Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
J Perinatol ; 37(5): 606-614, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28079875

RESUMO

OBJECTIVE: A neonatal illness severity score, The Score for Neonatal Acute Physiology-II (SNAP-II), predicts neurodevelopmental impairments at two years of age among children born extremely preterm. We sought to evaluate to what extent SNAP-II is predictive of cognitive and other neurodevelopmental impairments at 10 years of age. STUDY DESIGN: In a cohort of 874 children born before 28 weeks of gestation, we prospectively collected clinical, physiologic and laboratory data to calculate SNAP-II for each infant. When the children were 10 years old, examiners who were unaware of the child's medical history assessed neurodevelopmental outcomes, including neurocognitive, gross motor, social and communication functions, diagnosis and treatment of seizures or attention deficit hyperactivity disorder (ADHD), academic achievement, and quality of life. We used logistic regression to adjust for potential confounders. RESULTS: An undesirably high SNAP-II (⩾30), present in 23% of participants, was associated with an increased risk of cognitive impairment (IQ, executive function, language ability), adverse neurological outcomes (epilepsy, impaired gross motor function), behavioral abnormalities (attention deficit disorder and hyperactivity), social dysfunction (autistic spectrum disorder) and education-related adversities (school achievement and need for educational supports. In analyses that adjusted for potential confounders, Z-scores ⩽-1 on 11 of 18 cognitive outcomes were associated with SNAP-II in the highest category, and 6 of 18 were associated with SNAP-II in the intermediate category. Odds ratios and 95% confidence intervals ranged from 1.4 (1.01, 2.1) to 2.1 (1.4, 3.1). Similarly, 2 of the 8 social dysfunctions were associated with SNAP-II in the highest category, and 3 of 8 were associated with SNAP-II in the intermediate category. Odds ratios and 95% confidence intervals were slightly higher for these assessments, ranging from 1.6 (1.1, 2.4) to 2.3 (1.2, 4.6). CONCLUSION: Among very preterm newborns, physiologic derangements present in the first 12 postnatal hours are associated with dysfunctions in several neurodevelopmental domains at 10 years of age. We are unable to make inferences about causality.


Assuntos
Deficiências do Desenvolvimento/diagnóstico , Lactente Extremamente Prematuro/crescimento & desenvolvimento , Índice de Gravidade de Doença , Criança , Desenvolvimento Infantil , Deficiências do Desenvolvimento/fisiopatologia , Função Executiva , Feminino , Idade Gestacional , Humanos , Recém-Nascido , Modelos Logísticos , Masculino , Estudos Prospectivos , Qualidade de Vida , Estados Unidos
2.
J Perinatol ; 34(9): 705-10, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25010224

RESUMO

OBJECTIVE: Bronchopulmonary dysplasia (BPD) is the most common cause of pulmonary morbidity in premature infants and is associated with life-long morbidities. Developing drugs for the prevention of BPD would improve public health. We sought to determine characteristics of favorable randomized controlled trials (RCTs) of drugs for BPD prevention. STUDY DESIGN: We searched MEDLINE and EMBASE from 1992 to 2014 using the MeSH terms 'BPD' and 'respiratory distress syndrome, newborn'. We included a Cochrane Library search to ensure inclusion of all available RCTs. We identified RCTs with BPD as a primary or secondary outcome and determined the definition of BPD used by the study. We determined whether a phase I or phase II study-to determine drug safety, efficacy or optimal dose-was performed before the RCT. Finally, we searched the Cochrane Library for meta-analyses for each drug and used the results of available meta-analyses to define a favorable versus unfavorable RCT. RESULT: We identified 2026 articles; 47 RCTs met our inclusion criteria encompassing 21 drugs; 5 of the drugs reduced the incidence of BPD. We found data from phase I or II studies for 16 of the drugs, but only 1 demonstrated a reduction of BPD. CONCLUSION: The majority of the drugs studied in RCTs failed to reduce the incidence of BPD. Performing early-phase studies before phase III trials might provide necessary information on drugs and drug doses capable of preventing BPD, thus informing the development of future RCTs.


