Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Cell Biochem Biophys ; 45(3): 289-302, 2006.
Article in English | MEDLINE | ID: mdl-16845175

ABSTRACT

Advances in the technologies for labeling and imaging biological samples drive a constant progress in our capability of studying structures and their dynamics within cells and tissues. In the last decade, the development of numerous nonlinear optical microscopies has led to a new prospective both in basic research and in the potential development of very powerful noninvasive diagnostic tools. These techniques offer large advantages over conventional linear microscopy with regard to penetration depth, spatial resolution, three-dimensional optical sectioning, and lower photobleaching. Additionally, some of these techniques offer the opportunity for optically probing biological functions directly in living cells, as highlighted, for example, by the application of second harmonic generation to the optical measurement of electrical potential and activity in excitable cells. In parallel with imaging techniques, nonlinear microscopy has been developed into a new area for the selective disruption and manipulation of intracellular structures, providing an extremely useful tool of investigation in cell biology. In this review we present some basic features of nonlinear microscopy with regard both to imaging and manipulation, and show some examples to illustrate the advantages offered by these novel methodologies.


Subject(s)
Cells, Cultured/cytology , Cells, Cultured/physiology , Imaging, Three-Dimensional/methods , Micromanipulation/methods , Microscopy/methods , Animals , Humans , Imaging, Three-Dimensional/instrumentation , Imaging, Three-Dimensional/trends , Micromanipulation/instrumentation , Micromanipulation/trends , Microscopy/instrumentation , Microscopy/trends , Nonlinear Dynamics
3.
J Biomed Opt ; 10(2): 024014, 2005.
Article in English | MEDLINE | ID: mdl-15910088

ABSTRACT

Second-harmonic generation (SHG) is emerging as a powerful tool for the optical measurement of transmembrane potential in live cells with high sensitivity and temporal resolution. Using a patch clamp, we characterize the sensitivity of the SHG signal to transmembrane potential for the RH 237 dye in various normal and tumor cell types. SHG sensitivity shows a significant dependence on the type of cell, ranging from 10 to 17% per 100 mV. Furthermore, in the samples studied, tumor cell lines display a higher sensitivity compared to normal cells. In particular, the SHG sensitivity increases in the cell line Balb/c3T3 by the transformation induced with SV40 infection of the cells. We also demonstrate that fluorescent labeling of the membrane with RH 237 at the concentration used for SHG measurements does not induce any measurable alteration in the electrophysiological properties of the cells investigated. Therefore, SHG is suitable for the investigation of outstanding questions in electrophysiology and neurobiology.


Subject(s)
Cell Physiological Phenomena , Lasers , Neoplasms/physiopathology , Optics and Photonics/instrumentation , Animals , Cell Line , Coloring Agents , Humans , Membrane Potentials , Mice , Mice, Inbred BALB C , Models, Theoretical , Patch-Clamp Techniques , Pyridinium Compounds
4.
Biochem Soc Trans ; 32(Pt 5): 826-7, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15494025

ABSTRACT

Integrins are adhesion receptors capable of transmitting intracellular signals that regulate many different cellular functions. Among integrin-mediated signals, the activation of ion channels can be included. We demonstrated that a long-lasting activation of hERG (human ether-a-go-go-related gene) potassium channels occurs in both human neuroblastoma and leukaemia cells after the activation of the beta1 integrin subunit. This activation is apparently a determining factor inducing neurite extension and osteoclastic differentiation in both the cell types. More recently, we provided evidences that beta1 integrins and hERG channels co-precipitate in both the cell types. Preliminary results suggest that a macromolecular signalling complex indeed occurs between integrins and the hERG1 protein and that hERG channel activity can modulate integrin downstream signalling.


