Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
1.
Poult Sci ; 102(10): 102841, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37480657

ABSTRACT

Necrotic enteritis (NE) is a widespread infectious disease caused by Clostridium perfringens that inflicts major economic losses on the global poultry industry. Due to regulations on antibiotic use in poultry production, there is an urgent need for alternative strategies to mitigate the negative effects of NE. This paper presents a passive immunization technology that utilizes hyperimmune egg yolk immunoglobulin Y (IgY) specific to the major immunodominant antigens of C. perfringens. Egg yolk IgYs were generated by immunizing hens with 4 different recombinant C. perfringens antigens, and their protective effects against NE were evaluated in commercial broilers. Six different spray-dried egg powders were produced using recombinant C. perfringens antigens: α-toxin, NE B-like toxin (NetB; EB), elongation factor-Tu (ET), pyruvate:ferredoxin oxidoreductase, a mixture of 4 antigens (EM-1), and a nonimmunized control (EC). The challenged groups were either provided with different egg powders at a 1% level or no egg powders (EN). The NE challenge model based on Eimeria maxima and C. perfringens dual infection was used. In Experiments 1 and 2, the EB and ET groups exhibited increased body weight gain (BWG; P < 0.01), decreased NE lesion scores (P < 0.001), and reduced serum NetB levels (P < 0.01) compared to the EN and EC groups. IgY against NetB significantly reduced Leghorn male hepatocellular cytotoxicity in an in vitro test (P < 0.01). In Experiment 3, the protective effect of the IgYs mixture (EM-2) against C. perfringens antigens (NetB and EFTu) and Eimeria antigens (elongation factor-1-alpha: EF1α and Eimeria profilin: 3-1E) was tested. The EM-2 group showed similar body weight, BWG, and feed intake from d 7 to 22 compared to the NC group (P < 0.05). On d 20, the EM-2 group showed comparable intestinal permeability, NE lesion scores, and jejunal NetB and collagen adhesion protein levels to the NC group (P < 0.05). In conclusion, dietary mixture containing antibodies to NetB and EFTu provides protection against experimental NE in chickens through passive immunization.


Subject(s)
Bacterial Toxins , Clostridium Infections , Eimeria , Enteritis , Poultry Diseases , Animals , Female , Male , Clostridium perfringens , Chickens/metabolism , Bacterial Toxins/metabolism , Clostridium Infections/prevention & control , Clostridium Infections/veterinary , Egg Yolk/metabolism , Enteritis/prevention & control , Enteritis/veterinary , Antibodies, Bacterial , Body Weight , Peptide Elongation Factors/metabolism , Poultry Diseases/prevention & control , Necrosis/veterinary
2.
Arthritis Res Ther ; 21(1): 182, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31370858

ABSTRACT

BACKGROUND: Patients with rheumatoid arthritis (RA) experience extra-articular manifestations including osteoporosis and muscle wasting, which closely associate with severity of disease. Whilst therapeutic glucocorticoids (GCs) reduce inflammation in RA, their actions on muscle and bone metabolism in the context of chronic inflammation remain unclear. We utilised the TNF-tg model of chronic polyarthritis to ascertain the impact of therapeutic GCs on bone and muscle homeostasis in the context of systemic inflammation. METHODS: TNF-tg and wild-type (WT) animals received either vehicle or the GC corticosterone (100 µg/ml) in drinking water at onset of arthritis. Arthritis severity and clinical parameters were measured, serum collected for ELISA and muscle and bone biopsies collected for µCT, histology and mRNA analysis. In vivo findings were examined in primary cultures of osteoblasts, osteoclasts and myotubes. RESULTS: TNF-tg mice receiving GCs showed protection from inflammatory bone loss, characterised by a reduction in serum markers of bone resorption, osteoclast numbers and osteoclast activity. In contrast, muscle wasting was markedly increased in WT and TNF-tg animals receiving GCs, independently of inflammation. This was characterised by a reduction in muscle weight and fibre size, and an induction in anti-anabolic and catabolic signalling. CONCLUSIONS: This study demonstrates that when given in early onset chronic polyarthritis, oral GCs partially protect against inflammatory bone loss, but induce marked muscle wasting. These results suggest that in patients with inflammatory arthritis receiving GCs, the development of interventions to manage deleterious side effects in muscle should be prioritised.


