Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Cell Biochem ; 111(3): 643-52, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20589763

ABSTRACT

In several human cancers, ErbB2 over-expression facilitates the formation of constitutively active homodimers resistant to internalization which results in progressive signal amplification from the receptor, conducive to cell survival, proliferation, or metastasis. Here we report on studies of the influence of ErbB2 over-expression on localization and signaling in polarized Caco-2 and MDCK cells, two established models to study molecular trafficking. In these cells, ErbB2 is not over-expressed and shares basolateral localization with ErbB3. Over-expression of ErbB2 by transient transfection resulted in partial separation of the receptors by relocalization of ErbB2, but not ErbB3, to the apical surface, as shown by biotinylation of the apical or basolateral surfaces. These results were confirmed by immunofluorescence and confocal microscopy. Polarity controls indicated that the relocalization of ErbB2 is not the result of depolarization of the cells. Biotinylation and confocal microscopy also showed that apical, but not basolateral ErbB2 is activated at tyrosine 1139. This phosphotyrosine binds adaptor protein Grb2, as confirmed by immunoprecipitation. However, we found that it does not initiate the canonical Grb2-Ras-Raf-Erk pathway. Instead, our data supports the activation of a survival pathway via Bcl-2. The effects of ErbB2 over-expression were abrogated by the humanized anti-ErbB2 monoclonal antibody Herceptin added only from the apical side. The ability of apical ErbB2 to initiate an altered downstream cascade suggests that subcellular localization of the receptor plays an important role in regulating ErbB2 signaling in polarized epithelia.


Subject(s)
Cell Polarity , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction , Animals , Caco-2 Cells , Cell Line , Cell Survival , Dogs , Gene Expression , Humans , Protein Transport , Proto-Oncogene Proteins c-bcl-2 , Receptor, ErbB-2/pharmacology
2.
J Cell Sci ; 122(Pt 14): 2491-503, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19549684

ABSTRACT

Atypical PKC (PKC iota) is a key organizer of cellular asymmetry. Sequential extractions of intestinal cells showed a pool of enzymatically active PKC iota and the chaperone Hsp70.1 attached to the apical cytoskeleton. Pull-down experiments using purified and recombinant proteins showed a complex of Hsp70 and atypical PKC on filamentous keratins. Transgenic animals overexpressing keratin 8 displayed delocalization of Hsp70 and atypical PKC. Two different keratin-null mouse models, as well as keratin-8 knockdown cells in tissue culture, also showed redistribution of Hsp70 and a sharp decrease in the active form of atypical PKC, which was also reduced by Hsp70 knockdown. An in-vitro turn motif rephosphorylation assay indicated that PKC iota is dephosphorylated by prolonged activity. The Triton-soluble fraction could rephosphorylate PKC iota only when supplemented with the cytoskeletal pellet or filamentous highly purified keratins, a function abolished by immunodepletion of Hsp70 but rescued by recombinant Hsp70. We conclude that both filamentous keratins and Hsp70 are required for the rescue rephosphorylation of mature atypical PKC, regulating the subcellular distribution and steady-state levels of active PKC iota.


Subject(s)
Enterocytes/enzymology , HSP70 Heat-Shock Proteins/metabolism , Intermediate Filaments/enzymology , Isoenzymes/metabolism , Keratins/metabolism , Protein Kinase C/metabolism , Animals , Caco-2 Cells , HSP70 Heat-Shock Proteins/genetics , Humans , Isoenzymes/genetics , Keratin-18/metabolism , Keratin-19/metabolism , Keratin-8/metabolism , Keratins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphorylation , Protein Kinase C/genetics , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference
3.
J Cell Sci ; 121(Pt 5): 644-54, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18270268

