Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Hum Mutat ; 39(9): 1161-1172, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29858533

RESUMO

Mutations in FLNC for a long time are known in connection to neuromuscular disorders and only recently were described in association with various cardiomyopathies. Here, we report a new clinical phenotype of filaminopathy in four unrelated patients with early-onset restrictive cardiomyopathy (RCM) in combination with congenital myopathy due to FLNC mutations (NM_001458.4:c.3557C>T, p.A1186V, rs1114167361 in three probands and c.[3547G>C; 3548C>T], p.A1183L, rs1131692185 in one proband). In all cases, concurrent myopathy was confirmed by neurological examination, electromyography, and morphological studies. Three of the patients also presented with arthrogryposis. The pathogenicity of the described missense variants was verified by cellular and morphological studies and by in vivo modeling in zebrafish. Combination of in silico and experimental approaches revealed that FLNC missense variants localized in Ig-loop segments often lead to development of RCM. The described FLNC mutations associated with early-onset RCMP extend cardiac spectrum of filaminopathies and facilitate the differential diagnosis of restrictive cardiac phenotype associated with neuromuscular involvement in children.


Assuntos
Cardiomiopatia Restritiva/genética , Anormalidades Congênitas/genética , Filaminas/genética , Doenças Musculares/genética , Adolescente , Cardiomiopatia Restritiva/fisiopatologia , Pré-Escolar , Anormalidades Congênitas/fisiopatologia , Análise Mutacional de DNA , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Doenças Musculares/fisiopatologia , Mutação , Linhagem , Fenótipo
2.
Pflugers Arch ; 470(4): 681-691, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29380055

RESUMO

This study aims to improve the classification of smooth muscle types to better understand their normal and pathological functional phenotypes. Four different smooth muscle tissues (aorta, muscular arteries, intestine, urinary bladder) with a 5-fold difference in maximal shortening velocity were obtained from mice and classified according to expression of the inserted myosin heavy chain (SMHC-B). Western blotting and quantitative PCR analyses were used to determine 15 metabolic and 8 cell signaling key components in each tissue. The slow muscle type (aorta) with a 12 times lower SMHC-B had 6-fold lower expression of the phosphatase subunit MYPT1, a 7-fold higher expression of Rhokinase 1, and a 3-fold higher expression of the PKC target CPI17, compared to the faster (urinary bladder) smooth muscle. The slow muscle had higher expression of components involved in glucose uptake and glycolysis (type 1 glucose transporter, 3 times; hexokinase, 13 times) and in gluconeogenesis (phosphoenolpyruvate carboxykinase, 43 times), but lower expression of the metabolic sensing AMP-activated kinase, alpha 2 isoform (5 times). The slow type also had higher expression of enzymes involved in lipid metabolism (hormone-sensitive lipase, 10 times; lipoprotein lipase, 13 times; fatty acid synthase, 6 times; type 2 acetyl-coenzyme A carboxylase, 8 times). We present a refined division of smooth muscle into muscle types based on the analysis of contractile, metabolic, and signaling components. Slow compared to fast smooth muscle has a lower expression of the deactivating phosphatase and upregulated Ca2+ sensitizing pathways and is more adapted for sustained glucose and lipid metabolism.


Assuntos
Músculo Liso/metabolismo , Transdução de Sinais/fisiologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Aorta/metabolismo , Aorta/fisiologia , Cálcio/metabolismo , Feminino , Gluconeogênese/fisiologia , Glucose/metabolismo , Glicólise/fisiologia , Metabolismo dos Lipídeos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso/fisiologia , Cadeias Pesadas de Miosina/metabolismo , Isoformas de Proteínas/metabolismo , Regulação para Cima/fisiologia
3.
Acta Paediatr ; 107(4): 638-646, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29224235