Subject(s)
Arthritis/drug therapy , Bone Resorption/prevention & control , Corticosterone/therapeutic use , Muscle Cells/pathology , Muscular Atrophy/prevention & control , Osteoblasts/pathology , Osteoclasts/pathology , Animals , Arthritis/diagnosis , Arthritis/metabolism , Biopsy , Bone Resorption/metabolism , Bone Resorption/pathology , Cells, Cultured , Chronic Disease , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Glucocorticoids/therapeutic use , Mice , Mice, Inbred C57BL , Muscle Cells/drug effects , Muscle Cells/metabolism , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism
3.
Osteoarthritis Cartilage ; 22(5): 609-21, 2014 May.
Article in English | MEDLINE | ID: mdl-24632293

ABSTRACT

Osteoarthritis (OA) is an age-related condition and the leading cause of pain, disability and shortening of adult working life in the UK. The incidence of OA increases with age, with 25% of the over 50s population having OA of the knee. Despite promising preclinical data covering various molecule classes, there is regrettably at present no approved disease-modifying OA drugs (DMOADs). With the advent of next generation sequencing technologies, other therapeutic areas, in particular oncology, have experienced a paradigm shift towards defining disease by its molecular composition. This paradigm shift has enabled high resolution patient stratification and supported the emergence of personalised or precision medicines. In this review we evaluate the potential for the development of OA therapeutics to undergo a similar paradigm shift given that OA is increasingly being recognised as a heterogeneous disease affecting multiple joint tissues. We highlight the evidence for the role of these tissues in OA pathology as different "hallmarks" of OA biology and review the opportunities to identify and develop targeted disease-modifying pharmacological therapeutics. Finally, we consider whether it is feasible to expect the emergence of personalised disease-modifying medicines for patients with OA and how this might be achieved.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Molecular Targeted Therapy/methods , Osteoarthritis/drug therapy , Precision Medicine/methods , Adipose Tissue/drug effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bone Remodeling/drug effects , Humans , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , Osteoarthritis/physiopathology
4.
Phys Chem Chem Phys ; 15(40): 17268-76, 2013 Oct 28.
Article in English | MEDLINE | ID: mdl-24019182

ABSTRACT

A study of the mechanism of ethanol oxidation in alkaline solution on a platinum electrode modified with an irreversibly-deposited layer of lead has been carried out using in situ FTIR spectroscopy. This study provides support for the suggestion that the adsorption mechanism of ethanol is substantially modified in the presence of Pb, with a carbon-bonded intermediate being favoured leading to facile scission of the C-C bond in ethanol. We have found that the formation of carbonate takes place at potentials close to the thermodynamic value. At higher potentials, when Pb is lost to solution, the mechanism of oxidation of ethanol reverts to that found on a normal polycrystalline Pt surface, with the primary product being acetate.


Subject(s)
Electrodes , Ethanol/chemistry , Hydroxides/chemistry , Lead/chemistry , Platinum/chemistry , Potassium Compounds/chemistry , Spectroscopy, Fourier Transform Infrared , Electrochemistry , Oxidation-Reduction , Water/chemistry
5.
J Hum Hypertens ; 27(5): 321-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23076450

ABSTRACT

Most patients with hypertension need more than one drug to achieve blood pressure (BP) control. This randomized, double-blind, multifactorial study evaluated whether combinations of aliskiren and amlodipine provided superior BP reductions to component monotherapies in patients with hypertension (mean sitting diastolic BP (msDBP) 95-<110 mm Hg). Overall, 1688 patients were randomized to once-daily monotherapy with aliskiren 150 or 300 mg or amlodipine 5 or 10 mg, combination therapy with one of four corresponding aliskiren/amlodipine doses, or placebo for 8 weeks. At week 8 end point, aliskiren/amlodipine combinations provided significant msDBP reductions from baseline of 14.0-16.5 mm Hg, compared with reductions of 8.0 and 10.2 mm Hg for aliskiren 150 and 300 mg, respectively (P<0.001), and 11.0 and 13.8 mm Hg for amlodipine 5 and 10 mg, respectively (P<0.05). Aliskiren/amlodipine combinations provided reductions in mean sitting systolic BP 20.6-23.9 mm Hg, compared with decreases of 10.7 and 15.4 mm Hg for aliskiren 150 and 300 mg, respectively (P<0.001), and 15.8 and 21.0 mm Hg for amlodipine 5 (P< or =0.001) and 10 mg (P=NS), respectively. Aliskiren/amlodipine combination therapy provides greater BP lowering than either agent alone, hence offering an effective treatment option for patients with hypertension.


