabolism , Oxygen/metabolism , Animals , Base Sequence , Humans , Hypoxia/genetics , Hypoxia/metabolism , Ion Channels/genetics , Molecular Sequence Data , Signal Transduction/physiology
14.
J Biol Chem ; 278(25): 22316-24, 2003 Jun 20.
Article in English | MEDLINE | ID: mdl-12679337

ABSTRACT

Cellular responses to hypoxia can be acute or chronic. Acute responses mainly depend on oxygen-sensitive ion channels, whereas chronic responses rely on the hypoxia-inducible transcription factors (HIFs), which up-regulate the expression of enzymes, transporters, and growth factors. It is unknown whether the expression of genes coding for ion channels is also influenced by hypoxia. We report here that the alpha1H gene of T-type voltage-gated calcium channels is highly induced by lowering oxygen tension in PC12 cells. Accumulation of alpha1H mRNA in response to hypoxia is time- and dose-dependent and paralleled by an increase in the density of T-type calcium channel current recorded in patch clamped cells. HIF appears to be involved in the response to hypoxia, since cobalt chloride, desferrioxamine, and dimethyloxalylglycine, compounds that mimic HIF-regulated gene expression, replicate the hypoxic effect. Moreover, functional inhibition of HIF-2alpha protein accumulation using antisense HIF-2alpha oligonucleotides reverses the effect of hypoxia on T-type Ca2+ channel expression. Importantly, regulation by oxygen tension is specific for T-type calcium channels, since it is not observed with the L-, N-, and P/Q-channel types. These findings show for the first time that hypoxia induces an ion channel gene via a HIF-dependent mechanism and define a new role for the T-type calcium channels as regulators of cellular excitability and calcium influx under chronic hypoxia.


Subject(s)
Calcium Channels, T-Type/genetics , Cell Hypoxia/physiology , Animals , Base Sequence , Brain/physiology , Calcium Channels, T-Type/drug effects , Calcium Channels, T-Type/physiology , DNA Primers , Gene Expression Regulation, Neoplastic/drug effects , Molecular Sequence Data , Oligodeoxyribonucleotides, Antisense/pharmacology , PC12 Cells , Pheochromocytoma , Rats , Recombinant Proteins/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Thionucleotides/pharmacology , Transfection
15.
Org Lett ; 5(5): 693-6, 2003 Mar 06.
Article in English | MEDLINE | ID: mdl-12605492

ABSTRACT

A new approach for creating allele-specific inhibitors is demonstrated. In this approach, a receptor and ligand are engineered to contain complementary reactive groups that form a covalent bond via a proximity-accelerated reaction upon formation of the receptor-ligand complex, irreversibly modulating the biological function of the receptor. This approach is demonstrated in the cyclophilin-cyclosporin receptor-ligand system by introducing thiol and acrylamide functional groups in the receptor and ligand, respectively.


Subject(s)
Cyclophilin A/genetics , Cyclophilin A/metabolism , Cyclosporine/metabolism , Enzyme Inhibitors/metabolism , Receptors, Immunologic/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Alkylation , Alleles , Binding Sites , Cyclophilin A/pharmacology , Cyclosporine/chemistry , Cyclosporine/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Ligands , Mutagenesis, Site-Directed , Peptidylprolyl Isomerase/antagonists & inhibitors , Receptors, Immunologic/chemistry , Receptors, Immunologic/metabolism , Recombinant Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...