Subject(s)
Cation Transport Proteins/metabolism , Integrins/metabolism , Potassium Channels, Voltage-Gated/metabolism , Animals , Cell Line, Tumor , Ether-A-Go-Go Potassium Channels , Humans , Leukemia/metabolism , Neuroblastoma/metabolism , Potassium/metabolism , Protein Binding , Signal Transduction
6.
Leukemia ; 16(9): 1791-8, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12200695

ABSTRACT

An important target in the understanding of the pathogenesis of acute myeloid leukemias (AML) relies on deciphering the molecular features of normal and leukemic hemopoietic progenitors. In particular, the analysis of the mechanisms involved in the regulation of cell proliferation is decisive for the establishment of new targeted therapies. To gain further insight into this topic we report herein a novel approach by analyzing the role of HERG K(+) channels in the regulation of hemopoietic cell proliferation. These channels, encoded by the human ether-a-gò-gò-related gene (herg), belong to a family of K(+) channels, whose role in oncogenesis has been recently demonstrated. We report here that herg is switched off in normal peripheral blood mononuclear cells (PBMNC) as well as in circulating CD34(+) cells, however, it is rapidly turned on in the latter upon induction of the mitotic cycle. Moreover, hergappears to be constitutively activated in leukemic cell lines as well as in the majority of circulating blasts from primary AML. Evidence is also provided that HERG channel activity regulates cell proliferation in stimulated CD34(+) as well as in blast cells from AML patients. These results open new perspectives on the pathogenetic role of HERG K(+) channels in leukemias.


Subject(s)
Cation Transport Proteins , Cell Division/physiology , DNA-Binding Proteins , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Potassium Channels/physiology , Trans-Activators , Acute Disease , Antigens, CD34/metabolism , Benzimidazoles/pharmacology , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Hematopoietic Stem Cells/cytology , Humans , Immunoenzyme Techniques , Leukemia, Myeloid/pathology , Patch-Clamp Techniques , Potassium Channel Blockers , Potassium Channels/genetics , Sulfanilamides/pharmacology , Transcriptional Regulator ERG , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology
7.
Biochem Biophys Res Commun ; 286(5): 857-62, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11527377

ABSTRACT

Using the patch-clamp technique, we analysed changes in the biophysical properties of HERG potassium channels in neuroblastoma cells long-term exposed to hypoxia. In this condition, HERG channels underwent a profound modulation that consisted of: (i) a slowing in open-close kinetics; (ii) a marked hyperpolarizing shift in voltage dependence of steady-state activation; and (iii) a slowing of the inactivation removal. The overall physiological impact of these changes in neuroblastoma cells is an increase in the HERG window current in the range of the resting potential (V(REST)), which varied between -40 and -30 mV. Such a current modulation is suitable to stabilise the resting potential (V(REST)) in hypoxic environments.


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Hypoxia , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Trans-Activators , Cell Membrane/chemistry , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Kinetics , Membrane Potentials , Neuroblastoma/metabolism , Patch-Clamp Techniques , Transcriptional Regulator ERG , Tumor Cells, Cultured
8.
J Biol Chem ; 276(7): 4923-31, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11080495

ABSTRACT

Integrin receptors have been demonstrated to mediate either "inside-to-out" and "outside-to-in" signals, and by this way are capable of regulating many cellular functions, such as cell growth and differentiation, cell migration, and activation. Among the various integrin-centered signaling pathways discovered so far, we demonstrated that the modulation of the electrical potential of the plasma membrane (V(REST)) is an early integrin-mediated signal, which is related to neurite emission in neuroblastoma cells. This modulation is sustained by the activation of HERG K(+) channels, encoded by the ether-à-go-go-related gene (herg). The involvement of integrin-mediated signaling is being discovered in the hemopoietic system: in particular, osteoclasts are generated as well as induced to differentiate by interaction of osteoclast progenitors with the stromal cells, through the involvement of integrin receptors. We studied the effects of cell interaction with the extracellular matrix protein fibronectin (FN) in a human leukemic preosteoclastic cell line (FLG 29.1 cells), which has been demonstrated to express HERG currents. We report here that FLG 29.1 cells indeed adhere to purified FN through integrin receptors, and that this adhesion induces an osteoclast phenotype in these cells, as evidenced by the appearance of tartrate-resistant acid phosphatase, as well as by the increased expression of CD51/alpha(v)beta(3) integrin and calcitonin receptor. An early activation of HERG current (I(HERG)), without any increase in herg RNA or modifications of HERG protein was also observed in FN-adhering cells. This activation is apparently sustained by the beta(1) integrin subunit activation, through the involvement of a pertussis-toxin sensitive G(i) protein, and appears to be a determinant signal for the up-regulation of alpha(v)beta(3) integrin, as well as for the increased expression of calcitonin receptor.