Subject(s)
Amides/administration & dosage , Amlodipine/administration & dosage , Calcium Channel Blockers/administration & dosage , Fumarates/administration & dosage , Hypertension/drug therapy , Renin/antagonists & inhibitors , Adult , Aged , Amides/adverse effects , Amides/therapeutic use , Amlodipine/adverse effects , Amlodipine/therapeutic use , Blood Pressure/drug effects , Blood Pressure Monitoring, Ambulatory , Double-Blind Method , Drug Therapy, Combination , Female , Fumarates/adverse effects , Fumarates/therapeutic use , Humans , Male , Middle Aged
6.
Osteoarthritis Cartilage ; 18(4): 555-62, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20060953

ABSTRACT

OBJECTIVE: To examine whether beta2-adrenergic agonist-induced hypertrophy of the quadriceps skeletal muscle can modulate the severity of osteoarthritis (OA) in the rodent meniscectomy (MNX) model. METHODS: Male Lewis rats were subcutaneously administered with 1.5 mg/kg/day clenbuterol hydrochloride (n=15) or saline vehicle (n=20) for 14 days. Following pre-treatment, five animals from each group were sacrificed to assess the immediate effects of clenbuterol. The remaining animals underwent either invasive knee surgery (clenbuterol pre-treated n=10; saline pre-treated n=10) or a sham control surgical procedure (saline pre-treated n=5). During disease initiation and progression, weight bearing was assessed by hindlimb loading. Myosin heavy chain (MHC) protein isoforms were quantified by silver stained SDS PAGE. OA severity was graded by assessment of toluidine blue stained step coronal sections of the total knee joint. RESULTS: Clenbuterol treatment resulted in an increase in total bodyweight, growth rate and in quadriceps skeletal muscle mass. Meniscal surgery resulted in the development of OA-like lesions, changes to weight bearing, and changes in MHC protein expression in the quadriceps. Clenbuterol-induced skeletal muscle hypertrophy had no effect on either weight bearing or articular pathology following MNX surgery. CONCLUSIONS: Our data reveal that clenbuterol-induced skeletal muscle hypertrophy is unable to mimic the beneficial clinical effects of increased musculature derived through targeted strength training in humans, in a rodent model of MNX-induced OA. In addition we observed fibre-type switching to "slow twitch" in the quadriceps muscle during the induction of OA that warrants further investigation as to its relationship to joint stability.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Clenbuterol/analogs & derivatives , Clenbuterol/pharmacology , Hypertrophy/chemically induced , Muscle, Skeletal/drug effects , Osteoarthritis/drug therapy , Quadriceps Muscle/drug effects , Animals , Body Weight/drug effects , Disease Models, Animal , Male , Muscle, Skeletal/pathology , Myosin Heavy Chains/analysis , Osteoarthritis/physiopathology , Quadriceps Muscle/pathology , Rats , Rats, Inbred Lew/growth & development , Weight-Bearing/physiology
7.
Osteoarthritis Cartilage ; 17(4): 464-72, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19008124