RESUMO

AIM: Gather 'proof-of-concept' evidence of the adverse developmental potential of cotinine (a seemingly benign biomarker of recent nicotine/tobacco smoke exposure). METHODS: Pregnant C57 mice drank nicotine- or cotinine-laced water for 6 wks from conception (NPRE = 2% saccharin + 100 µg nicotine/mL; CPRE = 2% saccharin + 10 µg cotinine/mL) or 3 wks after birth (CPOST = 2% saccharin + 30 µg cotinine/mL). Controls drank 2% saccharin (CTRL). At 17 ± 1 weeks (male pups; CTRL n = 6; CPOST n = 6; CPRE n = 8; NPRE n = 9), we assessed (i) cardiovascular control during sleep; (ii) arterial reactivity ex vivo; and (iii) expression of genes involved in arterial constriction/dilation. RESULTS: Blood cotinine levels recapitulated those of passive smoker mothers' infants. Pups exposed to cotinine exhibited (i) mild bradycardia - hypotension at rest (p < 0.001); (ii) attenuated (CPRE , p < 0.0001) or reverse (CPOST ; p < 0.0001) BP stress reactivity; (iii) adrenergic hypocontractility (p < 0.0003), low protein kinase C (p < 0.001) and elevated adrenergic receptor mRNA (p < 0.05; all drug-treated arteries); and (iv) endothelial dysfunction (NPRE only). CONCLUSION: Cotinine has subtle, enduring developmental consequences. Some cardiovascular effects of nicotine can plausibly arise via conversion into cotinine. Low-level exposure to this metabolite may pose unrecognised perinatal risks. Adults must avoid inadvertently exposing a foetus or infant to cotinine as well as nicotine.


Assuntos
Pressão Sanguínea/fisiologia , Cotinina/metabolismo , Endotélio Vascular/fisiopatologia , Frequência Cardíaca/fisiologia , Troca Materno-Fetal/fisiologia , Nicotina/metabolismo , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Gravidez , Poluição por Fumaça de Tabaco/efeitos adversos
4.
Proc Natl Acad Sci U S A ; 110(40): 16241-6, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24046370

RESUMO

Thyroid hormone is a major regulator of thermogenesis, acting both in peripheral organs and on central autonomic pathways. Mice heterozygous for a point mutation in thyroid hormone receptor α1 display increased thermogenesis as a consequence of high sympathetic brown fat stimulation. Surprisingly, despite the hypermetabolism, their body temperature is not elevated. Here we show, using isolated tail arteries, that defective thyroid hormone receptor α1 signaling impairs acetylcholine-mediated vascular relaxation as well as phenylephrine-induced vasoconstriction. Using infrared thermography on conscious animals, we demonstrate that these defects severely interfere with appropriate peripheral heat conservation and dissipation, which in turn leads to compensatory alterations in brown fat activity. Consequently, when the vasoconstrictive defect in mice heterozygous for a point mutation in thyroid hormone receptor α1 was reversed with the selective α1-adrenergic agonist midodrine, the inappropriate heat loss over their tail surface was reduced, normalizing brown fat activity and energy expenditure. Our analyses demonstrate that thyroid hormone plays a key role in vascular heat conservation and dissipation processes, adding a unique aspect to its well-documented functions in thermoregulation. The data thus facilitate understanding of temperature hypersensitivity in patients with thyroid disorders. Moreover, the previously unrecognized connection between cardiovascular regulation and metabolic activity revealed in this study challenges the interpretation of several experimental paradigms and questions some of the currently derived hypotheses on the role of thyroid hormone in thermogenesis.


Assuntos
Tecido Adiposo Marrom/fisiologia , Regulação da Temperatura Corporal/fisiologia , Hipotireoidismo/fisiopatologia , Termogênese/fisiologia , Receptores alfa dos Hormônios Tireóideos/metabolismo , Tecido Adiposo Marrom/metabolismo , Análise de Variância , Animais , Temperatura Corporal , Camundongos , Mutação Puntual/genética , Reação em Cadeia da Polimerase em Tempo Real , Cauda/irrigação sanguínea , Termografia , Receptores alfa dos Hormônios Tireóideos/genética , Hormônios Tireóideos/metabolismo , Vasoconstrição/fisiologia
5.
FASEB J ; 28(4): 1593-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24371125

RESUMO

Sapje zebrafish carry a mutation in the dystrophin gene, which results in a premature stop codon, and a severe muscle phenotype. They display several of the structural characteristics of Duchenne muscular dystrophy (DMD). Ataluren (PTC124) is proposed to cause readthrough of premature stop codons and has been introduced as a potential treatment of genetic disorders. Clinical trials in DMD have shown promise, although with complex dose dependency. We have established physiology techniques, enabling high resolution of contractile function in skeletal muscle of zebrafish larvae. We aimed to provide a mechanical analysis of sapje larval muscle and examine effects of ataluren. Homozygous 5 d postfertilization (dpf) sapje larvae exhibited structural defects with 50% decrease in active tension. Ataluren (0.1-1 µM, 3-5 dpf) improved contractile function (~60% improvement of force at 0.5 µM) and dystrophin expression. Controls were not affected. Higher doses (5 µM, 35 µM) impaired contractile function, an effect also observed in controls, suggesting unspecific negative effects at high concentrations. In summary, Sapje larvae exhibit impaired contractile performance and provide a relevant DMD model for functional studies. Ataluren significantly improves skeletal muscle function in the sapje larvae, most likely reflecting an observed increase in dystrophin expression. The bell-shaped dose dependence in sapje resembles that previously reported in clinical DMD studies.