Subject(s)
Cation Transport Proteins , Cell Adhesion , DNA-Binding Proteins , Fibronectins/metabolism , Integrin beta1/physiology , Osteoclasts/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Receptors, Vitronectin/genetics , Trans-Activators , Antibodies, Monoclonal/immunology , Cell Differentiation , ERG1 Potassium Channel , Electric Conductivity , Ether-A-Go-Go Potassium Channels , Filaggrin Proteins , Humans , Integrin beta1/immunology , Leukemia , Osteoclasts/cytology , Patch-Clamp Techniques , Potassium Channels/genetics , RNA, Messenger/biosynthesis , Receptors, Calcitonin/biosynthesis , Receptors, Calcitonin/genetics , Receptors, Vitronectin/biosynthesis , Stem Cells/cytology , Stem Cells/metabolism , Transcriptional Regulator ERG , Tumor Cells, Cultured , Up-Regulation
9.
Br J Cancer ; 83(12): 1722-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11104572

ABSTRACT

HERG K(+)channels, besides contributing to regulate cardiac and neuronal excitability, are preferentially expressed in tumour cell lines of different histogenesis, where their role in the development and maintenance of the neoplastic phenotype is under study. We show here that both herg gene and HERG protein are expressed with high frequency in primary human endometrial cancers, as compared to normal and hyperplastic endometrium. RT-PCR and immunohistochemistry, using specific anti-HERG antibodies developed in our laboratory, were applied to tissue specimens obtained from 18 endometrial cancers and 11 non-cancerous endometrial tissues. herg RNA and HERG protein are expressed in 67% and 82%, respectively, of cancerous, while in only 18% of non-cancerous tissues. In particular, no expression was found in endometrial hyperplasia. Moreover, electrophysiological experiments confirmed the presence of functioning HERG channels on the plasma membrane of tumour cells. On the whole, these data are the first demonstration of the presence of HERG channels in primary human neoplasias, and could candidate HERG as a potential tool capable of marking cancerous versus hyperplastic endometrial growth.


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Endometrial Neoplasms/metabolism , Endometrium/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Trans-Activators , Aged , Aged, 80 and over , Benzimidazoles/pharmacology , Blotting, Western , ERG1 Potassium Channel , Electrophysiology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrium/pathology , Ether-A-Go-Go Potassium Channels , Female , Gene Expression Regulation , Humans , Hyperplasia , Immunohistochemistry , Membrane Potentials/genetics , Middle Aged , Potassium Channels/genetics , RNA/genetics , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sulfanilamides/pharmacology , Transcriptional Regulator ERG , Tumor Cells, Cultured
10.
FASEB J ; 14(15): 2601-10, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11099479

ABSTRACT

The human ether-a-go-go-related genes (herg) are expressed in tissues other than heart and brain where the HERG K(+) channels are known to regulate the repolarization of the heart action potential and the neuronal spike-frequency accommodation. We provide evidence that herg1 transcripts are present in human pancreatic islets that were used to study both insulin secretion and electrical activity with radioimmunoassay and single cell perforated patch-clamp techniques, respectively. Glucose- and arginine-induced islets insulin secretion data suggested a net increase of release under perfusion with antiarrhythmic drugs known to selectively block HERG channels. Indeed we could routinely isolate a K(+) current that was recognized as biophysically and pharmacologically similar to the HERG current. An analysis of the glucose- and arginine-induced electrical activity (several applications during 30 min) in terms of firing frequency and putative insulin release was done in control and in the presence of selective blockers of HERG channels: the firing frequency and the release increased by 32% and 77%, respectively. It is concluded that HERG channels have a crucial role in regulating insulin secretion and firing of human beta-cells. This raises the possibility that some genetically characterized hyperinsulinemic diseases of unknown origin might involve mutations in the HERG channels.