ABSTRACT

OBJECTIVE: To identify differentially expressed microRNAs (miRNAs) in human osteoarthritic (OA) cartilage and bone tissue and to determine their relevance to chondrocyte function. METHODS: Cartilage and bone was obtained from OA patients who underwent total knee joint replacement surgery or from post-mortem patients with no previous history of OA. MiRNA expression was quantified by real-time PCR (RT-PCR). Functional pathway analysis of miRNA was performed using Ingenuity Pathway Analysis. Primary chondrocytes were isolated by collagenase digestion and transfected with miRNA mimics and miRNA inhibitors using cationic lipid. Tumour Necrosis Factor-alpha (TNF-alpha) and Matrix metalloprotease 13 (MMP13) protein levels were measured by Enzyme-Linked ImmunoSorbent Assay (ELISA). RESULTS: In total we identified 17 miRNA that showed greater than 4-fold differential expression between OA and normal cartilage, and 30 miRNA that showed greater than 4-fold differential expression in OA bone. Functional pathway analysis of the predicted gene targets for miR-9, miR-98, which were upregulated in both OA bone and cartilage tissue, and miR-146, which was downregulated in OA cartilage, suggested that these miRNA mediate inflammatory functions and pathways. Over-expression of miR-9, miR-98 or miR-146 in isolated human chondrocytes reduced interleukin-1 beta (IL-1 beta) induced TNF-alpha production. Furthermore, inhibition and over-expression of miR-9 modulated MMP13 secretion. CONCLUSIONS: We have identified a number of differentially expressed miRNAs in late-stage human OA cartilage and bone. Functional analysis of miR-9, miR-98 and miR-146 in primary chondrocytes suggests a role in mediating the IL-1 beta induced production of TNF-alpha. MiR-9, upregulated in OA tissue, was found to inhibit secretion of the collagen type II-targeting metalloproteinase MMP13 in isolated human chondrocytes.


Subject(s)
Matrix Metalloproteinase 13/biosynthesis , MicroRNAs/physiology , Osteoarthritis, Knee/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Aged , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/metabolism , Computational Biology/methods , Gene Expression Profiling/methods , Gene Expression Regulation , Humans , Inflammation Mediators/physiology , MicroRNAs/genetics , Middle Aged , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Signal Transduction/genetics , Signal Transduction/physiology
8.
Osteoarthritis Cartilage ; 17(1): 124-31, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18562219

ABSTRACT

OBJECTIVE: To examine the role of mitogen-activated protein kinase-activated protein kinase 2 (MK2) in mediating the cellular response to pro-inflammatory cytokines in human primary osteoarthritis (OA) chondrocytes. METHODS: Delivery of a dominant negative MK2 was achieved in HeLa cells by adenoviral infection. Cellular heat shock protein (HSP27) activity was determined using a Bioplex assay. Primary OA chondrocytes were isolated by collagenase digestion of human articular cartilage. Phosphorylated MK2 was detected by immunoblotting and immunohistology. Transfection of primary chondrocytes with siRNA was achieved using cationic lipid and gene expression determined by real-time polymerase chain reaction. Production of prostaglandin E2 (PGE2) and matrixmetalloproteases (MMPs) was measured by enzyme-linked immunosorbent assay. RESULTS: Over-expression of a dominant negative MK2 inhibited HSP27 phosphorylation and significantly reduced both interleukin 1 (IL-1)beta and tumour necrosis factor (TNF)-alpha mediated release of PGE2 in HeLa cells over a 24h period. Phosphorylated MK2 was detected in OA articular cartilage and in isolated primary OA chondrocytes, where it was induced by IL-1beta. Transfection of OA chondrocytes with MK2 siRNA antisense significantly reduced both basal and IL-1beta induced PGE2 release. siRNA mediated MK2 knockdown also significantly reduced both basal and IL-1beta induced MMP13 expression and MMP13 and MMP3 protein release but had no effect on MMP1. CONCLUSIONS: Our data reveal that MK2 is active in OA human articular cartilage and in isolated primary human chondrocytes and that MK2 mediates the release of PGE2, MMP3 and MMP13. These findings suggest a role for MK2 in contributing to OA algesia and OA joint structural deterioration by mediating the downstream effects of p38 activation on PGE2 release and the expression and release of catabolic proteases.


Subject(s)
Cartilage, Articular/enzymology , Chondrocytes/enzymology , Intracellular Signaling Peptides and Proteins/physiology , Osteoarthritis, Knee/enzymology , Protein Serine-Threonine Kinases/physiology , Adenoviridae/genetics , Aged , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/pathology , Dinoprostone/metabolism , Enzyme Activation/drug effects , Genetic Vectors , Humans , Interleukin-1beta/pharmacology , MAP Kinase Signaling System , Male , Middle Aged , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Peptide Hydrolases/biosynthesis , Phosphorylation , RNA, Small Interfering/genetics , Transfection
9.
Article in English | MEDLINE | ID: mdl-18003224

ABSTRACT

We present a discussion of the use of vertically-aligned carbon nanofibers (VACNFs) as nanoscale elements that directly interface to biological whole-cell systems. VACNFs are compatible with a large subset of microfabrication processes, thereby enabling their incorporation into mesoscale hybrid systems that provide addressability of the VACNFs as either bulk electrode material, or as individually addressed nanoelectrodes. These VACNF devices are compatible with cell cultures, and electrochemical addressability of nanofibers can be maintained for extended periods within cell cultures. We present results that demonstrate possible use of VACNF devices as electrical and genetic substrates for tissue scaffolding applications.


