Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Physiol Biochem ; 55(4): 428-448, 2021 Jul 10.
Article in English | MEDLINE | ID: mdl-34242501

ABSTRACT

BACKGROUND/AIMS: Nociceptors detect noxious capsaicin (CAPS) via the transient receptor potential vanilloid 1 (TRPV1) ion channel, but coding mechanisms for relaying CAPS concentration [CAPS] remain obscure. Prolonged (up to 1h.) exposure to CAPS is used clinically to desensitise sensory fibres for treatment of neuropathic pain, but its signalling has typically been studied in cultures of dissociated sensory neurons employing low cell numbers and very short exposure times. Thus, it was pertinent to examine responses to longer CAPS exposures in large populations of adult neurons. METHODS: Confocal fluorescence microscopy was used to monitor the simultaneous excitation by CAPS of neuronal populations in intact L3/4 dorsal root ganglia (DRG) explants from adult pirt-GCaMP3 mice that express a cytoplasmic, genetically-encoded Ca2+ sensor in almost all primary sensory neurons. Peak analysis was performed using GraphPad Prism 9 to deconstruct the heterogenous and complex fluorescence signals observed into informative, readily-comparable measurements: number of signals, their lag time, maximum intensity relative to baseline (Max.) and duration. RESULTS: Exposure for 5 min. to CAPS activated plasmalemmal TRPV1 and led to increased fluorescence due to Ca2+ entry into DRG neurons (DRGNs), as it was prevented by capsazepine or removal of extracellular Ca2+. Increasing [CAPS] (0.3, 1 and 10 µM, respectively) evoked signals from more neurons (123, 275 and 390 from 5 DRG) with shorter average lag (6.4 ± 0.4, 3.3 ± 0.2 and 1.9 ± 0.1 min.) and longer duration (1.4 ± 0.2, 2.9 ± 0.2 and 4.8 ± 0.3 min.). Whilst raising [CAPS] produced a modest augmentation of Max. for individual neurons, those with large increases were selectively expedited; this contributed to a faster onset and higher peak of cumulative fluorescence for an enlarged responding neuronal population. CAPS caused many cells to fluctuate between high and low levels of fluorescence, with consecutive pulses increasing Max. and duration especially when exposure was extended from 5 to 20 min. Such signal facilitation counteracted tachyphylaxis, observed upon repeated exposure to 1 µM CAPS, preserving the cumulative fluorescence over time (signal density) in the population. CONCLUSION: Individual neurons within DRG differed extensively in the dynamics of response to CAPS, but systematic changes elicited by elevating [CAPS] increased signal density in a graded manner, unveiling a possible mechanism for population coding of responses to noxious chemicals. Signal density is sustained during prolonged and repeated exposure to CAPS, despite profound tachyphylaxis in some neurons, by signal facilitation in others. This may explain the burning sensation that persists for several hours when CAPS is used clinically.


Subject(s)
Calcium/metabolism , Capsaicin/pharmacology , Ganglia, Spinal/metabolism , Nociceptors/metabolism , Signal Transduction/drug effects , TRPV Cation Channels/metabolism , Animals , Female , Ganglia, Spinal/cytology , Male , Mice , Mice, Transgenic , Nociceptors/cytology , Signal Transduction/genetics , TRPV Cation Channels/genetics
2.
Int J Mol Sci ; 22(5)2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33806699

ABSTRACT

Nociceptors sense hazards via plasmalemmal cation channels, including transient receptor potential vanilloid 1 (TRPV1). Nerve growth factor (NGF) sensitises TRPV1 to capsaicin (CAPS), modulates nociceptor excitability and induces thermal hyperalgesia, but cellular mechanisms remain unclear. Confocal microscopy was used to image changes in intracellular Ca2+ concentration ([Ca2+]i) across neuronal populations in dorsal root ganglia (DRG) explants from pirt-GCaMP3 adult mice, which express a fluorescent reporter in their sensory neurons. Raised [Ca2+]i was detected in 84 neurons of three DRG explants exposed to NGF (100 ng/mL) and most (96%) of these were also excited by 1 µM CAPS. NGF elevated [Ca2+]i in about one-third of the neurons stimulated by 1 µM CAPS, whether applied before or after the latter. In neurons excitable by NGF, CAPS-evoked [Ca2+]i signals appeared significantly sooner (e.g., respective lags of 1.0 ± 0.1 and 1.9 ± 0.1 min), were much (>30%) brighter and lasted longer (6.6 ± 0.4 vs. 3.9 ± 0.2 min) relative to those non-responsive to the neurotrophin. CAPS tachyphylaxis lowered signal intensity by ~60% but was largely prevented by NGF. Increasing CAPS from 1 to 10 µM nearly doubled the number of cells activated but only modestly increased the amount co-activated by NGF. In conclusion, a sub-population of the CAPS-sensitive neurons in adult mouse DRG that can be excited by NGF is more sensitive to CAPS, responds with stronger signals and is further sensitised by transient exposure to the neurotrophin.