Assuntos
Distrofina/genética , Distrofia Muscular Animal/genética , Mutação , Oxidiazóis/farmacologia , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Western Blotting , Relação Dose-Resposta a Droga , Distrofina/metabolismo , Humanos , Larva/efeitos dos fármacos , Larva/genética , Larva/fisiologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/fisiopatologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatologia , Estresse Mecânico , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
6.
Neurourol Urodyn ; 34(2): 196-202, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25794194

RESUMO

AIM: To examine the role of protein kinase C (PKC) and non-muscle myosin in regulation of wall tension in the hypertrophied urinary bladder. METHODS: A partial urinary outflow obstruction was induced in the mouse. Tissue strips from sham operated controls and obstructed bladders were examined in vitro with quantitative gel electrophoresis, immunohistochemistry, and in vitro force recordings. RESULTS: Outlet obstruction (14-18 days) induced a significant growth of the bladder, 73 ± 6.13 mg compared to 19 ± 1 13 mg in sham operated controls. The hypertrophying bladder tissue had increased expression of non-muscle myosin B (SMemb) mainly localized to serosa and suburothelium. Direct activation of PKC with PDBu did not alter force in the control urinary bladder. In contrast, PDBu initiated a prominent and sustained contraction which had an increased sensitivity to the myosin type II inhibitor blebbistatin. CONCLUSIONS: PKC activates a significant contractile response in the wall of the hypertrophying urinary bladder, possibly supported by non-muscle myosin. This contractile component is not contributing to the physiological response to muscarinic stimulation, but might be separately regulated by other, yet unknown, mechanisms.


Assuntos
Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Músculo Liso/metabolismo , Miosina não Muscular Tipo IIB/efeitos dos fármacos , Miosina não Muscular Tipo IIB/metabolismo , Proteína Quinase C/metabolismo , Bexiga Urinária/metabolismo , Bexiga Urinária/patologia , Animais , Fenômenos Biomecânicos/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Hipertrofia , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Músculo Liso/efeitos dos fármacos , Dibutirato de 12,13-Forbol/farmacologia , Proteína Quinase C/efeitos dos fármacos , Bexiga Urinária/efeitos dos fármacos , Obstrução do Colo da Bexiga Urinária/metabolismo , Obstrução do Colo da Bexiga Urinária/patologia
7.
Dev Biol ; 381(2): 377-88, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23850871

RESUMO

The amyloid precursor protein (APP) is a transmembrane protein mostly recognized for its association with Alzheimer's disease. The physiological function of APP is still not completely understood much because of the redundancy between genes in the APP family. In this study we have used zebrafish to study the physiological function of the zebrafish APP homologue, appb, during development. We show that appb is expressed in post-mitotic neurons in the spinal cord. Knockdown of appb by 50-60% results in a behavioral phenotype with increased spontaneous coiling and prolonged touch-induced activity. The spinal cord motor neurons in these embryos show defective formation and axonal outgrowth patterning. Reduction in Appb also results in patterning defects and changed density of pre- and post-synapses in the neuromuscular junctions. Together, our data show that development of functional locomotion in zebrafish depends on a critical role of Appb in the patterning of motor neurons and neuromuscular junctions.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Sinapses Elétricas/metabolismo , Neurônios Motores/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Geneticamente Modificados/embriologia , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/metabolismo , Comportamento Animal , Padronização Corporal , Sinapses Elétricas/patologia , Embrião não Mamífero/metabolismo , Embrião não Mamífero/patologia , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Locomoção/fisiologia , Morfolinos/administração & dosagem , Neurônios Motores/patologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Neurogênese , Natação , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
8.
J Physiol ; 592(5): 915-26, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24297853