Subject(s)
Cell Culture Techniques/methods , Drug Carriers/chemistry , Electric Stimulation/methods , Electroporation/methods , Nanotubes, Carbon/chemistry , Nanotubes, Carbon/ultrastructure , Tissue Culture Techniques/methods , Transfection/methods , Materials Testing , Particle Size
10.
Osteoarthritis Cartilage ; 14(6): 597-608, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16647866

ABSTRACT

OBJECTIVE: RDC1 is a class A orphan G-protein coupled receptor of unknown function. The purpose of this study was to identify compound RDC1 agonists and use these as tools to determine the effect of RDC1 activation in human chondrocytes and cartilage explant tissue. METHODS: Computational chemistry was employed to build a homology model of the RDC1 receptor. A virtual screen of in-house compounds was then performed and positive hits screened for their ability to invoke a Ca2+ response in a recombinant RDC1 HEK293 cell line, as measured by FLIPR. The effect of RDC1 activation on human chondrocytes and cartilage explant gene expression was determined by quantitative real-time polymerase chain reaction (PCR), and these effects validated as being mediated by RDC1 using siRNA antisense. RESULTS: Tissue expression profiling demonstrated that RDC1 expression was predominant in cartilage tissue. Treatment of human primary chondrocytes with RDC1 agonist induced a Ca2+ response, suggesting the receptor is active in this tissue type. Treatment for 24h with RDC1 agonist led to altered expression of a number of genes associated with chondrocyte hypertrophy and increased matrix degradation in human primary chondrocytes, and elevated total matrix metalloproteinase (MMP) activity in cartilage explant. Transfection with RDC1 siRNA caused a >90% reduction in human primary chondrocyte RDC1 expression and significantly reduced the impact of RDC1 agonist on the previously identified RDC1-regulated genes. CONCLUSIONS: RDC1 activation in human chondrocytes and cartilage explant leads to changes in gene expression and activity associated with chondrocyte hypertrophy, angiogenesis and increased matrix degradation, suggesting signalling via the RDC1 receptor may play an important role in the early development of osteoarthritis (OA).


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Osteoarthritis, Knee/metabolism , Receptors, G-Protein-Coupled/metabolism , Calcium/metabolism , Cartilage, Articular/pathology , Cell Line , Chondrocytes/pathology , Extracellular Matrix/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Humans , Hypertrophy , Immunohistochemistry/methods , Matrix Metalloproteinases/analysis , Neovascularization, Pathologic/metabolism , Receptors, CXCR , Receptors, G-Protein-Coupled/agonists , Recombinant Proteins/metabolism , Signal Transduction/physiology , Transfection
11.
Meat Sci ; 69(1): 17-25, 2005 Jan.
Article in English | MEDLINE | ID: mdl-22062635

ABSTRACT

The present study investigated the relationship between fibre type distribution and slow (MHC-s) and fast (MHC-f) myosin heavy chain content on calpastatin and meat tenderness in longissimus dorsi (LD), tensor fasciae latae (TFL), semitendinosus (ST), trapezius (TZ) and supraspinatus (SS) muscles from six Mule×Charolais rams. Samples taken at slaughter were frozen either in liquid N(2) for analysis of MHC-s and MHC-f by immunoblotting, or in cooled isopentane for histochemical fibre typing. Calpastatin activity and an immunoreactive 135 kDa calpastatin band were analysed in samples taken 24 h postmortem. Shear force was determined on muscle chops taken at 24 h postmortem and conditioned until day 14. The intensity of MHC-s and MHC-f immunopositive bands correlated with %Type I and %Type II fibres identified histochemically (r(2)=0.612 and 0.366, respectively, p<0.001). Muscle specific differences were observed in MHC-s and MHC-f immunoreactivity, fibre type distribution, calpastatin activity, calpastatin 135 kDa immunoreactivity and shear force. MHC-s correlated positively with calpastatin activity (r(2)=0.725, p<0.001) and 135 kDa calpastatin (r(2)=0.228, p<0.01) across all muscle types. The data show that detection of MHC-s can be used to identify fibre type differences between ovine muscles and that this correlates with differences in calpastatin content and inhibitory activity, but not tenderness.