Subject(s)
Calcium Signaling/drug effects , Calcium/metabolism , Capsaicin/pharmacology , Ganglia, Spinal/drug effects , Nerve Growth Factor/pharmacology , Neurons/drug effects , Nociception/drug effects , Animals , Female , Ganglia, Spinal/metabolism , Hyperalgesia/metabolism , Male , Mice , Nerve Growth Factors/metabolism , Neurons/metabolism , Nociceptors/metabolism , Signal Transduction/drug effects , TRPV Cation Channels/metabolism
3.
FASEB J ; 26(12): 5035-48, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22942075

ABSTRACT

Botulinum neurotoxin (BoNT) A or E and tetanus toxin (TeTx) bind to motor-nerve endings and undergo distinct trafficking; their light-chain (LC) proteases cleave soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) peripherally or centrally and cause flaccid or spastic paralysis, respectively. To seek protein domains responsible for local blockade of transmitter release (BoNTs) rather than retroaxonal transport to spinal neurons (TeTx), their acceptor-binding moieties (H(C))--or in one case, heavy chain (HC)--were exchanged by gene recombination. Each chimera, expressed and purified from Escherichia coli, entered rat cerebellar neurons to cleave their substrates, blocked in vitro nerve-induced muscle contractions, and produced only flaccid paralysis in mice. Thus, the local cytosolic delivery of BoNT/A or BoNT/E proteases and the contrasting retrograde transport of TeTx are not specified solely by their HC or H(C); BoNT/A LC translocated locally irrespective of being targeted by either of the latter TeTx domains. In contrast, BoNT/E protease fused to a TeTx enzymatically inactive mutant (TeTIM) caused spastic paralysis and cleaved SNAP-25 in spinal cord but not the injected muscle. Apparently, TeTIM precludes cytosolic release of BoNT/E protease at motor nerve endings. It is deduced that the LCs of the toxins, acting in conjunction with HC domains, dictate their local or distant destinations.


Subject(s)
Botulinum Toxins/metabolism , Paralysis/metabolism , Recombinant Fusion Proteins/metabolism , Tetanus Toxin/metabolism , Animals , Blotting, Western , Botulinum Toxins/genetics , Botulinum Toxins/pharmacokinetics , Cerebellum/metabolism , Mice , Mutation , Neuromuscular Diseases/metabolism , Neurons/metabolism , Neurotoxins/genetics , Neurotoxins/metabolism , Neurotoxins/pharmacokinetics , Peptide Hydrolases/metabolism , Protein Transport , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Sciatic Nerve/physiopathology , Sciatic Nerve/surgery , Spinal Cord/metabolism , Synaptosomal-Associated Protein 25/metabolism , Tetanus Toxin/genetics , Tetanus Toxin/pharmacokinetics
4.
FEBS J ; 279(14): 2555-67, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22607388