Assuntos
Displasia Broncopulmonar/prevenção & controle , Displasia Broncopulmonar/tratamento farmacológico , Ensaios Clínicos Fase I como Assunto , Humanos , Recém-Nascido , Metanálise como Assunto , Ensaios Clínicos Controlados Aleatórios como Assunto
3.
J Muscle Res Cell Motil ; 30(5-6): 217-23, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19802526

RESUMO

In skeletal muscle, there is bidirectional signalling between the L-type Ca(2+) channel (1,4-dihydropyridine receptor; DHPR) and the type 1 ryanodine-sensitive Ca(2+) release channel (RyR1) of the sarcoplasmic reticulum (SR). In the case of "orthograde signalling" (i.e., excitation-contraction coupling), the conformation of RyR1 is controlled by depolarization-induced conformational changes of the DHPR resulting in Ca(2+) release from the SR. "Retrograde coupling" is manifested as enhanced L-type current. The nature of this retrograde signal, and its dependence on RyR1 conformation, are poorly understood. Here, we have examined L-type currents in normal myotubes after an exposure to ryanodine (200 microM, 1 h at 37 degrees C) sufficient to lock RyR1 in a non-conducting, inactivated, conformational state. This treatment caused an increase in L-type current at less depolarized test potentials in comparison to myotubes similarly exposed to vehicle as a result of a approximately 5 mV hyperpolarizing shift in the voltage-dependence of activation. Charge movements of ryanodine-treated myotubes were also shifted to more hyperpolarizing potentials (approximately 13 mV) relative to vehicle-treated myotubes. Enhancement of the L-type current by ryanodine was absent in dyspedic (RyR1 null) myotubes, indicating that ryanodine does not act directly on the DHPR. Our findings indicate that in retrograde signaling, the functional state of RyR1 influences conformational changes of the DHPR involved in activation of L-type current. This raises the possibility that physiological regulators of the conformational state of RyR1 (e.g., Ca(2+), CaM, CaMK, redox potential) may also affect DHPR gating.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/fisiologia , Ativação do Canal Iônico/fisiologia , Músculo Esquelético/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Rianodina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Knockout , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/efeitos dos fármacos , Conformação Proteica/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
4.
Biophys J ; 94(7): 2631-8, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18192376

RESUMO

In skeletal muscle, the L-type voltage-gated Ca(2+) channel (1,4-dihydropyridine receptor) serves as the voltage sensor for excitation-contraction (EC) coupling. In this study, we examined the effects of Rem, a member of the RGK (Rem, Rem2, Rad, Gem/Kir) family of Ras-related monomeric GTP-binding proteins, on the function of the skeletal muscle L-type Ca(2+) channel. EC coupling was found to be weakened in myotubes expressing Rem tagged with enhanced yellow fluorescent protein (YFP-Rem), as assayed by electrically evoked contractions and myoplasmic Ca(2+) transients. This impaired EC coupling was not a consequence of altered function of the type 1 ryanodine receptor, or of reduced Ca(2+) stores, since the application of 4-chloro-m-cresol, a direct type 1 ryanodine receptor activator, elicited myoplasmic Ca(2+) release in YFP-Rem-expressing myotubes that was not distinguishable from that in control myotubes. However, YFP-Rem reduced the magnitude of L-type Ca(2+) current by approximately 75% and produced a concomitant reduction in membrane-bound charge movements. Thus, our results indicate that Rem negatively regulates skeletal muscle EC coupling by reducing the number of functional L-type Ca(2+) channels in the plasma membrane.


Assuntos
Potenciais de Ação/fisiologia , Canais de Cálcio Tipo L/fisiologia , Cálcio/metabolismo , Ativação do Canal Iônico/fisiologia , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Contração Muscular/fisiologia , Fibras Musculares Esqueléticas/fisiologia , Animais , Animais Recém-Nascidos , Sinalização do Cálcio/fisiologia , Células Cultivadas , Camundongos , Camundongos Transgênicos
5.
Biochem Biophys Res Commun ; 336(1): 134-41, 2005 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-16139246

RESUMO

The dihydropyridine receptor (DHPR) alpha(1S) II-III loop has been shown to be critical for excitation-contraction (EC) coupling in skeletal muscle, but the importance of other cytoplasmic regions, especially the N-terminus (residues 1-51), remains unclear. In this study, we found that deletion of alpha(1S) residues 2-37 (weakly conserved with N-termini of other L-type Ca(2+) channels) had little effect on the ability of alpha(1S) to serve as a Ca(2+) channel or voltage sensor for EC coupling. Strikingly, deletion of 10 additional residues, which are conserved in L-type channels, resulted in ablation of DHPR function. Specifically, confocal microscopy and measurement of charge movement showed that removal of residues 2-47 resulted in a failure of sarcolemmal insertion. Our results indicate that the weakly conserved, distal alpha(1S) N-terminus is not critical for EC coupling or function as a Ca(2+) channel. However, integrity of the proximal alpha(1S) N-terminus is necessary for sarcolemmal expression of the DHPR.