ABSTRACT

Atypical protein kinase iota (PKCiota) is a key organizer of the apical domain in epithelial cells. Ezrin is a cytosolic protein that, upon activation by phosphorylation of T567, is localized under the apical membrane where it connects actin filaments to membrane proteins and recruits protein kinase A (PKA). To identify the kinase that phosphorylates ezrin T567 in simple epithelia, we analyzed the expression of active PKC and the appearance of T567-P during enterocyte differentiation in vivo. PKCiota phosphorylated ezrin on T567 in vitro, and in Sf9 cells that do not activate human ezrin. In CACO-2 human intestinal cells in culture, PKCiota co-immunoprecipitated with ezrin and was knocked down by shRNA expression. The resulting phenotype showed a modest decrease in total ezrin, but a steep decrease in T567 phosphorylation. The PKCiota-depleted cells showed fewer and shorter microvilli and redistribution of the PKA regulatory subunit. Expression of a dominant-negative form of PKCiota also decreased T567-P signal, and expression of a constitutively active PKCiota mutant showed depolarized distribution of T567-P. We conclude that, although other molecular mechanisms contribute to ezrin activation, apically localized phosphorylation by PKCiota is essential for the activation and normal distribution of ezrin at the early stages of intestinal epithelial cell differentiation.


Subject(s)
Cell Membrane/enzymology , Cytoskeletal Proteins/metabolism , Intestinal Mucosa/enzymology , Isoenzymes/metabolism , Membrane Microdomains/enzymology , Protein Kinase C/metabolism , Amino Acid Sequence/physiology , Animals , Binding Sites/physiology , Caco-2 Cells , Cell Differentiation/physiology , Cell Membrane/ultrastructure , Cell Polarity/physiology , Cytoskeletal Proteins/chemistry , Down-Regulation/physiology , Enzyme Activation/physiology , Humans , Insecta , Intestinal Mucosa/cytology , Isoenzymes/genetics , Membrane Microdomains/ultrastructure , Mice , Microvilli/enzymology , Microvilli/ultrastructure , Phosphorylation , Protein Kinase C/genetics , Protein Subunits/metabolism , RNA, Small Interfering/genetics , Tyrosine/metabolism
4.
Exp Cell Res ; 313(10): 2255-64, 2007 Jun 10.
Article in English | MEDLINE | ID: mdl-17425955

ABSTRACT

Intermediate filaments have long been considered mechanical components of the cell that provide resistance to deformation stress. Practical experimental problems, including insolubility, lack of good pharmacological antagonists, and the paucity of powerful genetic models have handicapped the research of other functions. In single-layered epithelial cells, keratin intermediate filaments are cortical, either apically polarized or apico-lateral. This review analyzes phenotypes of genetic manipulations of simple epithelial cell keratins in mice and Caenorhabditis elegans that strongly suggest a role of keratins in apico-basal polarization and membrane traffic. Published evidence that intermediate filaments can act as scaffolds for proteins involved in membrane traffic and signaling is also discussed. Such a scaffolding function would generate a highly polarized compartment within the cytoplasm of simple epithelial cells. While in most cases mechanistic explanations for the keratin-null or overexpression phenotypes are still missing, it is hoped that investigators will be encouraged to study these as yet poorly understood functions of intermediate filaments.


Subject(s)
Cell Polarity/physiology , Epithelial Cells/metabolism , Intermediate Filament Proteins/metabolism , Intermediate Filaments/metabolism , Animals , Epithelial Cells/ultrastructure , Exocytosis/physiology , Humans , Intermediate Filament Proteins/ultrastructure , Intermediate Filaments/ultrastructure , Invertebrates/metabolism , Keratins/metabolism , Keratins/ultrastructure , Mammals/metabolism , Mice , Microtubules/metabolism , Microtubules/ultrastructure
5.
Mol Biol Cell ; 18(3): 781-94, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17182859