Subject(s)
Arginine/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Trans-Activators , ATP-Binding Cassette Transporters , Benzimidazoles/pharmacology , ERG1 Potassium Channel , Electric Conductivity , Ether-A-Go-Go Potassium Channels , Humans , Insulin Secretion , Islets of Langerhans/drug effects , KATP Channels , Patch-Clamp Techniques , Potassium/metabolism , Potassium Channel Blockers , Potassium Channels, Inwardly Rectifying , Radioimmunoassay , Reproducibility of Results , Sulfanilamides/pharmacology , Tolbutamide/pharmacology , Transcriptional Regulator ERG
11.
Mech Dev ; 95(1-2): 239-43, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10906470

ABSTRACT

The expression pattern of K(+) currents is the principal regulator of electrical activity during development of the nervous and muscular system. We report here a study showing the expression pattern of HERG K(+) currents-encoding (erg) genes in various nervous and muscular tissues at different stages of quail embryo development.


Subject(s)
Cation Transport Proteins , Gene Expression Regulation, Developmental , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Quail/embryology , Quail/genetics , Amino Acid Sequence , Animals , Embryo, Nonmammalian/physiology , Ether-A-Go-Go Potassium Channels , Molecular Sequence Data , Muscles/embryology , Nervous System/embryology , Sequence Alignment
12.
Biochim Biophys Acta ; 1466(1-2): 278-88, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10825449

ABSTRACT

Hybrid polar compounds (HPCs) are powerful inducers of terminal differentiation of various types of tumors, including Friend murine erythroleukemia cells (MELCs). They are known to act synergistically with an increase in the extracellular concentration of cations, which causes a positive shift in the negative value of the ionic surface potential. Two HPCs, hexamethylenebisacetamide (HMBA) and suberoylanilide hydroxamic acid (SAHA), were adsorbed on self-assembled phospholipid monolayers supported on a mercury drop and the shift in the surface dipole potential chi of the lipid film due to their adsorption was estimated from charge measurements. At their optimal concentrations for inducing MELC terminal differentiation (5 mM for HMBA and 2.6 microM for SAHA), these HPCs cause a chi shift of about 15-20 mV, positive toward the hydrocarbon tails, both on neutral phosphatidylcholine films and on negatively or positively charged phosphatidylserine films. This strongly suggests that the nonspecific effect of HPCs of different structure in inducing cancer cells to rescue their differentiation program is related to a positive chi shift on the extracellular side of the cell membrane.


Subject(s)
Acetamides/chemistry , Antineoplastic Agents/chemistry , Hydroxamic Acids/chemistry , Malonates/chemistry , Phosphatidylcholines/chemistry , Phosphatidylserines/chemistry , Absorption , Mercury/chemistry , Molecular Structure , Phospholipids/chemistry , Vorinostat
13.
Leukemia ; 14(4): 735-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10764163

ABSTRACT

In liquid cultures of murine bone marrow cells stimulated with interleukin-3 and granulocyte/macrophage colony-stimulating factor, hypoxia (1% oxygen) induced a reversible block of hematopoiesis, maintaining the progenitors' expansion potential unreduced. Progenitors repopulating day-14 hypoxic cultures with cells or granulocyte/macrophage colony-forming units (CFU-GM) were found, on the basis of their maintenance in hypoxia (12% and 76%, respectively), to belong to different subsets, the latter being much more efficiently maintained. The maintenance in hypoxic cultures of progenitors detectable by marrow-repopulating ability (MRA) assay was 18% for MRAcell progenitors and 69% for MRACFU progenitors. Thus, the repopulation of hypoxic cultures with cells or CFU-GM closely reflected the presence of progenitors capable of repopulating, with cells or CFU-GM, the bone marrow of lethally irradiated syngeneic animals. Progenitors repopulating hypoxic cultures were, like MRA progenitors, significantly resistant to 5-fluorouracil, progenitors repopulating cultures with CFU-GM being two-fold more resistant than those repopulating cultures with cells. We concluded that the repopulation of day-14 hypoxic cultures occurring after their transfer to air is to be considered an indicator of the maintenance of MRA progenitors in hypoxia. The relevance of these results to stem cell biology and their potential practical applications are discussed.