Assuntos
Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Canais de Cálcio Tipo L/metabolismo , Células Cultivadas , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Homologia de Sequência de Aminoácidos
6.
Vet Parasitol ; 110(1-2): 131-5, 2002 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-12446098

RESUMO

Toxoplasma gondii infection was diagnosed in a full term stillborn reindeer (Rangifer tarandus) fetus. The fetus had encephalitis and placentitis associated with T. gondii. Tissue cysts were identified histologically in sections of brain and tachyzoites were present in placenta and the myocardium. Protozoa in the brain, heart, and placenta stained positively with T. gondii antibodies, but not with Neospora caninum antibodies in an immunohistochemical test. The dam of the fetus had a 1:12,800 titer to T. gondii in the modified agglutination test employing whole tachyzoites and mercaptoethanol. This is the first confirmed report of T. gondii infection in reindeer.


Assuntos
Doenças Fetais/parasitologia , Rena/parasitologia , Toxoplasma/isolamento & purificação , Toxoplasmose Animal/parasitologia , Animais , Anticorpos Antiprotozoários/sangue , Encéfalo/parasitologia , Encéfalo/patologia , Encefalite/parasitologia , Encefalite/patologia , Encefalite/veterinária , Evolução Fatal , Feminino , Doenças Fetais/patologia , Coração/parasitologia , Imuno-Histoquímica/veterinária , Masculino , Placenta/parasitologia , Placenta/patologia , Gravidez , Toxoplasmose Animal/patologia
7.
J Gen Physiol ; 118(5): 495-508, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11696608

RESUMO

A defining property of L-type Ca(2+) channels is their potentiation by both 1,4-dihydropyridine agonists and strong depolarization. In contrast, non-L-type channels are potentiated by neither agonist nor depolarization, suggesting that these two processes may by linked. In this study, we have tested whether the mechanisms of agonist- and depolarization-induced potentiation in the cardiac L-type channel (alpha(1C)) are linked. We found that the mutant L-type channel GFP-alpha(1C)(TQ-->YM), bearing the mutations T1066Y and Q1070M, was able to undergo depolarization-induced potentiation but not potentiation by agonist. Conversely, the chimeric channel GFP-CACC was potentiated by agonist but not by strong depolarization. These data indicate that the mechanisms of agonist- and depolarization-induced potentiation of alpha(1C) are distinct. Since neither GFP-CACC nor GFP-CCAA was potentiated significantly by depolarization, no single repeat of alpha(1C) appears to be responsible for depolarization-induced potentiation. Surprisingly, GFP-CACC displayed a low estimated open probability similar to that of the alpha(1C), but could not support depolarization-induced potentiation, demonstrating that a relatively low open probability alone is not sufficient for depolarization-induced potentiation to occur. Thus, depolarization-induced potentiation may be a global channel property requiring participation from all four homologous repeats.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/fisiologia , Di-Hidropiridinas/farmacologia , Coração/fisiologia , Potenciais de Ação , Animais , Eletrofisiologia , Potenciais da Membrana , Camundongos , Mutagênese Sítio-Dirigida , Mutação Puntual , Reação em Cadeia da Polimerase
8.
Bioconjug Chem ; 12(6): 1074-80, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11716702

RESUMO

CPT-11 is a clinically approved anticancer drug used for the treatment of advanced colorectal cancer. Upon administration, the carbamate side chain of the drug is hydrolyzed, resulting in the release of SN-38, an agent that has approximately 1000-fold increased cytotoxic activity. Since only a very small percentage of the injected dose of CPT-11 is converted to SN-38, there is a significant opportunity to improve its therapeutic efficacy and to diminish its systemic toxicity by selectively activating the drug within tumor sites. We envisioned that a mAb-human enzyme conjugate for CPT-11 activation would be of interest, particularly since the conjugate would likely be minimally immunogenic, and the prodrug is clinically approved. Toward this end, it was necessary to identify the most active human enzyme that could convert CPT-11 to SN-38. We isolated enzymes from human liver microsomes based on their abilities to effect the conversion and identified human carboxylesterase 2 (hCE-2) as having the greatest specific activity. hCE-2 was 26-fold more active than human carboxylesterase 1 and was 65% as active as rabbit liver carboxylesterase, the most active CPT-11 hydrolyzing enzyme known. The anti-p97 mAb 96.5 was linked to hCE-2, forming a conjugate that could bind to antigen-positive cancer cells and convert CPT-11 to SN-38. Cytotoxicity assays established that the conjugate led to the generation of active drug, but the kinetics of prodrug activation (48 pmol x min(-1) x mg(-1) was insufficient for immunologically specific prodrug activation. These results confirm the importance of hCE-2 for CPT-11 activation and underscore the importance of enzyme kinetics for selective prodrug activation.