ABSTRACT

In simple epithelial cells, attachment of microtubule-organizing centers (MTOCs) to intermediate filaments (IFs) enables their localization to the apical domain. It is released by cyclin-dependent kinase (Cdk)1 phosphorylation. Here, we identified a component of the gamma-tubulin ring complex, gamma-tubulin complex protein (GCP)6, as a keratin partner in yeast two-hybrid assays. This was validated by binding in vitro of both purified full-length HIS-tagged GCP6 and a GCP6(1397-1819) fragment to keratins, and pull-down with native IFs. Keratin binding was blocked by Cdk1-mediated phosphorylation of GCP6. GCP6 was apical in normal enterocytes but diffuse in K8-null cells. GCP6 knockdown with short hairpin RNAs (shRNAs) in CACO-2 cells resulted in gamma-tubulin signal scattered throughout the cytoplasm, microtubules (MTs) in the perinuclear and basal regions, and microtubule-nucleating activity localized deep in the cytoplasm. Expression of a small fragment GCP6(1397-1513) that competes binding to keratins in vitro displaced gamma-tubulin from the cytoskeleton and resulted in depolarization of gamma-tubulin and changes in the distribution of microtubules and microtubule nucleation sites. Expression of a full-length S1397D mutant in the Cdk1 phosphorylation site delocalized centrosomes. We conclude that GCP6 participates in the attachment of MTOCs to IFs in epithelial cells and is among the factors that determine the peculiar architecture of microtubules in polarized epithelia.


Subject(s)
Epithelial Cells/metabolism , Intermediate Filaments/metabolism , Keratins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Animals , CDC2 Protein Kinase/metabolism , COS Cells , Cell Polarity , Chlorocebus aethiops , Down-Regulation/genetics , Epithelial Cells/cytology , Histones/genetics , Humans , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Organizing Center/metabolism , Mutation/genetics , Phosphorylation , Promoter Regions, Genetic/genetics , Protein Binding , Protein Transport , RNA, Small Interfering/metabolism , Transcription, Genetic , Tubulin/metabolism
6.
Mol Biol Cell ; 17(7): 2931-41, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16624867

ABSTRACT

Muc4 serves as an intramembrane ligand for the receptor tyrosine kinase ErbB2. The time to complex formation and the stoichiometry of the complex were determined to be <15 min and 1:1 by analyses of Muc4 and ErbB2 coexpressed in insect cells and A375 tumor cells. In polarized CACO-2 cells, Muc4 expression causes relocalization of ErbB2, but not its heterodimerization partner ErbB3, to the apical cell surface, effectively segregating the two receptors. The apically located ErbB2 is phosphorylated on tyrosines 1139 and 1248. The phosphorylated ErbB2 in CACO-2 cells recruits the cytoplasmic adaptor protein Grb2, consistent with previous studies showing phosphotyrosine 1139 to be a Grb2 binding site. To address the issue of downstream signaling from apical ErbB2, we analyzed the three MAPK pathways of mammalian cells, Erk, p38, and JNK. Consistent with the more differentiated phenotype of the CACO-2 cells, p38 phosphorylation was robustly increased by Muc4 expression, with a consequent activation of Akt. In contrast, Erk and JNK phosphorylation was not changed. The ability of Muc4 to segregate ErbB2 and other ErbB receptors and to alter downstream signaling cascades in polarized epithelial cells suggests that it has a role in regulating ErbB2 in differentiated epithelia.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , Mucins/metabolism , Receptor, ErbB-2/metabolism , Binding Sites , Caco-2 Cells , Cell Polarity , Cytoplasm/chemistry , Cytoplasm/metabolism , Enzyme Activation , Epithelial Cells/chemistry , Epithelial Cells/metabolism , GRB2 Adaptor Protein/metabolism , Humans , Ligands , Mitogen-Activated Protein Kinases/metabolism , Mucin-4 , Mucins/analysis , Phosphorylation , Receptor, ErbB-2/analysis , Signal Transduction , Tyrosine/metabolism
7.
Mol Biol Cell ; 16(9): 4096-107, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15987737

ABSTRACT

Ezrin connects the apical F-actin scaffold to membrane proteins in the apical brush border of intestinal epithelial cells. Yet, the mechanisms that recruit ezrin to the apical domain remain obscure. Using stable CACO-2 transfectants expressing keratin 8 (K8) antisense RNA under a tetracycline-responsive element, we showed that the actin-ezrin scaffold cannot assemble in the absence of intermediate filaments (IFs). Overexpression of ezrin partially rescued this phenotype. Overexpression of K8 in mice also disrupted the assembly of the brush border, but ezrin distributed away from the apical membrane in spots along supernumerary IFs. In cytochalasin D-treated cells ezrin localized to a subapical compartment and coimmunoprecipitated with IFs. Overexpression of ezrin in undifferentiated cells showed a Triton-insoluble ezrin compartment negative for phospho-T567 (dormant) ezrin visualized as spots along IFs. Pulse-chase analysis showed that Triton-insoluble, newly synthesized ezrin transiently coimmunoprecipitates with IFs during the first 30 min of the chase. Dormant, but not active (p-T567), ezrin bound in vitro to isolated denatured keratins in Far-Western analysis and to native IFs in pull-down assays. We conclude that a transient association to IFs is an early step in the polarized assembly of apical ezrin in intestinal epithelial cells.