ABSTRACT

A targeted drug carrier (TDC) is described for transferring functional proteins or peptides into motor nerve terminals, a pivotal locus for therapeutics to treat neuromuscular disorders. It exploits the pronounced selectivity of botulinum neurotoxin type B (BoNT/B) for interacting with acceptors on these cholinergic nerve endings and becoming internalized. The gene encoding an innocuous BoNT/B protease-inactive mutant (BoTIM) was fused to that for core streptavidin, expressed in Escherichia coli and the purified protein was conjugated to surface-biotinylated liposomes. Such decorated liposomes, loaded with fluorescein as traceable cargo, acquired pronounced specificity for motor nerve terminals in isolated mouse hemidiaphragms and facilitated the intraneuronal transfer of the fluor, as revealed by confocal microscopy. Delivery of the protease light chain of botulinum neurotoxin type A (BoNT/A) via this TDC accelerated the onset of neuromuscular paralysis, indicative of improved translocation of this enzyme into the presynaptic cytosol with subsequent proteolytic inactivation of synaptosomal-associated protein of molecular mass 25 kDa (SNAP-25), an exocytotic soluble N-ethyl-maleimide-sensitive factor attachment protein receptor (SNARE) essential for neurotransmitter release. BoTIM-coupled liposomes, loaded with peptide inhibitors of proteases, yielded considerable attenuation of the neuroparalytic effects of BoNT/A or BoNT/F as a result of their cytosolic transfer, the first in situ demonstration of the ability of designer antiproteases to suppress the symptoms of botulism ex vivo. Delivery of the BoNT/A inhibitor by liposomes targeted with the full-length BoTIM proved more effective than that mediated by its C-terminal neuroacceptor-binding domain. This demonstrated versatility of TDC for nonviral cargo transfer into cholinergic nerve endings has unveiled its potential for direct delivery of functional targets into motor nerve endings.


Subject(s)
Botulinum Toxins/metabolism , Drug Delivery Systems/methods , Presynaptic Terminals/metabolism , Protease Inhibitors/administration & dosage , SNARE Proteins/metabolism , Animals , Botulinum Toxins/chemistry , Botulinum Toxins/genetics , Botulinum Toxins, Type A , Diaphragm/metabolism , Fluorescein/chemistry , Humans , Liposomes , Mice , Microscopy, Confocal , Motor Neurons/metabolism , Mutation , Nerve Endings/metabolism , Paralysis/metabolism , Paralysis/therapy , Protease Inhibitors/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Streptavidin/genetics , Streptavidin/metabolism , Synaptosomal-Associated Protein 25/metabolism
5.
Biochem J ; 444(1): 59-67, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22360156

ABSTRACT

Various human neurogenic hyper-excitability disorders are successfully treated with type A or B BoNT (botulinum neurotoxin). The BoNT/A complex is widely used because of its longer-lasting benefits; also, autonomic side-effects are more often reported for BoNT/B. To establish if this distinct effect of BoNT/B could be exploited therapeutically, BoNT/A was modified so that it would bind the more abundant BoNT/B acceptor in rodents while retaining its desirable persistent action. The advantageous protease and translocation domain of BoNT/A were recombinantly combined with the acceptor-binding moiety of type B [H(C)/B (C-terminal half of BoNT/B heavy chain)], creating the chimaera AB. This purified protein bound the BoNT/B acceptor, displayed enhanced capability relative to type A for intraneuronally delivering its protease, cleaved SNAP-25 (synaptosome-associated protein of 25 kDa) and induced a more prolonged neuromuscular paralysis than BoNT/A in mice. The BA chimaera, generated by substituting H(C)/A (C-terminal half of BoNT/A heavy chain) into BoNT/B, exhibited an extremely high specific activity, delivered the BoNT/B protease via the BoNT/A acceptor into neurons, or fibroblast-like synoviocytes that lack SNAP-25, cleaving the requisite isoforms of VAMP (vesicle-associated membrane protein). Both chimaeras inhibited neurotransmission in murine bladder smooth muscle. BA has the unique ability to reduce exocytosis from non-neuronal cells expressing the BoNT/A-acceptor and utilising VAMP, but not SNAP-25, in exocytosis.


Subject(s)
Botulinum Toxins, Type A/genetics , Botulinum Toxins/genetics , Exocytosis/drug effects , Recombinant Fusion Proteins/pharmacology , Animals , Cells, Cultured , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , In Vitro Techniques , Mice , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Neuromuscular Junction/drug effects , Neuromuscular Junction/physiopathology , Neurons/drug effects , Neurons/metabolism , Paralysis/chemically induced , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Phrenic Nerve/drug effects , Phrenic Nerve/physiopathology , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Synaptic Transmission/drug effects , Synaptosomal-Associated Protein 25/metabolism , Synovial Membrane/cytology , Urinary Bladder/drug effects , Urinary Bladder/physiology , Vesicle-Associated Membrane Protein 2/genetics , Vesicle-Associated Membrane Protein 2/metabolism
6.
FEBS J ; 278(23): 4454-66, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21645262