Assuntos
Antineoplásicos Fitogênicos/metabolismo , Camptotecina/análogos & derivados , Camptotecina/metabolismo , Hidrolases de Éster Carboxílico/isolamento & purificação , Sistemas de Liberação de Medicamentos , Pró-Fármacos/metabolismo , Sequência de Aminoácidos , Anticorpos Antineoplásicos/química , Sequência de Bases , Ligação Competitiva , Hidrolases de Éster Carboxílico/química , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Imunoconjugados/metabolismo , Irinotecano , Fígado/enzimologia , Dados de Sequência Molecular , Células Tumorais Cultivadas/efeitos dos fármacos
9.
Pflugers Arch ; 442(6): 859-66, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11680618

RESUMO

FLASH-EDT2--4',5'-bis(1,3,2-dithioarsolan-2-yl)fluorescein-(1,2-ethanedithiol)2--has been reported to fluoresce only after binding with high affinity to a specific tetracysteine motif (CCXXCC, "Cys4") and thus to provide a technique for labeling recombinant proteins in vivo (Griffin et al. Science 281:269-272). We have attempted to use FLASH-EDT2 as a site-specific label of the II-III loop of the dihydropyridine receptor (DHPR) in skeletal muscle. Upon expression in dysgenic myotubes (which lack endogenous alpha1s), an alpha1s mutated to contain CCRECC in the II-III loop was able to produce L-type calcium currents and to mediate skeletal-type excitation-contraction (EC) coupling, but FLASH-EDT2 labeling revealed no difference from non-transfected dysgenic myotubes. HeLa-S3 cells transfected with Cys4-containing calmodulin were significantly more fluorescent than non-transfected cells, whereas the difference between transfected and non-transfected cells was less apparent for CHO-K and HEK 293 cells. Because the fluorescence of non-transfected cells increased substantially after treatment with FLASH-EDT2, it suggested the possibility that FLASH binds to endogenous cysteine-containing proteins. This finding was confirmed in cuvette experiments in which FLASH-EDT2 fluorescence was observed after FLASH-EDT, was added to protein homogenates from myotubes or cell lines. The enhanced fluorescence was abolished by pretreatment of cells or cell homogenates with coumarine maleimide (CPM), which modifies cysteine residues covalently. Thus, enhanced FLASH fluorescence appears to occur both after binding to an introduced Cys4 motif and to endogenous, cysteine-containing proteins. Therefore, FLASH-EDT2 may be useful only for labeling those recombinant proteins that express at a very high level.


Assuntos
Cisteína/metabolismo , Fluoresceínas/metabolismo , Corantes Fluorescentes/metabolismo , Compostos Organometálicos/metabolismo , Proteínas/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células CHO , Canais de Cálcio Tipo L/metabolismo , Calmodulina/química , Calmodulina/genética , Linhagem Celular , Cricetinae , Condutividade Elétrica , Humanos , Indicadores e Reagentes , Espectroscopia de Ressonância Magnética , Camundongos , Músculo Esquelético/metabolismo , Mutagênese , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Reação em Cadeia da Polimerase , Proteínas Recombinantes , Espectrometria de Fluorescência , Transfecção
10.
Arch Biochem Biophys ; 388(1): 13-7, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11361129

RESUMO

In skeletal muscle, an anterograde signal from the dihydropyridine receptor (DHPR) to the ryanodine receptor (RyR1) is required for excitation-contraction (EC) coupling and a retrograde signal from RyR1 to the DHPR regulates the magnitude of the calcium current carried by the DHPR. As a tool for studying biosynthesis and targeting, we constructed a cDNA encoding green fluorescent protein (GFP) fused to the amino terminal of RyR1 and expressed it in dyspedic myotubes. The GFP-RyR1 was present in a restricted domain near the nucleus injected with cDNA and was fully functional, which places constraints on the location of the amino terminal in the folded structure of RyR1.


Assuntos
Proteínas Luminescentes/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Cálcio/farmacologia , Núcleo Celular/metabolismo , DNA Complementar/metabolismo , Eletrofisiologia , Proteínas de Fluorescência Verde , Camundongos , Modelos Biológicos , Músculo Esquelético/metabolismo , Músculos/citologia , Músculos/metabolismo , Dobramento de Proteína , Transdução de Sinais
11.
Proc Natl Acad Sci U S A ; 98(10): 5892-7, 2001 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-11320225

RESUMO

The II-III loop of the skeletal muscle dihydropyridine receptor (DHPR) alpha(1S) subunit is responsible for bidirectional-signaling interactions with the ryanodine receptor (RyR1): transmitting an orthograde, excitation-contraction (EC) coupling signal to RyR1 and receiving a retrograde, current-enhancing signal from RyR1. Previously, several reports argued for the importance of two distinct regions of the skeletal II-III loop (residues R681-L690 and residues L720-Q765, respectively), claiming for each a key function in DHPR-RyR1 communication. To address whether residues 720-765 of the II-III loop are sufficient to enable skeletal-type (Ca(2+) entry-independent) EC coupling and retrograde interaction with RyR1, we constructed a green fluorescent protein (GFP)-tagged chimera (GFP-SkLM) having rabbit skeletal (Sk) DHPR sequence except for a II-III loop (L) from the DHPR of the house fly, Musca domestica (M). The Musca II-III loop (75% dissimilarity to alpha(1S)) has no similarity to alpha(1S) in the regions R681-L690 and L720-Q765. GFP-SkLM expressed in dysgenic myotubes (which lack endogenous alpha(1S) subunits) was unable to restore EC coupling and displayed strongly reduced Ca(2+) current densities despite normal surface expression levels and correct triad targeting (colocalization with RyR1). Introducing rabbit alpha(1S) residues L720-L764 into the Musca II-III loop of GFP-SkLM (substitution for Musca DHPR residues E724-T755) completely restored bidirectional coupling, indicating its dependence on alpha(1S) loop residues 720-764 but its independence from other regions of the loop. Thus, 45 alpha(1S)-residues embedded in a very dissimilar background are sufficient to restore bidirectional coupling, indicating that these residues may be a site of a protein-protein interaction required for bidirectional coupling.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Músculo Esquelético/fisiologia , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/genética , Células Cultivadas , DNA Complementar , Dados de Sequência Molecular , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Técnicas de Patch-Clamp , Coelhos , Proteínas Recombinantes de Fusão/genética , Homologia de Sequência de Aminoácidos
12.
J Biol Chem ; 275(39): 29935-7, 2000 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-10915779

RESUMO

A peptide corresponding to residues 681-690 of the II-III loop of the skeletal muscle dihydropyridine receptor alpha(1) subunit (DHPR, alpha(1S)) has been reported to activate the skeletal muscle ryanodine receptor (RyR1) in vitro. Within this region of alpha(1S), a cluster of basic residues, Arg(681)-Lys(685), was previously reported to be indispensable for the activation of RyR1 in microsomal preparations and lipid bilayers. We have used an intact alpha(1S) subunit with scrambled sequence in this region of the II-III loop (alpha(1S)-scr) to test the importance of residues 681-690 and the basic motif for skeletal-type excitation-contraction (EC) coupling and retrograde signaling in vivo. When expressed in dysgenic myotubes (which lack endogenous alpha(1S)), alpha(1S)-scr restored calcium currents that were indistinguishable, in current density and voltage dependence, from those restored by wild-type alpha(1S). The scrambled DHPR also rescued skeletal-type EC coupling, as indicated by electrically evoked contractions in the presence of 0.5 mm Cd(2+) and 0.1 mm La(3+). Furthermore, the release of intracellular Ca(2+), as assayed by the indicator dye, Fluo-3, had similar kinetics and voltage dependence for alpha(1S) and alpha(1S)-scr. These data suggest that residues 681-690 of the alpha(1S) II-III loop are not essential in muscle cells for normal functioning of the DHPR, including skeletal-type EC coupling and retrograde signaling.


Assuntos
Contração Muscular/fisiologia , Músculo Esquelético/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Camundongos , Modelos Moleculares , Oligopeptídeos/química , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Estrutura Secundária de Proteína , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Transdução de Sinais
13.
J Biol Chem ; 275(30): 23169-74, 2000 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-10801875

RESUMO

We have used the yeast two-hybrid technique and expression of truncated/mutated dihydropyridine receptors (DHPRs) to investigate whether the carboxyl tail of the DHPR is involved in targeting to junctions between the sarcolemma and sarcoplasmic reticulum in skeletal muscle. The carboxyl tail was extremely reactive in yeast two-hybrid library screens, with the reactivity residing in amino acids 1621-1647 and abolished by a point mutation (V1642D). Dysgenic myotubes were injected with cDNA encoding green fluorescent protein fused to the amino terminus of DHPRs truncated after either residue 1620 (Delta1621-1873) or residue 1542 (Delta1543-1873) or of full-length DHPRs with the V1642D mutation (V1642D). For either Delta1621-1873 or V1642D, the restoration of excitation-contraction coupling was reduced approximately 40%, and the number of functional DHPRs in the sarcolemma was reduced approximately 30%, compared with the wild-type DHPR. The restoration of excitation-contraction coupling and surface expression was more drastically reduced (by approximately 90 and approximately 55%, respectively) for Delta1543-1873. Fluorescence microscopy revealed that Delta1621-1873 and V1642D were concentrated in a longitudinally restricted region near the injected nucleus, whereas wild-type DHPRs were present relatively uniformly along the length of a myotube. The intensity of fluorescence was greatly reduced for Delta1543-1873, indicating a low level of protein expression. Thus, residues 1543-1647 appear to play a role in the biosynthetic processing, transport, and/or anchoring of DHPRs, with residues 1543-1620 being particularly important for expression.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Músculo Esquelético/metabolismo , Animais , Canais de Cálcio Tipo L/genética , DNA Complementar , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Microscopia de Fluorescência , Coelhos , Técnicas do Sistema de Duplo-Híbrido
14.
Pflugers Arch ; 439(5): 555-8, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10764214