Subject(s)
Epithelial Cells/metabolism , Intermediate Filaments/metabolism , Intestinal Mucosa/metabolism , Phosphoproteins/metabolism , Animals , Caco-2 Cells , Cell Differentiation/physiology , Cell Line , Cytoskeletal Proteins , Enterocytes/cytology , Enterocytes/metabolism , Humans , Intestinal Mucosa/cytology , Keratin-8 , Keratins/biosynthesis , Keratins/genetics , Mice , Mice, Transgenic , Octoxynol
8.
J Biol Chem ; 278(32): 30142-7, 2003 Aug 08.
Article in English | MEDLINE | ID: mdl-12748185

ABSTRACT

Muc4/Sialomucin complex (SMC) acts as an intramembrane ligand for the receptor tyrosine kinase ErbB2, inducing a limited phosphorylation of the receptor. Because Muc4/SMC is found at the apical surfaces of polarized epithelial cells and ErbB2 is often basolateral, the question arises as to whether these components become associated in polarized cells. To address this question, we examined the localization of these proteins in polarized human colon carcinoma CACO-2 cells. Dual color immunofluorescence analysis by confocal microscopy demonstrated the basolateral localization of the ErbB2 in these cells; it is primarily co-localized with E-cadherin at adherens junctions. Expression of apical Muc4/SMC in these cells by transient transfection results in the localization of the ErbB2 at the apical surface. Two-color confocal microscopy indicated that ErbB2 is colocalized with Muc4/SMC in the transfected cells but not in untransfected cells in the same culture. The change of localization of ErbB2 was confirmed by cell surface biotinylation of apical and basolateral proteins, followed by streptavidin precipitation and the subsequent detection of ErbB2 by immunoblotting. In contrast, Na+/K+-ATPase maintains its basolateral localization in Muc4/SMC-transfected cells, indicating that the translocation of ErbB2 is not the result of depolarization of the cells. A potential physiological role for the apical localization of ErbB2 is indicated by the fact that ErbB2 phosphorylated at tyrosine 1248 is found predominantly in Muc4/SMC-transfected cells, but not in untransfected cells, and is co-localized with the apical Muc4/SMC. The ability of Muc4/SMC to alter the localization of ErbB2, particularly a phosphorylated form of it, in epithelial cells, suggests that it has an important role in regulating ErbB2 signaling.


Subject(s)
Epithelial Cells/metabolism , Mucins/chemistry , Receptor, ErbB-2/metabolism , Biotinylation , Cadherins/metabolism , Electrophoresis, Polyacrylamide Gel , Humans , Immunoblotting , Ligands , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Mucin-4 , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Protein Transport , Sialomucins , Signal Transduction , Sodium-Potassium-Exchanging ATPase/metabolism , Streptavidin/pharmacology , Transfection , Tumor Cells, Cultured , Tyrosine/metabolism
9.
Am J Physiol Renal Physiol ; 285(2): F230-40, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12709392

ABSTRACT

We have previously shown that microtubule-organizing centers (MTOCs) attach to the apical network of intermediate filaments (IFs) in epithelial cells in culture and in epithelia in vivo. Because that attachment is important for the architecture of microtubules (MTs) in epithelia, we analyzed whether chemical anoxia in LLC-PK1 and CACO-2 cells or unilateral ischemia-reperfusion in rat kidney (performed under fluorane anesthesia) had an effect on the binding and distribution of MTOCs. In culture, we found that chemical anoxia induces MTOC detachment from IFs by morphological and biochemical criteria. In reperfused rat proximal tubules, noncentrosomal MTOCs were fully detached from the cytoskeleton and scattered throughout the cytoplasm at 3 days after reperfusion, when brush borders were mostly reassembled. At that time, MTs were also fully reassembled but, as expected, lacked their normal apicobasal orientation. Two apical membrane markers expressed in S2 and S3 segments were depolarized at the same stage. At 8 days after reperfusion, membrane polarity, MTOCs, and MTs were back to normal. Na+-K+-ATPase was also found redistributed, not to the apical domain but rather to an intracellular compartment, as described by others (Alejandro VS, Nelson W, Huie P, Sibley RK, Dafoe D, Kuo P, Scandling JD Jr., and Myers BD. Kidney Int 48: 1308-1315, 1995). The prolonged depolarization of the apical membrane may have implications in the pathophysiology of acute renal failure.