Subject(s)
Bone Marrow Cells/cytology , Cell Hypoxia , Animals , Antimetabolites/pharmacology , Cells, Cultured , Colony-Forming Units Assay , Drug Resistance , Fluorouracil/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Mice , Mice, Inbred CBA , Radiation Chimera
14.
Leukemia ; 14(12): 2112-7, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11187900

ABSTRACT

Although dephosphorylation of tyrosine containing proteins is considered a necessary step in the induction of leukemia cell differentiation by hybrid polar compounds (HPC), the crucial actors in this step remain unknown. We present evidence that tyrosine phosphorylation of JAK1 and JAK2 is down-regulated in murine erythroleukemia cells (MELC) within the first 6 h of their exposure to the prototype of the HPC family, hexamethylenebisacetamide (HMBA), with full recovery at 14 h. Further evidence that the JAKs-centered signalling pathway is switched off early by HMBA was provided by the demonstration that STAT5, a downstream member of this pathway, turned out to be completely dephosphorylated at 6 h in HMBA-treated cells. This JAKs dephosphorylation did not occur in HMBA-resistant clones, suggesting that JAKs are possible targets of the dephosphorylative process required for leukemia cell commitment to differentiation. These results may have impact on leukemia therapy based on JAKs inhibitors.


Subject(s)
Acetamides/pharmacology , Cell Differentiation/drug effects , Leukemia, Erythroblastic, Acute/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins , Animals , Janus Kinase 1 , Janus Kinase 2 , Leukemia, Erythroblastic, Acute/pathology , Mice , Phosphorylation , Tumor Cells, Cultured
15.
J Neurobiol ; 40(2): 214-25, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10413451

ABSTRACT

The modulation of herg gene and HERG currents (I(HERG)) was studied in SH-SY5Y neuroblastoma (NB) cells treated with all-trans-retinoic acid (RA) in the absence or presence of the neurotrophin brain-derived neurotrophic factor (BDNF). Both treatments produced a strong increase in the percentage of cells differentiated along the neuronal pathway, with an orientation to a cholinergic phenotype, while a minority of cells displayed a glial phenotype particularly evident after long-term exposure to the inducers. Differentiation of NB cells was accompanied by an increase in herg gene transcription, which attained its maximum after 6 days of treatment with RA and was not further increased by BDNF. This effect evidently reflected on HERG currents: In fact, RA produced an increase in HERG current density which was strongly potentiated by BDNF. Moreover, RA treatment affected the biophysical properties of I(HERG), inducing an increase in the deactivation time constant and a left shift of the activation curve. These effects were not substantially affected by BDNF. This modulation of I(HERG) influenced the value of the resting potential (V(REST)), which resulted significantly hyperpolarized in (RA with or without BDNF)-treated cells. Interestingly, these effects were absent in the glial population, which prevailed in cultures after long-term exposure to the inducers. On the whole, we demonstrate that besides expressing IRK currents, NB cells display another strategy to hyperpolarize their V(REST), based on the appropriate modulation of HERG currents. Different from what happens in normal neuroblast development, the latter are never lost by cancer cells despite the progression of these cells along the neuronal differentiative pathway, raising intriguing questions about the role of HERG currents in tumour behavior.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/physiology , Neuroblastoma/pathology , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Potassium/metabolism , Trans-Activators , Tretinoin/pharmacology , Acetylcholine/metabolism , Cell Differentiation/drug effects , Drug Synergism , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Fluorescent Antibody Technique, Indirect , Humans , Ion Transport/drug effects , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Patch-Clamp Techniques , Potassium Channels/biosynthesis , Potassium Channels/genetics , Transcriptional Regulator ERG , Tumor Cells, Cultured/drug effects
16.
FASEB J ; 13(8): 953-62, 1999 May.
Article in English | MEDLINE | ID: mdl-10224238

ABSTRACT

Toxins isolated from a variety of venoms are tools for probing the physiological function and structure of ion channels. The ether-a-go-go-related genes (erg) codify for the K+ channels (ERG), which are crucial in neurons and are impaired in human long-QT syndrome and Drosophila 'seizure' mutants. We have isolated a peptide from the scorpion Centruroides noxius Hoffmann that has no sequence homologies with other toxins, and demonstrate that it specifically inhibits (IC50=16+/-1 nM) only ERG channels of different species and distinct histogenesis. These results open up the possibility of investigating ERG channel structure-function relationships and novel pharmacological tools with potential therapeutic efficacy.