RESUMO

Patch-clamping and the simultaneous fluorescence measurement of cytoplasmic Ca2+ ([Ca2+]i) were used to analyze the effect of serum on the functional features of excitation-contraction (E-C) coupling in mouse skeletal myotubes. In high-serum-treated (10%) myotubes, depolarization elicited Ca2+ release which continued for tens of milliseconds following the end of the pulse, after which [Ca2+]i decayed slowly. In low-serum-treated (0.5%) myotubes, the Ca2+ transient caused by depolarization had an increased rate of rise and peak amplitude, and [Ca2+]i began to decay rapidly upon repolarization. When a depolarizing pulse (0.5-1.0 s) was applied to low-serum-treated myotubes during a Ca2+ transient induced by 5-10 mM caffeine, repolarization usually caused the caffeine transient to terminate rapidly (RISC; repolarization-induced stop of caffeine-induced Ca2+ release). The RISC was less prominent in high-serum-treated myotubes. These results suggest that low serum promotes the maturation of myotubes so that Ca(2+)-release and Ca(2+)-removal activities are accelerated. Additionally, the essential features of the communication between the voltage sensor and the Ca(2+)-release channel are shared by myotubes and adult muscle fibers.


Assuntos
Proteínas Sanguíneas/farmacologia , Contração Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/citologia , Fatores Etários , Animais , Animais Recém-Nascidos , Cafeína/farmacologia , Cálcio/farmacocinética , Canais de Cálcio Tipo L/fisiologia , Células Cultivadas , Eletrofisiologia , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/citologia , Inibidores de Fosfodiesterase/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia
15.
Kidney Int ; 57(3): 794-802, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10720931

RESUMO

Calcium is an important intracellular signaling molecule, and altered calcium channel function can cause widespread cellular changes. Genetic mutations in calcium channels that cause what appear to be trivial alterations of calcium currents in vitro can result in serious diseases in muscles and the nervous system. This article reviews calcium channelopathies in humans and mice.


Assuntos
Canais de Cálcio/genética , Doenças Musculares/genética , Mutação/fisiologia , Doenças do Sistema Nervoso/genética , Animais , Canais de Cálcio/química , Canais de Cálcio/fisiologia , Eletrofisiologia , Humanos , Músculo Esquelético/metabolismo , Doenças do Sistema Nervoso/metabolismo , Relação Estrutura-Atividade
16.
Brain Res ; 849(1-2): 162-8, 1999 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-10592298

RESUMO

The Scn8a gene encodes a neuronal, voltage-gated sodium channel, which is highly expressed in both cerebellar Purkinje neurons and spinal motoneurons [D.L. Burgess, D.C. Kohrman, J. Galt, N.W. Plummer, J.M. Jones, B. Spear, M.H. Meisler, Mutation of a new sodium channel gene, Scn8a, in the mouse mutant 'motor endplate disease', Nature Genetics 10 (1995) 461-465; K.L. Schaller, D.M. Krzemien, P.J. Yarowsky, B.K. Krueger, J.H. Caldwell, A novel, abundant sodium channel expressed in neurons and glia, J. Neurosci. 15 (1995) 3231-3242]. Sodium channels in Purkinje cells produce an unusual, "resurgent" current when the cells are repolarized to intermediate potentials (-60 to -20 mV) following a strong depolarization that completely inactivates transient sodium current [I.M. Raman, L.K. Sprunger, M.H. Meisler, B.P. Bean, Altered subthreshold sodium currents and disrupted firing patterns in Purkinje neurons of Scn8a mutant mice, Neuron 19 (1997) 881-891; I.M. Raman, B.P. Bean, Resurgent sodium current and action potential formation in dissociated cerebellar Purkinje neurons, J. Neurosci. 17 (1997) 4517-4526]. Here, we have examined whether large spinal neurons (predominantly motoneurons), isolated from P6-P8 mice and cultured overnight, produce sodium currents resembling those either of Purkinje cells or of Xenopus oocytes after heterologous expression of Scn8a. We found that P10-P14 Purkinje cells exhibited resurgent current (ranging from -3.6 to -15.4 pA/pF in 16 cells at -40 mV), but cultured spinal neurons had little or no such current (<0.5 pA/pF in 13 of 16 cells; -1.2 to -2.3 pA/pF in three of 16 cells). Furthermore, unlike Scn8a channels heterologously expressed in Xenopus oocytes [M.R. Smith, R.D. Smith, N.W. Plummer, M.H. Meisler, A.L. Goldin, Functional analysis of the mouse Scn8a sodium channel. J. Neurosci. 18 (1998) 6093-6102], there was not a prominent component of persistent sodium current in either Purkinje neurons or large spinal neurons. Based on analysis of cells from mice with a Scn8a null mutation, Scn8a channels appear to contribute significantly to total sodium current in both in P10-P14 Purkinje cells (approximately 40%; [21]) and cultured P7-P8 spinal motoneurons (approximately 70% [K.D. García, L.K. Sprunger, M.H. Meisler, K.G. Beam, The sodium channel Scn8a is the major contributor to the postnatal developmental increase of sodium current density in spinal motoneurons, J. Neurosci. 18 (1998) 5234-5239]). Thus, the presence or absence of resurgent current, and of persistent sodium current, appears to depend on cellular factors other than the mere presence of the Scn8a transcript.


Assuntos
Neurônios/fisiologia , Células de Purkinje/fisiologia , Canais de Sódio/fisiologia , Medula Espinal/fisiologia , Potenciais de Ação , Animais , Animais Recém-Nascidos , Células Cultivadas , Cerebelo/citologia , Cerebelo/fisiologia , Camundongos , Neurônios Motores/citologia , Neurônios Motores/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.6 , Proteínas do Tecido Nervoso/fisiologia , Neurônios/citologia , Técnicas de Patch-Clamp , Células de Purkinje/citologia , Medula Espinal/citologia
17.
J Gen Physiol ; 114(3): 393-403, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10469729

RESUMO

The skeletal and cardiac muscle dihydropyridine receptors (DHPRs) differ with respect to their rates of channel activation and in the means by which they control Ca2+ release from the sarcoplasmic reticulum (Adams, B.A., and K.G. Beam. 1990. FASEB J. 4:2809-2816). We have examined the functional properties of skeletal (SkEIIIK) and cardiac (CEIIIK) DHPRs in which a highly conserved glutamate residue in the pore region of repeat III was mutated to a positively charged lysine residue. Using expression in dysgenic myotubes, we have characterized macroscopic ionic currents, intramembrane gating currents, and intracellular Ca2+ transients attributable to these two mutant DHPRs. CEIIIK supported very small inward Ca2+ currents at a few potentials (from -20 to +20 mV) and large outward cesium currents at potentials greater than +20 mV. SkEIIIK failed to support inward Ca2+ flux at any potential. However, large, slowly activating outward cesium currents were observed at all potentials greater than + 20 mV. The difference in skeletal and cardiac Ca2+ channel activation kinetics was conserved for outward currents through CEIIIK and SkEIIIK, even at very depolarized potentials (at +100 mV; SkEIIIK: tau(act) = 30.7 +/- 1.9 ms, n = 11; CEIIIK: tau(act) = 2.9 +/- 0.5 ms, n = 7). Expression of SkEIIIK in dysgenic myotubes restored both evoked contractions and depolarization-dependent intracellular Ca(2+) transients with parameters of voltage dependence (V(0.5) = 6.5 +/- 3.2 mV and k = 9.3 +/- 0.7 mV, n = 5) similar to those for the wild-type DHPR (Garcia, J., T. Tanabe, and K.G. Beam. 1994. J. Gen. Physiol. 103:125-147). However, CEIIIK-expressing myotubes never contracted and failed to exhibit depolarization-dependent intracellular Ca2+ transients at any potential. Thus, high Ca2+ permeation is required for cardiac-type excitation-contraction coupling reconstituted in dysgenic myotubes, but not skeletal-type. The strong rectification of the EIIIK channels made it possible to obtain measurements of gating currents upon repolarization to -50 mV (Qoff) following either brief (20 ms) or long (200 ms) depolarizing pulses to various test potentials. For SkEIIIK, and not CEIIK, Qoff was significantly (P < 0.001) larger after longer depolarizations to +60 mV (121.4 +/- 2.0%, n = 6). The increase in Qoff for long depolarizations exhibited a voltage dependence similar to that of channel activation. Thus, the increase in Q(off) may reflect a voltage sensor movement required for activation of L-type Ca2+ current and suggests that most DHPRs in skeletal muscle undergo this voltage-dependent transition.


Assuntos
Canais de Cálcio/metabolismo , Coração/fisiologia , Ativação do Canal Iônico/fisiologia , Proteínas Musculares/metabolismo , Músculo Esquelético/fisiologia , Algoritmos , Substituição de Aminoácidos/genética , Substituição de Aminoácidos/fisiologia , Animais , Canais de Cálcio/genética , Canais de Cálcio/fisiologia , Canais de Cálcio Tipo L , Células Cultivadas , DNA Complementar/biossíntese , DNA Complementar/genética , Eletrofisiologia , Ativação do Canal Iônico/genética , Cinética , Potenciais da Membrana/fisiologia , Camundongos , Microtúbulos/metabolismo , Microtúbulos/fisiologia , Contração Muscular/fisiologia , Proteínas Musculares/genética , Proteínas Musculares/fisiologia , Contração Miocárdica/fisiologia
18.
J Biol Chem ; 274(31): 21913-9, 1999 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-10419512

RESUMO

The dihydropyridine receptor (DHPR) in the skeletal muscle plasmalemma functions as both voltage-gated Ca(2+) channel and voltage sensor for excitation-contraction (EC) coupling. As voltage sensor, the DHPR regulates intracellular Ca(2+) release via the skeletal isoform of the ryanodine receptor (RyR-1). Interaction with RyR-1 also feeds back to increase the Ca(2+) current mediated by the DHPR. To identify regions of the DHPR important for receiving this signal from RyR-1, we expressed in dysgenic myotubes a chimera (SkLC) having skeletal (Sk) DHPR sequence except for a cardiac (C) II-III loop (L). Tagging with green fluorescent protein (GFP) enabled identification of expressing myotubes. Dysgenic myotubes expressing GFP-SkLC or SkLC lacked EC coupling and had very small Ca(2+) currents. Introducing a short skeletal segment (alpha(1S) residues 720-765) into the cardiac II-III loop (replacing alpha(1C) residues 851-896) of GFP-SkLC restored both EC coupling and Ca(2+) current densities like those of the wild type skeletal DHPR. This 46-amino acid stretch of skeletal sequence was recently shown to be capable of transferring strong, skeletal-type EC coupling to an otherwise cardiac DHPR (Nakai, J., Tanabe, T., Konno, T., Adams, B., and Beam, K.G. (1998) J. Biol. Chem. 273, 24983-24986). Thus, this segment of the skeletal II-III loop contains a motif required for both skeletal-type EC coupling and RyR-1-mediated enhancement of Ca(2+) current.


Assuntos
Canais de Cálcio/química , Canais de Cálcio/fisiologia , Músculo Esquelético/fisiologia , Receptor Cross-Talk/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Canais de Cálcio/genética , Canais de Cálcio Tipo L , Células Cultivadas , Proteínas de Fluorescência Verde , Coração/fisiologia , Proteínas Luminescentes/genética , Potenciais da Membrana , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Músculo Esquelético/citologia , Músculo Esquelético/fisiopatologia , Técnicas de Patch-Clamp , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Mapeamento por Restrição , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Transdução de Sinais
20.
J Biol Chem ; 273(39): 24983-6, 1998 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-9737952

RESUMO

Skeletal and cardiac muscles express distinct isoforms of the dihydropyridine receptor (DHPR), a type of voltage-gated Ca2+ channel that is important for excitation-contraction (EC) coupling. However, entry of Ca2+ through the channel is not required for skeletal muscle-type EC coupling. Previous work (Tanabe, T., Beam, K. G., Adams, B. A., Niidome, T., and Numa, S. (1990) Nature 346, 567-569) revealed that the loop between repeats II and III (II-III loop) is an important determinant of skeletal-type EC coupling. In the present study we have further dissected the regions of the II-III loop critical for skeletal-type EC coupling by expression of cDNA constructs in dysgenic myotubes. Because Ser687 of the skeletal II-III loop has been reported to be rapidly phosphorylated in vitro, we substituted this serine with alanine, the corresponding cardiac residue. This alanine-substituted skeletal DHPR retained the ability to mediate skeletal-type EC coupling. Weak skeletal-type EC coupling was produced by a chimeric DHPR, which was entirely cardiac except for a small amount of skeletal sequence (residues 725-742) in the II-III loop. Skeletal-type coupling was stronger when both residues 725-742 and adjacent residues were skeletal (e.g. a chimera containing skeletal residues 711-765). However, residues 725-742 appeared to be critical because skeletal-type coupling was not produced either by a chimera with skeletal residues 711-732 or by one with skeletal residues 734-765.


Assuntos
Canais de Cálcio/metabolismo , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Sequência de Aminoácidos , Cálcio/metabolismo , Canais de Cálcio/química , Canais de Cálcio Tipo L , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dados de Sequência Molecular , Contração Muscular , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Miocárdio/citologia , Fosforilação , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...