RESUMO

The role of the small GTP-binding protein Rac1 in smooth muscle contraction was examined using small molecule inhibitors (EHT1864, NSC23766) and a novel smooth muscle-specific, conditional, Rac1 knockout mouse strain. EHT1864, which affects nucleotide binding and inhibits Rac1 activity, concentration-dependently inhibited the contractile responses induced by several different modes of activation (high-K+, phenylephrine, carbachol and protein kinase C activation by phorbol-12,13-dibutyrate) in several different visceral (urinary bladder, ileum) and vascular (mesenteric artery, saphenous artery, aorta) smooth muscle tissues. This contractile inhibition was associated with inhibition of the Ca2+ transient. Knockout of Rac1 (with a 50% loss of Rac1 protein) lowered active stress in the urinary bladder and the saphenous artery consistent with a role of Rac1 in facilitating smooth muscle contraction. NSC23766, which blocks interaction between Rac1 and some guanine nucleotide exchange factors, specifically inhibited the α1 receptor responses (phenylephrine) in vascular tissues and potentiated prostaglandin F2α and thromboxane (U46619) receptor responses. The latter potentiating effect occurred at lowered intracellular [Ca2+]. These results show that Rac1 activity is required for active contraction in smooth muscle, probably via enabling an adequate Ca2+ transient. At the same time, specific agonists recruit Rac1 signalling via upstream modulators, resulting in either a potentiation of contraction via Ca2+ mobilization (α1 receptor stimulation) or an attenuated contraction via inhibition of Ca2+ sensitization (prostaglandin and thromboxane receptors).


Assuntos
Sinalização do Cálcio/fisiologia , Contração Isométrica/fisiologia , Músculo Liso/fisiologia , Neuropeptídeos/metabolismo , Transdução de Sinais/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Feminino , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropeptídeos/efeitos dos fármacos , Neuropeptídeos/genética , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/genética
9.
Nat Commun ; 15(1): 1950, 2024 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-38431640

RESUMO

In muscular dystrophies, muscle fibers loose integrity and die, causing significant suffering and premature death. Strikingly, the extraocular muscles (EOMs) are spared, functioning well despite the disease progression. Although EOMs have been shown to differ from body musculature, the mechanisms underlying this inherent resistance to muscle dystrophies remain unknown. Here, we demonstrate important differences in gene expression as a response to muscle dystrophies between the EOMs and trunk muscles in zebrafish via transcriptomic profiling. We show that the LIM-protein Fhl2 is increased in response to the knockout of desmin, plectin and obscurin, cytoskeletal proteins whose knockout causes different muscle dystrophies, and contributes to disease protection of the EOMs. Moreover, we show that ectopic expression of fhl2b can partially rescue the muscle phenotype in the zebrafish Duchenne muscular dystrophy model sapje, significantly improving their survival. Therefore, Fhl2 is a protective agent and a candidate target gene for therapy of muscular dystrophies.


Assuntos
Proteínas com Domínio LIM , Proteínas Musculares , Distrofia Muscular de Duchenne , Músculos Oculomotores , Animais , Proteínas do Citoesqueleto/metabolismo , Distrofina/genética , Expressão Ectópica do Gene , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Músculos Oculomotores/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas Musculares/metabolismo , Proteínas com Domínio LIM/metabolismo
10.
Front Physiol ; 14: 1275736, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38028806

RESUMO

Recent successful cardiac transplantation from pig to non-human primates and the first pig-to-human transplantation has put the focus on the properties of the pig heart. In contrast to the coronary arteries, the coronary veins are less well characterized and the aim was to examine the mechanical and pharmacological properties of coronary veins in comparison to the arteries. Vessel segments from the left anterior descending coronary artery (LAD) and the concomitant vein were isolated from pig hearts in cardioplegia and examined in vitro. The wall thickness, active tension and active stress at optimal circumference were lower in coronary veins, reflecting the lower intravascular pressure in vivo. Reverse transcription polymerase chain reaction (RT-PCR) analysis of myosin isoforms showed that the vein could be characterized as having a slower smooth muscle phenotype compared to the artery. Both vessel types contracted in response to the thromboxane agonist U46619 with EC50 values of about 20 nM. The artery contracted in response to acetylcholine. Precontracted arteries relaxed in noradrenaline and substance P. In contrast, the veins relaxed in acetylcholine, contracted in noradrenaline and were unresponsive to substance P. In conclusion, these results demonstrate significant differences between the coronary artery and vein in the smooth muscle properties and in the responses to sympathetic and parasympathetic stimuli.