ABSTRACT

A major unmet clinical need exists for long-acting neurotherapeutics to alleviate chronic pain in patients unresponsive to available nonaddictive analgesics. Herein, a new strategy is described for the development of potent and specific inhibitors of the neuronal exocytosis of transmitters and pain mediators that exhibit unique antinociceptive activity. This entailed recombinant production in Escherichia coli of two serotypes of botulinum neurotoxin (BoNT) (BoNT(A) and BoNT(E) ), which are proteins that are known to block the release of transmitters by targeting and entering nerve endings, where their proteases cleave and inactivate a protein, synaptosomal protein of M(r) 25 000 (SNAP-25), that is essential for Ca(2+) -regulated exocytosis. Site-directed mutagenesis of Leu428 and Leu429 in BoNT(A) revealed that the remarkable longevity of its neuroparalytic action is attributable to a dileucine-containing motif. BoNT(E) acts transiently, because it lacks these residues, but is a superior inhibitor of transient receptor potential vanilloid type 1-mediated release of pain peptides from sensory nerves. The advantageous features of each serotype were harnessed by attaching the BoNT(E) protease moiety to an enzymically inactive mutant of BoNT(A) . The resultant purified composite protein could target motoneurons by binding to the BoNT(A) ectoacceptor and persistently produce BoNT(E) -truncated SNAP-25. As this enzyme lasted for more than 1 month (as compared with 5 days for BoNT(E) alone), such a dramatic extension in the lifetime of this BoNT(E) protease is attributable to a stabilizing influence of the BoNT(A) mutant. Most importantly, injecting this novel biotherapeutic into the foot pads of rats resulted in extended amelioration of inflammatory pain. Thus, a new generation of biotherapeutics has been created with the potential to give long-term relief of pain.


Subject(s)
Analgesics/pharmacology , Botulinum Toxins/pharmacology , Chronic Pain/drug therapy , Neuromuscular Agents/pharmacology , Neurotoxins/pharmacology , Analgesics/therapeutic use , Animals , Botulinum Toxins/genetics , Botulinum Toxins/metabolism , Botulinum Toxins, Type A/genetics , Botulinum Toxins, Type A/metabolism , Botulinum Toxins, Type A/pharmacology , Chronic Pain/metabolism , Exocytosis , Humans , Leucine/genetics , Mutagenesis, Site-Directed , Mutation , Neuromuscular Agents/therapeutic use , Neurotoxins/genetics , Neurotoxins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , SNARE Proteins/genetics , SNARE Proteins/metabolism , Synaptosomal-Associated Protein 25/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
7.
J Biol Chem ; 286(8): 6375-85, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21138836

ABSTRACT

Blockade of neurotransmitter release by botulinum neurotoxin type A (BoNT(A)) underlies the severe neuroparalytic symptoms of human botulism, which can last a few years. The structural basis for this remarkable persistence remains unclear. Herein, recombinant BoNT(A) was found to match the neurotoxicity of that from Clostridium botulinum, producing persistent cleavage of synaptosomal-associated protein of 25 kDa (SNAP-25) and neuromuscular paralysis. When two leucines near the C terminus of the protease light chain of A (LC(A)) were mutated, its inhibition of exocytosis was followed by fast recovery of intact SNAP-25 in cerebellar neurons and neuromuscular transmission in vivo. Deletion of 6-7 N terminus residues diminished BoNT(A) activity but did not alter the longevity of its SNAP-25 cleavage and neuromuscular paralysis. Furthermore, genetically fusing LC(E) to a BoNT(A) enzymically inactive mutant (BoTIM(A)) yielded a novel LC(E)-BoTIM(A) protein that targets neurons, and the BoTIM(A) moiety also delivers and stabilizes the inhibitory LC(E), giving a potent and persistent cleavage of SNAP-25 with associated neuromuscular paralysis. Moreover, its neurotropism was extended to sensory neurons normally insensitive to BoNT(E). LC(E-)BoTIM(A)(AA) with the above-identified dileucine mutated gave transient neuromuscular paralysis similar to BoNT(E), reaffirming that these residues are critical for the persistent action of LC(E)-BoTIM(A) as well as BoNT(A). LC(E)-BoTIM(A) inhibited release of calcitonin gene-related peptide from sensory neurons mediated by transient receptor potential vanilloid type 1 and attenuated capsaicin-evoked nociceptive behavior in rats, following intraplantar injection. Thus, a long acting, versatile composite toxin has been developed with therapeutic potential for pain and conditions caused by overactive cholinergic nerves.


Subject(s)
Botulinum Toxins, Type A/pharmacology , Cerebellum/metabolism , Leucine , Neuromuscular Agents/pharmacology , Recombinant Fusion Proteins/pharmacology , Sensory Receptor Cells/metabolism , Synaptic Transmission/drug effects , Animals , Botulinum Toxins, Type A/genetics , Calcitonin/genetics , Calcitonin/metabolism , Cerebellum/cytology , Female , Male , Mice , Mutation , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Sensory Receptor Cells/cytology , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism , Time Factors
8.
J Biol Chem ; 283(25): 16993-7002, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18400760

ABSTRACT

Hyperexcitability disorders of cholinergically innervated muscles are treatable with botulinum neurotoxin (BoNT) A. The seven serotypes (A-G) potently block neurotransmission by binding to presynaptic receptors, undergoing endocytosis, transferring to the cytosol, and inactivating proteins essential for vesicle fusion. Although BoNT/A and BoNT/E cleave SNAP-25, albeit at distinct sites, BoNT/E blocks neurotransmission faster and more potently. To identify the domains responsible for these characteristics, the C-terminal heavy chain portions of BoNT/A and BoNT/E were exchanged to create chimeras AE and EA. After high yield expression in Escherichia coli, these single chain chimeras were purified by two-step chromatography and activated by conversion to disulfide-linked dichains. In vitro, each entered neurons, cleaved SNAP-25, and blocked neuromuscular transmission while causing flaccid paralysis in vivo. Acidification-dependent translocation of the light chain to the cytosol occurred more rapidly for BoNT/E and EA than for BoNT/A and AE because the latter pair remained susceptible for longer to inhibitors of the vesicular proton pump, and BoNT/A proved less sensitive. The receptor-binding and protease domains do not seem to be responsible for the speeds of intoxication; rather the N-terminal halves of their heavy chains are implicated, with dissimilar rates of cytosolic transfer of the light chains being due to differences in pH sensitivity. AE produced the most persistent muscle weakening and therefore has therapeutic potential. Thus, proof of principle is provided for tailoring the pharmacological properties of these toxins by protein engineering.


Subject(s)
Botulinum Toxins, Type A/chemistry , Botulinum Toxins/chemistry , Gene Expression Regulation , Animals , Cells, Cultured , Cytosol/metabolism , Escherichia coli/metabolism , Hydrogen-Ion Concentration , Mice , Neurons/metabolism , Protein Engineering/methods , Protein Structure, Tertiary , Protein Transport , Protons , Recombinant Fusion Proteins/chemistry
9.
BJU Int ; 100(5): 1075-80, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17784887

ABSTRACT

OBJECTIVE To monitor the presence and cleavage of synaptosomal-associated protein of 25 kDa (SNAP-25) by botulinum toxin type A (botox-A), in human detrusor muscle, as the effects of botox-A in the urinary bladder last significantly longer than when applied for disorders of striated muscles. PATIENTS AND METHODS Tissue samples were obtained from eight patients with end-stage neurogenic bladder at different times after injection with botox-A. The resected bladder domes were examined using biochemical and immunohistological techniques. RESULTS The presence of intact SNAP-25 in human bladder was detected, for the first time, in all samples by both Western blotting and immunofluorescence. By contrast, detection of a band potentially representing toxin-cleaved SNAP-25(A) required its enrichment by precipitation with a specific antibody. This putative product was present in four of six patients treated with botox-A 5 weeks to 11 months previously, but could not be detected in one patient 30 months after botox injection, and in an untreated control. Fluorescence microscopy showed no obvious effects of the toxin treatment on the presence and pattern of SNAP-25-positive neurones. CONCLUSIONS A limited amount of SNAP-25 appears to be cleaved in nerves that innervate the smooth detrusor muscle in most patients who had been injected with botox-A; its absolute identification was precluded by the sensitivity of the detection. This protein was detectable much longer after toxin treatment than published for rodent striated muscle, and thus could contribute to the clinically reported longer duration of the effectiveness of botox-A.