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Scorpion Venoms/chemistry , Scorpion Venoms/isolation & purification , Scorpion Venoms/toxicity , Trans-Activators , Action Potentials/drug effects , Amino Acid Sequence , Animals , Cell Line , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Endocrine Glands/drug effects , Endocrine Glands/metabolism , Ether-A-Go-Go Potassium Channels , Guinea Pigs , Humans , Kinetics , Mice , Molecular Sequence Data , Myocardium/metabolism , Neurons/drug effects , Neurons/metabolism , Potassium Channels/genetics , Rats , Scorpion Venoms/genetics , Scorpions , Sequence Homology, Amino Acid , Transcriptional Regulator ERG
17.
Eur J Neurosci ; 11(3): 753-60, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10103069

ABSTRACT

ERG (ether-à-go-go-related gene) K+ channels are crucial in human heart physiology (h-ERG), but are also found in neuronal cells and are impaired in Drosophila 'seizure' mutants. Their biophysical properties include the relatively fast kinetics of the inactivation gate and much slower kinetics of the activation gate. In order to elucidate how the complex time- and voltage-dependent activation properties of ERG channels underlies distinct roles in excitability, we investigated different types of ERG channels intrinsically present in cells or heterologously expressed in mammalian cells or Xenopus oocytes. Voltage-dependent activation curves were highly dependent on the features of the eliciting protocols. Only very long preconditioning times produced true steady-state relationships, a fact that has been largely neglected in the past, hampering the comparison of published data on ERG channels. Beyond this technical aspect, the slow activation property of ERG can be responsible for unsuspected physiological roles. We found that around the midpoint of the activation curve, the time constant of ERG open-close kinetics is of the order of 10-15 s. During sustained trains of depolarizations, e.g. those produced in neuronal firing, this leads to the use-dependent accumulation of open-state ERG channels. Accumulation is not observed in a mutant with a fast activation gate. In conclusion, it is well established that other K+ channels (i.e. Ca2+-activated and M) control the spike-frequency adaptation, but our results support the notion that the purely voltage-dependent activation property of ERG channels would allow a slow inhibitory physiological role in rapid neuronal signalling.


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Ion Channel Gating/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Potassium Channels/metabolism , Trans-Activators , Animals , ERG1 Potassium Channel , Electric Stimulation , Electrophysiology , Ether-A-Go-Go Potassium Channels , Ganglia, Spinal/cytology , Humans , Kidney/cytology , Leukemia , Membrane Potentials/physiology , Mice , Mutagenesis/physiology , Neuroblastoma , Oocytes/physiology , Rats , Transcriptional Regulator ERG , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/physiology , Xenopus
18.
Psychother Psychosom ; 68(1): 46-50, 1999.
Article in English | MEDLINE | ID: mdl-9873242

ABSTRACT

BACKGROUND: To explore the impact of a prevention program on the eating and body attitudes of a sample of adolescent schoolgirls. METHODS: The program involved lessons and group discussions of general adolescent problems and eating disorders. A total of 254 16-year-old schoolgirls were evaluated, of whom 154 participated in the program and a further 154 subjects formed the control group. Variations in weight, Eating Attitudes Test and Eating Disorders Inventory at a 1-year follow-up were compared for the two groups. RESULTS: Among high-risk subjects, no significant differences were found between the prevention and the control group. The preventive program appeared to reduce significantly body dissatisfaction and to decrease the risk of bulimic attitudes in low-risk subjects. CONCLUSIONS: Providing schoolgirls with the correct information about eating disorders did not encourage unhealthy attitudes to eating and weight regulation practices. However, for high-risk subjects more intensive and specific intervention may be required, for which further research is needed.