11.
Front Cardiovasc Med ; 10: 1105257, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36891241

RESUMO

Stone heart (ischemic contracture) is a rare and serious condition observed in the heart after periods of warm ischemia. The underlying mechanisms are largely unknown and treatment options are lacking. In view of the possibilities for cardiac donation after circulatory death (DCD), introducing risks for ischemic damage, we have investigated stone heart in pigs. Following cessation of ventilation, circulatory death (systolic pressure <8 mmHg) occurred within 13.1 ± 1.2 min; and a stone heart, manifested with asystole, increased left ventricular wall thickness and stiffness, established after a further 17 ± 6 min. Adenosine triphosphate and phosphocreatine levels decreased by about 50% in the stone heart. Electron microscopy showed deteriorated structure with contraction bands, Z-line streaming and swollen mitochondria. Synchrotron based small angle X-ray scattering of trabecular samples from stone hearts revealed attachment of myosin to actin, without volume changes in the sarcomeres. Ca2+ sensitivity, determined in permeabilized muscle, was increased in stone heart samples. An in vitro model for stone heart, using isolated trabecular muscle exposed to hypoxia/zero glucose, exhibited the main characteristics of stone heart in whole animals, with a fall in high-energy phosphates and development of muscle contracture. The stone heart condition in vitro was significantly attenuated by the myosin inhibitor MYK-461 (Mavacamten). In conclusion, the stone heart is a hypercontracted state associated with myosin binding to actin and increased Ca2+ sensitivity. The hypercontractile state, once developed, is poorly reversible. The myosin inhibitor MYK-461, which is clinically approved for other indications, could be a promising venue for prevention.

12.
FASEB Bioadv ; 5(2): 71-84, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36816513

RESUMO

Excitability and contraction of cardiac muscle from brain-dead donors critically influence the success of heart transplantation. Membrane physiology, Ca2+-handling, and force production of cardiac muscle and the contractile properties of coronary arteries were studied in hearts of brain-dead pigs. Cardiac muscle and vascular function after 12 h brain death (decapitation between C2 and C3) were compared with properties of fresh tissue. In both isolated cardiomyocytes (whole-cell patch clamp) and trabecular muscle (conventional microelectrodes), action potential duration was shorter in brain dead, compared to controls. Cellular shortening and Ca2+ transients were attenuated in the brain dead, and linked to lower mRNA expression of L-type calcium channels and a slightly lower ICa,L, current, as well as to a lower expression of phospholamban. The current-voltage relationship and the current above the equilibrium potential of the inward K+ (IK1) channel were altered in the brain-dead group, associated with lower mRNA expression of the Kir2.2 channel. Delayed K+ currents were detected (IKr, IKs) and were not different between groups. The transient outward K+ current (Ito) was not observed in the pig heart. Coronary arteries exhibited increased contractility and sensitivity to the thromboxane analogue (U46619), and unaltered endothelial relaxation. In conclusion, brain death involves changes in cardiac cellular excitation which might lower contractility after transplantation. Changes in the inward rectifier K+ channel can be associated with an increased risk for arrhythmia. Increased reactivity of coronary arteries may lead to increased risk of vascular spasm, although endothelial relaxant function was well preserved.

13.
Nat Commun ; 14(1): 3312, 2023 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-37286550

RESUMO

Mutations in thyroid hormone receptor α1 (TRα1) cause Resistance to Thyroid Hormone α (RTHα), a disorder characterized by hypothyroidism in TRα1-expressing tissues including the heart. Surprisingly, we report that treatment of RTHα patients with thyroxine to overcome tissue hormone resistance does not elevate their heart rate. Cardiac telemetry in male, TRα1 mutant, mice indicates that such persistent bradycardia is caused by an intrinsic cardiac defect and not due to altered autonomic control. Transcriptomic analyses show preserved, thyroid hormone (T3)-dependent upregulation of pacemaker channels (Hcn2, Hcn4), but irreversibly reduced expression of several ion channel genes controlling heart rate. Exposure of TRα1 mutant male mice to higher maternal T3 concentrations in utero, restores altered expression and DNA methylation of ion channels, including Ryr2. Our findings indicate that target genes other than Hcn2 and Hcn4 mediate T3-induced tachycardia and suggest that treatment of RTHα patients with thyroxine in high dosage without concomitant tachycardia, is possible.


Assuntos
Síndrome da Resistência aos Hormônios Tireóideos , Tiroxina , Masculino , Animais , Camundongos , Tiroxina/uso terapêutico , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , Hormônios Tireóideos , Receptores alfa dos Hormônios Tireóideos/genética , Receptores alfa dos Hormônios Tireóideos/metabolismo , Mutação , Taquicardia/genética
14.
Dev Biol ; 354(1): 67-76, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21457708

RESUMO

Urodele amphibians, like the newt, are the "champions of regeneration" as they are able to regenerate many body parts and tissues. Previous experiments, however, have suggested that the newt heart has only a limited regeneration capacity, similar to the human heart. Using a novel, reproducible ventricular resection model, we show for the first time that adult newt hearts can fully regenerate without any evidence of scarring. This process is governed by increased proliferation and the up-regulation of cardiac transcription factors normally expressed during developmental cardiogenesis. Furthermore, we are able to identify cells within the newly regenerated regions of the myocardium that express the LIM-homeodomain protein Islet1 and GATA4, transcription factors found in cardiac progenitors. Information acquired from using the newt as a model organism may help to shed light on the regeneration deficits demonstrated in damaged human hearts.