12.
J Pharmacol Exp Ther ; 311(3): 1225-31, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15243031

ABSTRACT

The intracellular mechanisms that regulate changes in postnatal myosin heavy chain (MHC) expression are not well established. The major objective of this study was to examine the acute and chronic effects of administration of BRL-47672, the prodrug of the beta2-adrenoceptor agonist clenbuterol on MHC and MyoD transcription factor expression to determine whether or not changes in MHC composition are preceded by changes in MyoD protein expression. To assess to what extent the use of BRL-47672 minimized cardiovascular effects, its hemodynamic actions were compared with those of clenbuterol. The effect of BRL-47672 on heart rate, mean arterial blood pressure, and hindquarters vascular conductance was significantly less than that of clenbuterol after a single i.p. injection (250 microg kg(-1) body mass). In the main study, 4-week old rats were given BRL-47672 (900 microg kg(-1) body mass) or an equivalent volume of saline (control) daily for 1, 28, or 56 days. Soleus muscle (SOL) was excised and MHC and MyoD expression analyzed. After 4 weeks, SOL from the BRL-47672-treated animals had significantly faster MHC composition (49 +/- 2% MHCIIA) compared with those from the control animal (39 +/- 3% MHCIIA, P <0.05). MyoD expression increased by 40% after 1 day of BRL-47672 administration (P <0.05) before a change in MHC composition. In conclusion, these data suggest that increased expression of fast-type MHCIIA expression in rat SOL induced by BRL-47672 administration is preceded by changes in the level of MyoD transcription factor expression.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Hemodynamics/drug effects , Muscle, Skeletal/metabolism , MyoD Protein/biosynthesis , Myosin Heavy Chains/metabolism , Adenosine Triphosphatases/metabolism , Animals , Blood Pressure/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Clenbuterol/pharmacology , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Heart Rate/drug effects , Male , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , MyoD Protein/genetics , Myofibrils/metabolism , Organic Chemicals , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects , Time Factors
13.
J Gen Physiol ; 118(5): 457-70, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11696605

ABSTRACT

The alpha1I T-type calcium channel inactivates almost 10-fold more slowly than the other family members (alpha1G and alpha1H) or most native T-channels. We have examined the underlying mechanisms using whole-cell recordings from rat alpha1I stably expressed in HEK293 cells. We found several kinetic differences between alpha1G and alpha1I, including some properties that at first appear qualitatively different. Notably, alpha1I tail currents require two or even three exponentials, whereas alpha1G tails were well described by a single exponential over a wide voltage range. Also, closed-state inactivation is more significant for alpha1I, even for relatively strong depolarizations. Despite these differences, gating of alpha1I can be described by the same kinetic scheme used for alpha1G, where voltage sensor movement is allosterically coupled to inactivation. Nearly all of the rate constants in the model are 5-12-fold slower for alpha1I, but the microscopic rate for channel closing is fourfold faster. This suggests that T-channels share a common gating mechanism, but with considerable quantitative variability.


Subject(s)
Calcium Channels, T-Type/physiology , Ion Channel Gating/physiology , Action Potentials , Animals , Cell Culture Techniques , Electrophysiology , Kinetics , Models, Theoretical , Rats
14.
Biophys J ; 81(2): 814-26, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11463627