Subject(s)
Feeding and Eating Disorders/prevention & control , Health Knowledge, Attitudes, Practice , Health Promotion/methods , Psychotherapy, Group/methods , Adolescent , Adolescent Behavior/psychology , Analysis of Variance , Body Image , Body Mass Index , Female , Follow-Up Studies , Humans , Interpersonal Relations , Risk Factors
19.
Biochem Biophys Res Commun ; 244(3): 706-11, 1998 Mar 27.
Article in English | MEDLINE | ID: mdl-9535729

ABSTRACT

The modulation of inward K+ conductances was studied during neuronal differentiation of human SH-SY5Y neuroblastoma cells. Under standard culture conditions, these cells express the herg gene, and the HERG current is the main inward K+ current regulating their Vrest. After 10-20 days exposure to Retinoic Acid (RA), SH-SY5Y cells showed, in addition to HERG currents, a novel current characterized by inward rectification, dependence on the extracellular K+ concentration, and blockade by Cs+ and Ba2+, the main features of the IRK1 current. The appearance of this current is accompanied by a strong hyperpolarisation of Vrest. RT-PCR experiments confirmed that a transcript of the IRK1 (Kir 2.1) gene actually appears in SH-SY5Y cells treated for 10-20 days with RA. On the whole, data here presented demonstrate that RA-induced neuronal differentiation of neuroblastoma cells is accompanied by the switch from a HERG-driven to a IRK1-driven control of Vrest, similarly to what happens in normal differentiating neurons; however, in tumor cells, this switch does not imply the abolition of HERG channel expression.


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Neurons/drug effects , Potassium Channels, Inwardly Rectifying , Potassium Channels, Voltage-Gated , Potassium Channels/biosynthesis , Trans-Activators , Tretinoin/pharmacology , Cell Differentiation , ERG1 Potassium Channel , Electric Conductivity , Ether-A-Go-Go Potassium Channels , Gene Expression , Humans , Neuroblastoma , Patch-Clamp Techniques , Potassium Channels/genetics , RNA, Messenger/analysis , Time Factors , Transcriptional Regulator ERG , Tumor Cells, Cultured
20.
Cancer Res ; 58(4): 815-22, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9485040

ABSTRACT

The human ether-a-go-go-related gene (herg) encodes a K+ current (IHERG) that plays a fundamental role in heart excitability by regulating the action potential repolarization (IKr); mutations of this gene are responsible for the chromosome 7-linked long QT syndrome (LQT2). In this report, we show that in a variety (n = 17) of tumor cell lines of different species (human and murine) and distinct histogenesis (neuroblastoma, rhabdomyosarcoma, adenocarcinoma, lung microcytoma, pituitary tumors, insulinoma beta-cells, and monoblastic leukemia), a novel K+ inward-rectifier current (IIR), which is biophysically and pharmacologically similar to IHERG, can be recorded with the patch-clamp technique. Northern blot experiments with a human herg cDNA probe revealed that both in human and murine clones the very high expression of herg transcripts can be quantified in at least three clearly identifiable bands, suggesting an alternative splicing of HERG mRNA. Moreover, we cloned a cDNA encoding for IIR from the SH-SY5Y human neuroblastoma. The sequence of this cDNA result was practically identical to that already reported for herg, indicating a high conservation of this gene in tumors. Consistently, the expression of this clone in Xenopus oocytes showed that the encoded K+ channel had substantially all of the biophysical and pharmacological properties of the native IIR described for tumor cells. In addition, in the tumor clones studied, IIR governs the resting potential, whereas it could not be detected either by the patch clamp or the Northern blot techniques in cells obtained from primary cell cultures of parental tissues (sensory neurons and myotubes), whose resting potential is controlled by the classical K+ anomalous rectifier current. This current substitution had a profound impact on the resting potential, which was markedly depolarized in tumors as compared with normal cells. These results suggest that IIR is normally only expressed during the early stages of cell differentiation frozen by neoplastic transformation, playing an important pathophysiological role in the regulatory mechanisms of neoplastic cell survival. In fact, because of its biophysical features, IIR, besides keeping the resting potential within the depolarized values required for unlimited tumor growth, could also appear suitable to afford a selective advantage in an ischemic environment.


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Neoplasms/genetics , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Trans-Activators , Amino Acid Sequence , Animals , Base Sequence , Biophysical Phenomena , Biophysics , Cell Lineage , Cell Transformation, Neoplastic , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Mice , Molecular Sequence Data , Muscles/physiology , Neuroblastoma/genetics , Neurons/physiology , Transcriptional Regulator ERG , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...