Assuntos
Traumatismos Cardíacos/fisiopatologia , Coração/fisiopatologia , Regeneração , Salamandridae/fisiologia , Animais , Proliferação de Células , Fator de Transcrição GATA4/genética , Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas com Homeodomínio LIM , Microscopia Confocal , Microscopia de Fluorescência , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Salamandridae/genética , Fatores de Transcrição
15.
BJU Int ; 109(1): 132-40, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21883831

RESUMO

OBJECTIVE: To study muscarinic/purinergic receptor activation and Rho-kinase/protein kinase C (PKC) signalling during smooth muscle contraction in normal and hypertrophic mouse urinary bladders. METHODS: Partial urinary outflow obstruction was induced in adult female (10-12 weeks) C57Bl/6 mice and comparisons were made with sham-operated controls. Bladder preparations were examined in vitro. Expression of signalling proteins was examined using Western blot analysis. RESULTS: Obstructed bladders increased more than threefold in weight and were found to have enhanced muscarinic and attenuated purinergic components during nerve-induced contractions. The contractile response to carbachol was shifted towards lower concentrations of carbachol for the peak response and had a markedly enhanced sustained component. The amplitude of the α,ß-methylene ATP-induced responses was lowered. Rho-kinase inhibitor Y27632 (10 µM) inhibited peak and sustained contractile responses to carbachol in control bladders (peak by 38%; plateau 57%) and obstructed bladders (peak 37% plateau 47%). PKC inhibitor GF109203X (1 µM) inhibited carbachol contractions in controls (peak by 29%; plateau 29%) and obstructed bladders (peak 17%; plateau 12%). Inhibition by a similar extent was observed after nerve stimulation. Sensitivity to Ca(2+) in high-K(+) depolarized intact tissues increased in obstructed bladders. This increased receptor-independent Ca(2+)-sensitivity was abolished by Y27632. Tissue contents of the myosin-binding phosphatase subunit MYPT-1 and catalytic phosphatase subunit PP1ß, were decreased and the contents of RhoGDI, RhoA and CPI-17 increased. A decrease in the Rho-kinase isoform ROCK-1 was observed. CONCLUSION: Based on these results, one can speculate that Rho-kinase inhibition would preferentially target the pathological phasic activity in the urinary bladder rather than inhibit the physiological receptor-mediated bladder emptying.


Assuntos
Contração Muscular/fisiologia , Proteína Quinase C/metabolismo , Obstrução do Colo da Bexiga Urinária/enzimologia , Bexiga Urinária/fisiopatologia , Quinases Associadas a rho/metabolismo , Animais , Western Blotting , Modelos Animais de Doenças , Feminino , Hipertrofia/enzimologia , Hipertrofia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Bexiga Urinária/enzimologia , Bexiga Urinária/patologia , Obstrução do Colo da Bexiga Urinária/patologia , Obstrução do Colo da Bexiga Urinária/fisiopatologia
16.
J Theor Biol ; 297: 176-86, 2012 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-22108241

RESUMO

The main function of smooth muscle is to maintain/regulate the size of different hollow organs through contraction and relaxation. The magnitude of the active force during contraction is dependent on the number of attached cross-bridges, which can be linked to the overlap between the thin and thick filaments. The relevance of filament overlap and the active cross-bridges in smooth muscle is investigated through a mechanical model founded on Hill's three-element model. The mechanical model describes a sarcomere-equivalent contractile unit supported by structural observations with a distinct filament overlap and a realistic framework for the filament sliding behavior based on force-velocity experiments. The mechanical model is coupled to the four-state latch-model by Hai and Murphy to capture the electromechanical activation from intracellular calcium concentration to load-bearing cross-bridges. The model is fitted to isometric experiments performed on the pig carotid media and on isotonic quick-release experiments found in the literature. The proposed coupled mechanochemical model with the description of the filament overlap, which has a significant influence on the results, is able to predict isometric experimental data performed at different muscle lengths. The relevance of the filament overlap and the load-bearing cross-bridges is investigated through the model by simulating additional scenarios that has been documented in the literature.