ABSTRACT

We previously concluded that the Kv2.1 K(+) channel inactivates preferentially from partially activated closed states. We report here that the Kv3.1 channel also exhibits two key features of this inactivation mechanism: a U-shaped voltage dependence measured at 10 s and stronger inactivation with repetitive pulses than with a single long depolarization. More surprisingly, slow inactivation of the Kv1 Shaker K(+) channel (Shaker B Delta 6--46) also has a U-shaped voltage dependence for 10-s depolarizations. The time and voltage dependence of recovery from inactivation reveals two distinct components for Shaker. Strong depolarizations favor inactivation that is reduced by K(o)(+) or by partial block by TEA(o), as previously reported for slow inactivation of Shaker. However, depolarizations near 0 mV favor inactivation that recovers rapidly, with strong voltage dependence (as for Kv2.1 and 3.1). The fraction of channels that recover rapidly is increased in TEA(o) or high K(o)(+). We introduce the term U-type inactivation for the mechanism that is dominant in Kv2.1 and Kv3.1. U-type inactivation also makes a major but previously unrecognized contribution to slow inactivation of Shaker.


Subject(s)
Neuropeptides/antagonists & inhibitors , Neuropeptides/metabolism , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Allosteric Regulation/drug effects , Animals , Drosophila Proteins , Drosophila melanogaster , Electrophysiology , Kinetics , Membrane Potentials/drug effects , Models, Biological , Neuropeptides/genetics , Oocytes/drug effects , Oocytes/metabolism , Potassium Channels/genetics , RNA/genetics , RNA/metabolism , Shaker Superfamily of Potassium Channels , Shaw Potassium Channels , Tetraethylammonium/pharmacology , Xenopus laevis
15.
Biophys J ; 79(6): 3052-62, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11106611

ABSTRACT

We have examined permeation by Ca(2+) and Ba(2+), and block by Mg(2+), using whole-cell recordings from alpha1G T-type calcium channels stably expressed in HEK 293 cells. Without Mg(o)(2+), inward currents were comparable with Ca(2+) and Ba(2+). Surprisingly, three other results indicate that alpha1G is actually selective for Ca(2+) over Ba(2+). 1) Mg(2+) block is approximately 7-fold more potent with Ba(2+) than with Ca(2+). With near-physiological (1 mM) Mg(o)(2+), inward currents were approximately 3-fold larger with 2 mM Ca(2+) than with 2 mM Ba(2+). The stronger competition between Ca(2+) and Mg(2+) implies that Ca(2+) binds more tightly than Ba(2+). 2) Outward currents (carried by Na(+)) are blocked more strongly by Ca(2+) than by Ba(2+). 3) The reversal potential is more positive with Ca(2+) than with Ba(2+), thus P(Ca) > P(Ba). We conclude that alpha1G can distinguish Ca(2+) from Ba(2+), despite the similar inward currents in the absence of Mg(o)(2+). Our results can be explained by a 2-site, 3-barrier model if Ca(2+) enters the pore 2-fold more easily than Ba(2+) but exits the pore at a 2-fold lower rate.


Subject(s)
Barium/pharmacology , Calcium Channels, T-Type/physiology , Calcium/pharmacology , Magnesium/pharmacology , Animals , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/drug effects , Cations, Divalent/pharmacology , Cell Line , Humans , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Rats , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Transfection
16.
Biophys J ; 78(4): 1872-80, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10733967

ABSTRACT

In whole-cell recordings from HEK293 cells stably transfected with the delayed rectifier K(+) channel Kv2.1, long depolarizations produce current-dependent changes in [K(+)](i) that mimic inactivation and changes in ion selectivity. With 10 mM K(o)(+) or K(i)(+), and 140-160 mM Na(i,o)(+), long depolarizations shifted the reversal potential (V(R)) toward E(Na). However, similar shifts in V(R) were observed when Na(i,o)(+) was replaced with N-methyl-D-glucamine (NMG(+))(i, o). In that condition, [K(+)](o) did not change significantly, but the results could be quantitatively explained by changes in [K(+)](i). For example, a mean outward K(+) current of 1 nA for 2 s could decrease [K(+)](i) from 10 mM to 3 mM in a 10 pF cell. Dialysis by the recording pipette reduced but did not fully prevent changes in [K(+)](i). With 10 mM K(i,o)(+), 150 mM Na(i)(+), and 140 mM NMG(o)(+), steps to +20 mV produced a positive shift in V(R), as expected from depletion of K(i)(+), but opposite to the shift expected from a decreased K(+)/Na(+) selectivity. Long steps to V(R) caused inactivation, but no change in V(R). We conclude that current-dependent changes in [K(+)](i) need to be carefully evaluated when studying large K(+) currents in small cells.