Subject(s)
Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/physiopathology , Microtubule-Organizing Center/physiology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Actins/physiology , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Adenosine Triphosphate/metabolism , Animals , Biomarkers , Caco-2 Cells , Cell Polarity/physiology , Centrosome/physiology , Humans , LLC-PK1 Cells , Membrane Potentials/physiology , Microvilli/pathology , Microvilli/physiology , Sodium-Calcium Exchanger/analysis , Swine
10.
Am J Physiol Cell Physiol ; 284(2): C429-38, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12529251

ABSTRACT

The cystic fibrosis transmembrane conductance regulator (CFTR) channel is regulated by cAMP-dependent vesicle traffic and exocytosis to the apical membrane in some cell types, but this has not been demonstrated in the intestinal crypt. The distribution of CFTR, lactase (control), and fluid secretion were determined in rat jejunum after cAMP activation in the presence of nocodazole and primaquine to disrupt vesicle traffic. CFTR and lactase were localized by immunofluorescence, and surface proteins were detected by biotinylation of enterocytes. Immunoprecipitates from biotinylated and nonbiotinylated cells were analyzed by streptavidin detection and immunoblots. Immunolocalization confirmed a cAMP-dependent shift of CFTR but not lactase from a subapical compartment to the apical surface associated with fluid secretion that was reduced in the presence of primaquine and nocodazole. Analysis of immunoblots from immunoprecipitates after biotinylation revealed a 3.8 +/- 1.7-fold (P < 0.005) increase of surface-exposed CFTR after vasoactive intestinal peptide (VIP). These measurements provide independent corroboration supporting a role for vesicle traffic in regulating CFTR and cAMP-induced fluid transport in the intestine.


Subject(s)
Cyclic AMP/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Jejunum/metabolism , Transport Vesicles/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Cell Compartmentation/drug effects , Cell Compartmentation/physiology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Epithelial Cells/drug effects , Exocytosis/drug effects , Exocytosis/physiology , Fluorescent Antibody Technique , Intestinal Mucosa/drug effects , Jejunum/drug effects , Male , Microtubules/drug effects , Microtubules/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Transport Vesicles/drug effects , Vasoactive Intestinal Peptide/pharmacology
11.
J Biol Chem ; 277(40): 37848-54, 2002 Oct 04.
Article in English | MEDLINE | ID: mdl-12151413

ABSTRACT

We have shown previously that centrosomes and other microtubule-organizing centers (MTOCs) attach to the apical intermediate filament (IF) network in CACO-2 cells. In this cell line, intermediate filaments do not disorganize during mitosis. Therefore, we speculated that the trigger of the G(2)-M boundary may also detach MTOCs from their IF anchor. If that was the case, at least one of the proteins involved in the attachment must be phosphorylated by p34(cdc2) (cdk1). Using confocal microscopy and standard biochemical analysis, we found that p34(cdc2)-mediated phosphorylation indeed released MTOCs from IFs in permeabilized cells. In isolated, immunoprecipitated multiprotein complexes containing both gamma-tubulin and cytokeratin 19, p34(cdc2) phosphorylated only one protein, and phosphorylation released cytokeratin 19 from the complexes. We conclude that this as yet unidentified protein is a part of the molecular mechanism that attaches MTOCs to IFs in interphase.