Assuntos
Citoesqueleto de Actina/fisiologia , Contração Isométrica/fisiologia , Modelos Biológicos , Músculo Liso/fisiologia , Animais , Cálcio/metabolismo , Mecanotransdução Celular/fisiologia , Músculo Liso/metabolismo , Fosforilação , Estresse Mecânico , Sus scrofa
17.
EMBO J ; 26(21): 4535-45, 2007 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-17932484

RESUMO

Thyroid hormone, via its nuclear receptors TRalpha and TRbeta, controls metabolism by acting locally in peripheral tissues and centrally by regulating sympathetic signaling. We have defined aporeceptor regulation of metabolism by using mice heterozygous for a mutant TRalpha1 with low affinity to T3. The animals were hypermetabolic, showing strongly reduced fat depots, hyperphagia and resistance to diet-induced obesity accompanied by induction of genes involved in glucose handling and fatty acid metabolism in liver and adipose tissues. Increased lipid mobilization and beta-oxidation occurred in adipose tissues, whereas blockade of sympathetic signaling to brown adipose tissue normalized the metabolic phenotype despite a continued perturbed hormone signaling in this cell type. The results define a novel and important role for the TRalpha1 aporeceptor in governing metabolic homeostasis. Furthermore, the data demonstrate that a nuclear hormone receptor affecting sympathetic signaling can override its autonomous effects in peripheral tissues.


Assuntos
Receptores alfa dos Hormônios Tireóideos/fisiologia , Tecido Adiposo/metabolismo , Animais , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Lipídeos/química , Fígado/metabolismo , Metabolismo , Camundongos , Camundongos Transgênicos , Ligação Proteica , Receptores dos Hormônios Tireóideos/metabolismo , Transdução de Sinais , Sistema Nervoso Simpático/metabolismo , Receptores alfa dos Hormônios Tireóideos/metabolismo
18.
J Vasc Res ; 48(3): 206-18, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21099227

RESUMO

BACKGROUND/AIMS: Sepsis-induced lung injury is characterized by pulmonary hypertension, edema and deteriorated gas exchange. As in vivo studies have indicated that bacterial endotoxin predominantly induces a pulmonary venous constriction, we aimed to investigate effects of endotoxin on isolated porcine pulmonary vessels. METHODS: Pulmonary arteries and veins were examined using in vitro isometric force recordings. Endothelin-receptor protein expression and distribution were analyzed by Western blot and immunohistochemistry. Freshly isolated preparations and vessels incubated (24 h) with/without endotoxin (10 µg·ml(-1)) were compared. The contractile responses to phenylephrine, UK14.304, U46619, PGF(2α), endothelin-1 (ET-1) and sarafotoxin were recorded, as well as the relaxation in response to acetylcholine, isoproterenol and nitroprusside. RESULTS: In freshly isolated vessels, phenylephrine-induced contractions had a 5-times larger amplitude in arteries than in veins. The amplitude of the contractions in response to sarafotoxin was nearly 2 times larger in veins than in arteries, but there was no difference in responses to ET-1. Endotoxin markedly reduced phenylephrine-induced contractions in both arteries and veins, whereas the responses to ET-1 and sarafotoxin were augmented in veins only. No apparent changes in ET receptor expression or distribution were detected with Western blot or immunohistochemistry. CONCLUSION: Endotoxin differentially and selectively alters the contractile responses of porcine pulmonary vessels in vitro, towards a situation where the α-1 adrenergic responses of arteries are attenuated and the ET responses of veins are augmented. In situations with high adrenergic activity and high circulating ET levels, such as sepsis, these results may provide a mechanism contributing to pulmonary hypertension and edema formation.


Assuntos
Endotoxinas/farmacologia , Artéria Pulmonar/efeitos dos fármacos , Veias Pulmonares/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Animais , Western Blotting , Relação Dose-Resposta a Droga , Feminino , Imuno-Histoquímica , Masculino , Miografia , Técnicas de Cultura de Órgãos , Artéria Pulmonar/metabolismo , Veias Pulmonares/metabolismo , Receptores de Endotelina/efeitos dos fármacos , Receptores de Endotelina/metabolismo , Sus scrofa , Vasoconstritores/farmacologia , Vasodilatadores/farmacologia
19.
Circ Res ; 104(6): 813-21, 2009 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-19213955