Subject(s)
Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Biophysical Phenomena , Biophysics , Cell Line , Delayed Rectifier Potassium Channels , Humans , Intracellular Fluid/metabolism , Kinetics , Membrane Potentials , Potassium Channels/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Shab Potassium Channels , Sodium/metabolism , Transfection
17.
J Muscle Res Cell Motil ; 20(4): 417-24, 1999 May.
Article in English | MEDLINE | ID: mdl-10531622

ABSTRACT

Members of the calpain proteinase family are present in all mammalian cells, although a novel calpain 94 kDa isoform is found almost exclusively in skeletal muscle. p94 is difficult to purify from muscle and recombinant p94 autolyses rapidly when expressed in COS cells. However, in vivo the enzyme may be stabilised by interaction with titin, which has two well-characterised binding sites for p94 at the N2- and M-lines. Both these titin subdomains are subject to muscle-specific alternative splicing, which could be related to p94 expression level or stability in muscles of different fibre type. In this study, porcine longissimus dorsi (LD), trapezius (TZ) and adductor longus (AL) were characterised as fast, intermediate and slow using commercially available specific anti-human fast- and slow-myosin heavy chain mAbs and also by conventional histochemistry. p94 was quantified both in whole muscle preparations and single fibres by western blotting using an anti-p94 antiserum generated by expressing a recombinant p94 sequence as a GST fusion protein antigen. SDS PAGE and immunoblotting revealed a single band of approximately 94 kDa with identical mobility in all muscle and fibre preparations. The intensity of the 94 kDa band was greater in LD (22 +/- 1.7 densitometric units mean +/- SEM, n = 3) than TZ and AL (10 +/- 2.3 and 6 +/- 0.9 units, respectively). Expressed as a ratio relative to actin immunoreactivity, p94 is present in all types of single fibres isolated from TZ, but at a significantly lower level (P < 0.01) in slow type I (0.08 +/- 0.01, n = 9), compared to fast IIA/IIB fibres (0.22 +/- 0.02, n = 26). No evidence was seen for rapid or variable rate of p94 degradation in either type of fibre. These data suggest a positive correlation between p94 expression level and fast glycolytic characteristics in porcine muscle.


Subject(s)
Calpain/biosynthesis , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/metabolism , Animals , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/pathology , Muscle, Skeletal/pathology , Protein Isoforms/metabolism , Swine , Time Factors
19.
J Gen Physiol ; 114(2): 185-201, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10435997

ABSTRACT

We have examined the kinetics of whole-cell T-current in HEK 293 cells stably expressing the alpha1G channel, with symmetrical Na(+)(i) and Na(+)(o) and 2 mM Ca(2+)(o). After brief strong depolarization to activate the channels (2 ms at +60 mV; holding potential -100 mV), currents relaxed exponentially at all voltages. The time constant of the relaxation was exponentially voltage dependent from -120 to -70 mV (e-fold for 31 mV; tau = 2.5 ms at -100 mV), but tau = 12-17 ms from-40 to +60 mV. This suggests a mixture of voltage-dependent deactivation (dominating at very negative voltages) and nearly voltage-independent inactivation. Inactivation measured by test pulses following that protocol was consistent with open-state inactivation. During depolarizations lasting 100-300 ms, inactivation was strong but incomplete (approximately 98%). Inactivation was also produced by long, weak depolarizations (tau = 220 ms at -80 mV; V(1/2) = -82 mV), which could not be explained by voltage-independent inactivation exclusively from the open state. Recovery from inactivation was exponential and fast (tau = 85 ms at -100 mV), but weakly voltage dependent. Recovery was similar after 60-ms steps to -20 mV or 600-ms steps to -70 mV, suggesting rapid equilibration of open- and closed-state inactivation. There was little current at -100 mV during recovery from inactivation, consistent with

Subject(s)
Calcium Channels/physiology , Ion Channel Gating/physiology , Algorithms , Animals , Calcium Channels, T-Type , Electric Stimulation , Electrophysiology , Kinetics , Membrane Potentials/physiology , Models, Neurological , Patch-Clamp Techniques , Rats , Sodium/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...