Subject(s)
CDC2 Protein Kinase/metabolism , Intermediate Filaments/metabolism , Intestinal Mucosa/metabolism , CDC2 Protein Kinase/drug effects , Caco-2 Cells , Cell Culture Techniques/methods , Cell Membrane Permeability , Humans , Intermediate Filaments/ultrastructure , Intestinal Mucosa/pathology , Nocodazole/pharmacology , Phosphorylation , Saponins/pharmacology
12.
J Physiol ; 541(Pt 2): 411-21, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12042348

ABSTRACT

The durations of transmembrane action potentials recorded from single myocytes isolated from the endocardial surface of hypertrophied left ventricles of rats were increased, compared to the durations recorded from normal left ventricular cells at 36-37 degrees C. Exposure to phalloidin (1-20 microM, < 20 min), a specific stabilizer of the non-myofibrillar actin microfilament component of the cardiac cytoskeleton, had no effect on action potential duration of normal cells, but significantly shortened the prolonged action potentials of hypertrophied cells. Cytochalasin D (5-50 microM), a disrupter of the actin microfilaments, also had little effect on action potential duration of normal cells. However, cytochalasin D further increased the action potential duration of hypertrophied cells at 10 min exposure. The addition of phalloidin to solutions containing cytochalasin D, reduced the latter's increase of action potential duration in hypertrophied cells. Whole-cell transient outward K(+) current (I(to1)) density was significantly decreased in hypertrophied cells. At a test potential of +60 mV, the mean I(to1) density recorded from normal cells was 13.5 +/- 1.1 pA pF(-1) (n = 18) compared to 4.17 +/- 1.2 pA pF(-1) for LVH cells (n = 22; P < 0.05). Phalloidin (20 microM) increased and cytochalasin D (50 microM) decreased whole-cell I(to1) in hypertrophied cells but had no effect on I(to1), in normal cells. When equimolar concentrations were used, phalloidin, 10 microM, reversed the decrease in I(to1) brought about by cytochalasin D, 10 microM, in hypertrophied cells. The L-type calcium current density was reduced in LVH compared to normal cells. Phalloidin (20 microM) and cytochalasin D (50 microM) had no effect on I(Ca,L) in normal or LVH myocytes. The decrease in I(to1) in hypertrophied cells and the altered I(to1) responsiveness to phalloidin and cytochalasin D reflect modification of I(to1) channel function mediated, in part, through hypertrophy-altered cytoskeletal actin microfilament regulation of I(to1).


Subject(s)
Actins/physiology , Cytoskeleton/physiology , Hypertrophy, Left Ventricular/physiopathology , Microfilament Proteins/physiology , Potassium Channels/physiology , Actins/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Calcium Channels, L-Type/drug effects , Cell Separation , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Electrophysiology , Hypertrophy, Left Ventricular/pathology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microfilament Proteins/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Patch-Clamp Techniques , Phalloidine/pharmacology , Potassium Channels/drug effects , Rats , Rats, Sprague-Dawley
13.
Curr Top Membr Transp ; 24: 251-294, 1985.
Article in English | MEDLINE | ID: mdl-32287478

ABSTRACT

This chapter focuses on protein sorting in the secretory pathway. From primary and secondary biosynthetic sites in the cytosol and mitochondrial matrix, respectively, proteins and lipids are distributed to more than 30 final destinations in membranes or membrane-bound spaces, where they carry out their programmed function. Molecular sorting is defined, in its most general sense, as the sum of the mechanisms that determine the distribution of a given molecule from its site of synthesis to its site of function in the cell. The final site of residence of a protein in a eukaryotic cell is determined by a combination of various factors, acting in concert: (1) site of synthesis, (2) sorting signals or zip codes, (3) signal recognition or decoding mechanisms, (4) cotranslational or posttranslational mechanisms for translocation across membranes, (5) specific fusion-fission interactions between intracellular vesicular compartments, and (6) restrictions to the lateral mobility in the plane of the bilayer. Improvements in cell fractionation, protein separation, and immune precipitation procedures in the past decade have made them possible. Very little is known about the mechanisms that mediate the localization and concentration of specific proteins and lipids within organelles. Various experimental model systems have become available for their study. The advent of recombinant DNA technology has shortened the time needed for obtaining the primary structure of proteins to a few months.

SELECTION OF CITATIONS
SEARCH DETAIL
...