Subject(s)
Botulinum Toxins, Type A/therapeutic use , Meningomyelocele/complications , Muscle, Smooth/drug effects , Neuromuscular Agents/therapeutic use , Synaptosomal-Associated Protein 25/metabolism , Urinary Bladder, Neurogenic/drug therapy , Adolescent , Adult , Aged , Blotting, Western , Child , Female , Humans , Immunohistochemistry , Male , Multiple Sclerosis/complications , Muscle, Smooth/pathology , Sensitivity and Specificity , Urinary Bladder, Neurogenic/pathology , Urodynamics
10.
Methods Enzymol ; 403: 499-512, 2005.
Article in English | MEDLINE | ID: mdl-16473615

ABSTRACT

The Rab family of small GTPases are key regulators of membrane trafficking in eukaryotic cells. Rab11, one member of this family, plays a role in regulating various cellular functions such as plasma membrane recycling, phagocytosis, and cytokinesis. A family of Rab11-binding proteins has been identified and termed the Rab11 family interacting proteins or Rab11-FIPs. Rab11-FIP3, a member of this Rab11-binding protein family, in addition to interacting with Rab11, is also capable of interaction with members of the ADP-Ribosylation Factor (ARF) GTPase family. Here we describe the purification of Rab11-FIP3 and report its biological properties in eukaryotic cells as visualized by immunofluorescence microscopy.


Subject(s)
Carrier Proteins/isolation & purification , Carrier Proteins/physiology , Amino Acid Sequence , Base Sequence , Carrier Proteins/chemistry , Carrier Proteins/metabolism , DNA Primers , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Molecular Sequence Data , Plasmids , Protein Binding , Subcellular Fractions/metabolism
11.
Biochem Biophys Res Commun ; 319(1): 83-94, 2004 Jun 18.
Article in English | MEDLINE | ID: mdl-15158446

ABSTRACT

The Rab11-family interacting protein 3 (Rab11-FIP3), also known as Arfophilin and Eferin, is a Rab11 and ADP-ribosylation factor (ARF) binding protein of unknown function. Here, we sought to investigate the subcellular localisation and elucidate the function of Rab11-FIP3 in eukaryotic membrane trafficking. Utilising a polyclonal antibody specific for Rab11-FIP3, we have demonstrated by immunofluorescence microscopy that Rab11-FIP3 colocalises with Rab11 in a distinctive pericentrosomal location in A431 cells. Additionally, we found that Rab11-FIP3 localises to punctate vesicular structures dispersed throughout A431 cells. We have demonstrated that both Rab11 and Rab11-FIP3 localise to the cleavage furrow during cytokinesis, and that Rab11-FIP3 localisation is dependent on both microtubule and actin filament integrity. We show that Rab11-FIP3 does not enter brefeldin A (BFA) induced membrane tubules that are positive for the transferrin receptor (TfnR). Furthermore, we show that expression of an amino-terminally truncated mutant of Rab11-FIP3 (Rab11-FIP3((244-756))) does not inhibit transferrin (Tfn) recycling in HeLa cells. It is likely that Rab11-FIP3 is involved in trafficking events other than Tfn trafficking; these may include the transport of endosomally derived membrane to the cleavage furrow during cytokinesis.


Subject(s)
Carrier Proteins/chemistry , Centrosome/ultrastructure , rab GTP-Binding Proteins/chemistry , ADP-Ribosylation Factors/metabolism , Actins/chemistry , Amino Acid Sequence , Brefeldin A/pharmacology , Cell Division , Cell Line, Tumor , Centrosome/metabolism , Cloning, Molecular , DNA, Complementary/metabolism , Endosomes/metabolism , HeLa Cells , Humans , Immunoblotting , Interphase , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Synthesis Inhibitors/pharmacology , Receptors, Transferrin/chemistry , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Transfection , Transferrin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...