RESUMO

Reduced cardiac output is one of the consequences of myocarditis. Bosentan, an endothelin-1 receptor (ET1R) antagonist, could be useful to reduce cardiac afterload, preserving cardiac output. In this study, we investigated the potential therapeutic use of bosentan in an animal model of viral myocarditis. Using a mouse model of coxsackievirus B3 (CVB3)-induced myocarditis, we demonstrated preserved ejection fraction (EF) and fractional shortening (FS) by treatment with bosentan (68+/-5.8% EF and 40+/-3.7% FS for treated versus 48+/-2.2% EF and 25+/-2.6% FS for controls; P=0.028). However, bosentan enhanced cardiac viral load (10.4+/-6.7% in the bosentan group versus 5.0+/-5.5% in control group; P=0.02), likely through enhancement of p38 mitogen-activated protein kinase (MAPK) phosphorylation (0.77+/-0.40% ATF2 activation in the bosentan group versus 0.03+/-0.02% in controls; P=0.0002), mediated by endothelin receptor type-A. We further demonstrate that a water soluble inhibitor of p38 MAPK, SB203580 HCl, is a potent inhibitor of virus replication in the heart (0.28% antisense viral genome stained area for 3 mg/kg dose versus 2.9% stained area for controls; P=0.01), attenuates CVB3-induced myocardial damage (blinded cardiac histopathologic scores of 1.8+/-1.6 and 2.05+/-1.2 for the 3 mg/kg and 10 mg/kg doses, respectively, versus 3.25+/-1.2 for the controls), and preserves cardiac function (69+/-3.5% EF for 3 mg/kg dose and 71+/-6.7% EF for 10 mg/kg dose versus 60+/-1.5% EF control; P=0.038 and P=0.045, as compared to control, respectively). Bosentan, a prescribed vasodilator, improves cardiac function but enhances viral load and myocarditis severity through ETRA mediated p38 MAPK activation; p38 MAPK is a desirable antiviral target. Caution must be exercised during treatment of suspected infectious myocarditis with supportive vasoactive remedies.


Assuntos
Anti-Hipertensivos/farmacologia , Infecções por Coxsackievirus/enzimologia , Infecções por Coxsackievirus/fisiopatologia , Antagonistas do Receptor de Endotelina A , Enterovirus Humano B , Miocardite/enzimologia , Miocardite/fisiopatologia , Volume Sistólico/efeitos dos fármacos , Sulfonamidas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Bosentana , Baixo Débito Cardíaco/tratamento farmacológico , Baixo Débito Cardíaco/enzimologia , Baixo Débito Cardíaco/fisiopatologia , Baixo Débito Cardíaco/virologia , Infecções por Coxsackievirus/tratamento farmacológico , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Humanos , Camundongos , Miocardite/tratamento farmacológico , Miocardite/virologia , Carga Viral/métodos
20.
Pulm Pharmacol Ther ; 24(4): 361-6, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21255673

RESUMO

Different mouse strains display different degrees of inflammation-induced airway hyperresponsiveness in vivo. It is not known whether these variations are attributable to distinct properties of the airway smooth muscle. Therefore, tracheal ring segments from C57BL/6 and BALB/c mice were exposed to three different pro-inflammatory stimuli for 4 days while maintained under tissue-culture conditions: tumour necrosis factor α (100 ng/ml), the Toll-like receptor (TLR) 3 agonist polyI:C (10 µg/ml), and the TLR4 agonist LPS (10 µg/ml). The contractile responses to carbachol, 5-hydroxytryptamine (5-HT) and bradykinin were assessed after culture. In addition, gene expression of TLR1-TLR9, pivotal inflammatory signal transduction proteins (jun-kinase, p38 and p65) and critical negative regulators of inflammation (A20, Itch, Tax1bp1 and RNF11) were studied in tracheal smooth muscle strips, fresh and following treatment for 4 days with LPS, from both strains. No differences between the strains were detected regarding the response of freshly isolated preparations to carbachol, 5-HT and bradykinin. After stimulation with pro-inflammatory mediators, contractions in response to 5-HT and bradykinin, but not to carbachol, were up-regulated. This up-regulation was markedly larger in BALB/c than in C57BL/6 segments and depended on the type of inflammatory stimulus. Expression of the genes investigated did not differ between the two strains. These findings indicate that strain differences in airway hyperresponsiveness can be linked to differences in the responsiveness of airway smooth muscle to pro-inflammatory mediators per se. The differences do not appear to be due to differential expression of TLR or common inflammatory transduction and repressor proteins.


Assuntos
Inflamação/fisiopatologia , Músculo Liso/fisiologia , Traqueia/fisiologia , Animais , Bradicinina/farmacologia , Carbacol/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Contração Muscular/efeitos dos fármacos , Serotonina/farmacologia , Transdução de Sinais , Especificidade da Espécie , Receptores Toll-Like/fisiologia , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA