Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38798483

RESUMEN

Fibrosis is a common outcome of numerous pathologies, including chronic kidney disease (CKD), a progressive renal function deterioration. Current approaches to target activated fibroblasts, key effector contributors to fibrotic tissue remodeling, lack specificity. Here, we report Gucy1α1 as a specific kidney fibroblast marker. Gucy1α1 levels significantly increased over the course of two clinically relevant murine CKD models and directly correlated with established fibrosis markers. Immunofluorescent (IF) imaging showed that Gucy1α1 comprehensively labelled cortical and medullary quiescent and activated fibroblasts in the control kidney and throughout injury progression, respectively. Unlike traditionally used markers platelet derived growth factor receptor beta (Pdgfrß) and vimentin (Vim), Gucy1α1 did not overlap with off-target populations such as podocytes. Notably, Gucy1α1 labelled kidney fibroblasts in both male and female mice. Furthermore, we observed elevated GUCY1α1 expression in the human fibrotic kidney and lung. Studies in the murine models of cardiac and liver fibrosis revealed Gucy1α1 elevation in activated Pdgfrß-, Vim- and alpha smooth muscle actin (αSma)-expressing fibroblasts paralleling injury progression and resolution. Overall, we demonstrate Gucy1α1 as an exclusive fibroblast marker in both sexes. Due to its multiorgan translational potential, GUCY1α1 might provide a novel promising strategy to specifically target and mechanistically examine fibroblasts.

2.
Cell Rep ; 43(5): 114149, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38678560

RESUMEN

Loss of muscle mass is a feature of chronic illness and aging. Here, we report that skeletal muscle-specific thrombospondin-1 transgenic mice (Thbs1 Tg) have profound muscle atrophy with age-dependent decreases in exercise capacity and premature lethality. Mechanistically, Thbs1 activates transforming growth factor ß (TGFß)-Smad2/3 signaling, which also induces activating transcription factor 4 (ATF4) expression that together modulates the autophagy-lysosomal pathway (ALP) and ubiquitin-proteasome system (UPS) to facilitate muscle atrophy. Indeed, myofiber-specific inhibition of TGFß-receptor signaling represses the induction of ATF4, normalizes ALP and UPS, and partially restores muscle mass in Thbs1 Tg mice. Similarly, myofiber-specific deletion of Smad2 and Smad3 or the Atf4 gene antagonizes Thbs1-induced muscle atrophy. More importantly, Thbs1-/- mice show significantly reduced levels of denervation- and caloric restriction-mediated muscle atrophy, along with blunted TGFß-Smad3-ATF4 signaling. Thus, Thbs1-mediated TGFß-Smad3-ATF4 signaling in skeletal muscle regulates tissue rarefaction, suggesting a target for atrophy-based muscle diseases and sarcopenia with aging.


Asunto(s)
Factor de Transcripción Activador 4 , Músculo Esquelético , Atrofia Muscular , Transducción de Señal , Proteína Smad2 , Proteína smad3 , Trombospondina 1 , Factor de Crecimiento Transformador beta , Animales , Masculino , Ratones , Factor de Transcripción Activador 4/metabolismo , Autofagia , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Trombospondina 1/metabolismo , Trombospondina 1/genética , Factor de Crecimiento Transformador beta/metabolismo
3.
Cells ; 12(17)2023 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-37681905

RESUMEN

RATIONALE: The adult cardiac extracellular matrix (ECM) is largely comprised of type I collagen. In addition to serving as the primary structural support component of the cardiac ECM, type I collagen also provides an organizational platform for other ECM proteins, matricellular proteins, and signaling components that impact cellular stress sensing in vivo. OBJECTIVE: Here we investigated how the content and integrity of type I collagen affect cardiac structure function and response to injury. METHODS AND RESULTS: We generated and characterized Col1a2-/- mice using standard gene targeting. Col1a2-/- mice were viable, although by young adulthood their hearts showed alterations in ECM mechanical properties, as well as an unanticipated activation of cardiac fibroblasts and induction of a progressive fibrotic response. This included augmented TGFß activity, increases in fibroblast number, and progressive cardiac hypertrophy, with reduced functional performance by 9 months of age. Col1a2-loxP-targeted mice were also generated and crossed with the tamoxifen-inducible Postn-MerCreMer mice to delete the Col1a2 gene in myofibroblasts with pressure overload injury. Interestingly, while germline Col1a2-/- mice showed gradual pathologic hypertrophy and fibrosis with aging, the acute deletion of Col1a2 from activated adult myofibroblasts showed a loss of total collagen deposition with acute cardiac injury and an acute reduction in pressure overload-induce cardiac hypertrophy. However, this reduction in hypertrophy due to myofibroblast-specific Col1a2 deletion was lost after 2 and 6 weeks of pressure overload, as fibrotic deposition accumulated. CONCLUSIONS: Defective type I collagen in the heart alters the structural integrity of the ECM and leads to cardiomyopathy in adulthood, with fibroblast expansion, activation, and alternate fibrotic ECM deposition. However, acute inhibition of type I collagen production can have an anti-fibrotic and anti-hypertrophic effect.


Asunto(s)
Cardiomiopatías , Colágeno Tipo I , Animales , Ratones , Cardiomegalia/genética , Colágeno Tipo I/genética , Fibrosis
4.
Sci Adv ; 9(34): eadi2767, 2023 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-37624892

RESUMEN

Mitochondrial permeability transition pore (MPTP) formation contributes to ischemia-reperfusion injury in the heart and several degenerative diseases, including muscular dystrophy (MD). MD is a family of genetic disorders characterized by progressive muscle necrosis and premature death. It has been proposed that the MPTP has two molecular components, the adenine nucleotide translocase (ANT) family of proteins and an unknown component that requires the chaperone cyclophilin D (CypD) to activate. This model was examined in vivo by deleting the gene encoding ANT1 (Slc25a4) or CypD (Ppif) in a δ-sarcoglycan (Sgcd) gene-deleted mouse model of MD, revealing that dystrophic mice lacking Slc25a4 were partially protected from cell death and MD pathology. Dystrophic mice lacking both Slc25a4 and Ppif together were almost completely protected from necrotic cell death and MD disease. This study provides direct evidence that ANT1 and CypD are required MPTP components governing in vivo cell death, suggesting a previously unrecognized therapeutic approach in MD and other necrotic diseases.


Asunto(s)
Distrofias Musculares , Animales , Ratones , Necrosis , Muerte Celular , Peptidil-Prolil Isomerasa F , Modelos Animales de Enfermedad
5.
Proc Natl Acad Sci U S A ; 120(19): e2213696120, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37126682

RESUMEN

To better understand the genetic basis of heart disease, we identified a variant in the Flightless-I homolog (FLII) gene that generates a R1243H missense change and predisposes to cardiac remodeling across multiple previous human genome-wide association studies (GWAS). Since this gene is of unknown function in the mammalian heart we generated gain- and loss-of-function genetically altered mice, as well as knock-in mice with the syntenic R1245H amino acid substitution, which showed that Flii protein binds the sarcomeric actin thin filament and influences its length. Deletion of Flii from the heart, or mice with the R1245H amino acid substitution, show cardiomyopathy due to shortening of the actin thin filaments. Mechanistically, Flii is a known actin binding protein that we show associates with tropomodulin-1 (TMOD1) to regulate sarcomere thin filament length. Indeed, overexpression of leiomodin-2 in the heart, which lengthens the actin-containing thin filaments, partially rescued disease due to heart-specific deletion of Flii. Collectively, the identified FLII human variant likely increases cardiomyopathy risk through an alteration in sarcomere structure and associated contractile dynamics, like other sarcomere gene-based familial cardiomyopathies.


Asunto(s)
Actinas , Cardiomiopatías , Humanos , Animales , Ratones , Actinas/metabolismo , Sarcómeros/metabolismo , Estudio de Asociación del Genoma Completo , Citoesqueleto de Actina/metabolismo , Cardiomiopatías/metabolismo , Mamíferos/genética , Proteínas de Microfilamentos/metabolismo , Transactivadores/metabolismo , Tropomodulina/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas Musculares/metabolismo
6.
Front Physiol ; 14: 1054169, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36733907

RESUMEN

Introduction: The ribosomal protein L3-like (RPL3L) is a heart and skeletal muscle-specific ribosomal protein and paralogue of the more ubiquitously expressed RPL3 protein. Mutations in the human RPL3L gene are linked to childhood cardiomyopathy and age-related atrial fibrillation, yet the function of RPL3L in the mammalian heart remains unknown. Methods and Results: Here, we observed that mouse cardiac ventricles express RPL3 at birth, where it is gradually replaced by RPL3L in adulthood but re-expressed with induction of hypertrophy in adults. Rpl3l gene-deleted mice were generated to examine the role of this gene in the heart, although Rpl3l -/- mice showed no overt changes in cardiac structure or function at baseline or after pressure overload hypertrophy, likely because RPL3 expression was upregulated and maintained in adulthood. mRNA expression analysis and ribosome profiling failed to show differences between the hearts of Rpl3l null and wild type mice in adulthood. Moreover, ribosomes lacking RPL3L showed no differences in localization within cardiomyocytes compared to wild type controls, nor was there an alteration in cardiac tissue ultrastructure or mitochondrial function in adult Rpl3l -/- mice. Similarly, overexpression of either RPL3 or RPL3L with adeno-associated virus -9 in the hearts of mice did not cause discernable pathology. However, by 18 months of age Rpl3l -/- null mice had significantly smaller hearts compared to wild type littermates. Conclusion: Thus, deletion of Rpl3l forces maintenance of RPL3 expression within the heart that appears to fully compensate for the loss of RPL3L, although older Rpl3l -/- mice showed a mild but significant reduction in heart weight.

7.
Cell Stem Cell ; 30(1): 96-111.e6, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36516837

RESUMEN

The efficacy and safety of gene-therapy strategies for indications like tissue damage hinge on precision; yet, current methods afford little spatial or temporal control of payload delivery. Here, we find that tissue-regeneration enhancer elements (TREEs) isolated from zebrafish can direct targeted, injury-associated gene expression from viral DNA vectors delivered systemically in small and large adult mammalian species. When employed in combination with CRISPR-based epigenome editing tools in mice, zebrafish TREEs stimulated or repressed the expression of endogenous genes after ischemic myocardial infarction. Intravenously delivered recombinant AAV vectors designed with a TREE to direct a constitutively active YAP factor boosted indicators of cardiac regeneration in mice and improved the function of the injured heart. Our findings establish the application of contextual enhancer elements as a potential therapeutic platform for spatiotemporally controlled tissue regeneration in mammals.


Asunto(s)
Elementos de Facilitación Genéticos , Terapia Genética , Corazón , Infarto del Miocardio , Miocitos Cardíacos , Regeneración , Animales , Ratones , Proliferación Celular , Corazón/fisiología , Infarto del Miocardio/genética , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Pez Cebra/genética , Terapia Genética/métodos , Regeneración/genética
8.
Mol Metab ; 62: 101528, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35717025

RESUMEN

OBJECTIVE: Mitochondrial capacity is critical to adapt the high energy demand of the heart to circadian oscillations and diseased states. Glucocorticoids regulate the circadian cycle of energy metabolism, but little is known about how circadian timing of exogenous glucocorticoid dosing directly regulates heart metabolism through cardiomyocyte-autonomous mechanisms. While chronic once-daily intake of glucocorticoids promotes metabolic stress and heart failure, we recently discovered that intermittent once-weekly dosing of exogenous glucocorticoids promoted muscle metabolism in normal and obese skeletal muscle. However, the effects of glucocorticoid intermittence on heart metabolism and heart failure remain unknown. Here we investigated the extent to which circadian time of dosing regulates the effects of the glucocorticoid prednisone in heart metabolism and function in conditions of single pulse or chronic intermittent dosing. METHODS AND RESULTS: In WT mice, we found that prednisone improved cardiac content of NAD+ and ATP with light-phase dosing (ZT0), while the effects were blocked by dark-phase dosing (ZT12). The drug effects on mitochondrial function were cardiomyocyte-autonomous, as shown by inducible cardiomyocyte-restricted glucocorticoid receptor (GR) ablation, and depended on an intact cardiomyocyte clock, as shown by inducible cardiomyocyte-restricted ablation of Brain and Muscle ARNT-like 1 (BMAL1). Conjugating time-of-dosing with chronic intermittence, we found that once-weekly prednisone improved metabolism and function in heart after myocardial injury dependent on circadian time of intake, i.e. with light-phase but not dark-phase dosing. CONCLUSIONS: Our study identifies cardiac-autonomous mechanisms through which circadian-specific intermittent dosing reconverts glucocorticoid drugs to metabolic boosters for the heart.


Asunto(s)
Relojes Circadianos , Insuficiencia Cardíaca , Animales , Relojes Circadianos/fisiología , Glucocorticoides/metabolismo , Glucocorticoides/farmacología , Insuficiencia Cardíaca/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Prednisona/metabolismo , Prednisona/farmacología
10.
Nat Commun ; 12(1): 3928, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34168130

RESUMEN

The thrombospondin (Thbs) family of secreted matricellular proteins are stress- and injury-induced mediators of cellular attachment dynamics and extracellular matrix protein production. Here we show that Thbs1, but not Thbs2, Thbs3 or Thbs4, induces lethal cardiac atrophy when overexpressed. Mechanistically, Thbs1 binds and activates the endoplasmic reticulum stress effector PERK, inducing its downstream transcription factor ATF4 and causing lethal autophagy-mediated cardiac atrophy. Antithetically, Thbs1-/- mice develop greater cardiac hypertrophy with pressure overload stimulation and show reduced fasting-induced atrophy. Deletion of Thbs1 effectors/receptors, including ATF6α, CD36 or CD47 does not diminish Thbs1-dependent cardiac atrophy. However, deletion of the gene encoding PERK in Thbs1 transgenic mice blunts the induction of ATF4 and autophagy, and largely corrects the lethal cardiac atrophy. Finally, overexpression of PERK or ATF4 using AAV9 gene-transfer similarly promotes cardiac atrophy and lethality. Hence, we identified Thbs1-mediated PERK-eIF2α-ATF4-induced autophagy as a critical regulator of cardiomyocyte size in the stressed heart.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Miocardio/patología , Trombospondinas/metabolismo , eIF-2 Quinasa/metabolismo , Factor de Transcripción Activador 4/genética , Animales , Atrofia , Autofagia/fisiología , Cardiomegalia/genética , Cardiomegalia/patología , Estrés del Retículo Endoplásmico/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Expresión Génica , Lisosomas/metabolismo , Masculino , Ratones Transgénicos , Miocitos Cardíacos/patología , Proteolisis , Trombospondinas/genética , eIF-2 Quinasa/genética
11.
J Vis Exp ; (171)2021 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-34057439

RESUMEN

Cardiovascular disease is the most prevalent cause of mortality worldwide and is often marked by heightened cardiac fibrosis that can lead to increased ventricular stiffness with altered cardiac function. This increase in cardiac ventricular fibrosis is due to activation of resident fibroblasts, although how these cells operate within the 3-dimensional (3-D) heart, at baseline or after activation, is not well understood. To examine how fibroblasts contribute to heart disease and their dynamics in the 3-D heart, a refined CLARITY-based tissue clearing and imaging method was developed that shows fluorescently labeled cardiac fibroblasts within the entire mouse heart. Tissue resident fibroblasts were genetically labeled using Rosa26-loxP-eGFP florescent reporter mice crossed with the cardiac fibroblast expressing Tcf21-MerCreMer knock-in line. This technique was used to observe fibroblast localization dynamics throughout the entire adult left ventricle in healthy mice and in fibrotic mouse models of heart disease. Interestingly, in one injury model, unique patterns of cardiac fibroblasts were observed in the injured mouse heart that followed bands of wrapped fibers in the contractile direction. In ischemic injury models, fibroblast death occurred, followed by repopulation from the infarct border zone. Collectively, this refined cardiac tissue clarifying technique and digitized imaging system allows for 3-D visualization of cardiac fibroblasts in the heart without the limitations of antibody penetration failure or previous issues surrounding lost fluorescence due to tissue processing.


Asunto(s)
Modelos Animales de Enfermedad , Fibroblastos , Cardiopatías , Animales , Fibroblastos/patología , Fibrosis , Corazón , Ratones , Ratones Endogámicos C57BL , Miocardio/patología
12.
Circ Res ; 127(3): 379-390, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32299299

RESUMEN

RATIONALE: Mitochondrial Ca2+ loading augments oxidative metabolism to match functional demands during times of increased work or injury. However, mitochondrial Ca2+ overload also directly causes mitochondrial rupture and cardiomyocyte death during ischemia-reperfusion injury by inducing mitochondrial permeability transition pore opening. The MCU (mitochondrial Ca2+ uniporter) mediates mitochondrial Ca2+ influx, and its activity is modulated by partner proteins in its molecular complex, including the MCUb subunit. OBJECTIVE: Here, we sought to examine the function of the MCUb subunit of the MCU-complex in regulating mitochondria Ca2+ influx dynamics, acute cardiac injury, and long-term adaptation after ischemic injury. METHODS AND RESULTS: Cardiomyocyte-specific MCUb overexpressing transgenic mice and Mcub gene-deleted (Mcub-/-) mice were generated to dissect the molecular function of this protein in the heart. We observed that MCUb protein is undetectable in the adult mouse heart at baseline, but mRNA and protein are induced after ischemia-reperfusion injury. MCUb overexpressing mice demonstrated inhibited mitochondrial Ca2+ uptake in cardiomyocytes and partial protection from ischemia-reperfusion injury by reducing mitochondrial permeability transition pore opening. Antithetically, deletion of the Mcub gene exacerbated pathological cardiac remodeling and infarct expansion after ischemic injury in association with greater mitochondrial Ca2+ uptake. Furthermore, hindlimb remote ischemic preconditioning induced MCUb expression in the heart, which was associated with decreased mitochondrial Ca2+ uptake, collectively suggesting that induction of MCUb protein in the heart is protective. Similarly, mouse embryonic fibroblasts from Mcub-/- mice were more sensitive to Ca2+ overload. CONCLUSIONS: Our studies suggest that Mcub is a protective cardiac inducible gene that reduces mitochondrial Ca2+ influx and permeability transition pore opening after ischemic injury to reduce ongoing pathological remodeling.


Asunto(s)
Calcio/metabolismo , Miembro Posterior/irrigación sanguínea , Proteínas de la Membrana/metabolismo , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/metabolismo , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Remodelación Ventricular , Animales , Señalización del Calcio , Muerte Celular , Línea Celular , Modelos Animales de Enfermedad , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Precondicionamiento Isquémico , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Cardíacas/patología , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Proteínas Mitocondriales/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/patología
14.
Nature ; 577(7790): 405-409, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31775156

RESUMEN

Clinical trials using adult stem cells to regenerate damaged heart tissue continue to this day1,2, despite ongoing questions of efficacy and a lack of mechanistic understanding of the underlying biological effect3. The rationale for these cell therapy trials is derived from animal studies that show a modest but reproducible improvement in cardiac function in models of cardiac ischaemic injury4,5. Here we examine the mechanistic basis for cell therapy in mice after ischaemia-reperfusion injury, and find that-although heart function is enhanced-it is not associated with the production of new cardiomyocytes. Cell therapy improved heart function through an acute sterile immune response characterized by the temporal and regional induction of CCR2+ and CX3CR1+ macrophages. Intracardiac injection of two distinct types of adult stem cells, cells killed by freezing and thawing or a chemical inducer of the innate immune response all induced a similar regional accumulation of CCR2+ and CX3CR1+ macrophages, and provided functional rejuvenation to the heart after ischaemia-reperfusion injury. This selective macrophage response altered the activity of cardiac fibroblasts, reduced the extracellular matrix content in the border zone and enhanced the mechanical properties of the injured area. The functional benefit of cardiac cell therapy is thus due to an acute inflammatory-based wound-healing response that rejuvenates the infarcted area of the heart.


Asunto(s)
Inmunidad Innata , Miocitos Cardíacos/inmunología , Trasplante de Células Madre , Células Madre , Animales , Diferenciación Celular , Femenino , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/trasplante , Rejuvenecimiento
15.
Sci Adv ; 5(8): eaaw4597, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31489369

RESUMEN

The mitochondrial permeability transition pore (MPTP) has resisted molecular identification. The original model of the MPTP that proposed the adenine nucleotide translocator (ANT) as the inner membrane pore-forming component was challenged when mitochondria from Ant1/2 double null mouse liver still had MPTP activity. Because mice express three Ant genes, we reinvestigated whether the ANTs comprise the MPTP. Liver mitochondria from Ant1, Ant2, and Ant4 deficient mice were highly refractory to Ca2+-induced MPTP formation, and when also given cyclosporine A (CsA), the MPTP was completely inhibited. Moreover, liver mitochondria from mice with quadruple deletion of Ant1, Ant2, Ant4, and Ppif (cyclophilin D, target of CsA) lacked Ca2+-induced MPTP formation. Inner-membrane patch clamping in mitochondria from Ant1, Ant2, and Ant4 triple null mouse embryonic fibroblasts showed a loss of MPTP activity. Our findings suggest a model for the MPTP consisting of two distinct molecular components: The ANTs and an unknown species requiring CypD.


Asunto(s)
Nucleótidos de Adenina/genética , Mitocondrias/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Necrosis por Permeabilidad de la Transmembrana Mitocondrial/genética , Peptidil-Prolil Isomerasa F/genética , Eliminación de Secuencia/genética , Animales , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Noqueados , Poro de Transición de la Permeabilidad Mitocondrial
16.
JCI Insight ; 52019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30964448

RESUMEN

Mitogen-activated protein kinase (MAPK) signaling consists of an array of successively acting kinases. The extracellular signal-regulated kinases 1/2 (ERK1/2) are major components of the greater MAPK cascade that transduce growth factor signaling at the cell membrane. Here we investigated ERK1/2 signaling in skeletal muscle homeostasis and disease. Using mouse genetics, we observed that the muscle-specific expression of a constitutively active MEK1 mutant promotes greater ERK1/2 signaling that mediates fiber-type switching to a slow, oxidative phenotype with type I myosin heavy chain expression. Using a conditional and temporally regulated Cre strategy as well as Mapk1 (ERK2) and Mapk3 (ERK1) genetically targeted mice, MEK1-ERK2 signaling was shown to underlie this fast-to-slow fiber type switching in adult skeletal muscle as well as during development. Physiologic assessment of these activated MEK1-ERK1/2 mice showed enhanced metabolic activity and oxygen consumption with greater muscle fatigue resistance. Moreover, induction of MEK1-ERK1/2 signaling increased dystrophin and utrophin protein expression in a mouse model of limb-girdle muscle dystrophy and protected myofibers from damage. In summary, sustained MEK1-ERK1/2 activity in skeletal muscle produces a fast-to-slow fiber-type switch that protects from muscular dystrophy, suggesting a therapeutic approach to enhance the metabolic effectiveness of muscle and protect from dystrophic disease.


Asunto(s)
Sistema de Señalización de MAP Quinasas/genética , Fatiga Muscular/genética , Fibras Musculares de Contracción Rápida/metabolismo , Fibras Musculares de Contracción Lenta/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/genética , Consumo de Oxígeno/genética , Animales , Modelos Animales de Enfermedad , Distrofina/metabolismo , MAP Quinasa Quinasa 1/genética , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/fisiopatología , Índice de Severidad de la Enfermedad , Utrofina/metabolismo
17.
J Biol Chem ; 294(22): 8918-8929, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31006653

RESUMEN

Valosin-containing protein (VCP), also known as p97, is an ATPase with diverse cellular functions, although the most highly characterized is targeting of misfolded or aggregated proteins to degradation pathways, including the endoplasmic reticulum-associated degradation (ERAD) pathway. However, how VCP functions in the heart has not been carefully examined despite the fact that human mutations in VCP cause Paget disease of bone and frontotemporal dementia, an autosomal dominant multisystem proteinopathy that includes disease in the heart, skeletal muscle, brain, and bone. Here we generated heart-specific transgenic mice overexpressing WT VCP or a VCPK524A mutant with deficient ATPase activity. Transgenic mice overexpressing WT VCP exhibit normal cardiac structure and function, whereas mutant VCP-overexpressing mice develop cardiomyopathy. Mechanistically, mutant VCP-overexpressing hearts up-regulate ERAD complex components and have elevated levels of ubiquitinated proteins prior to manifestation of cardiomyopathy, suggesting dysregulation of ERAD and inefficient clearance of proteins targeted for proteasomal degradation. The hearts of mutant VCP transgenic mice also exhibit profound defects in cardiomyocyte nuclear morphology with increased nuclear envelope proteins and nuclear lamins. Proteomics revealed overwhelming interactions of endogenous VCP with ribosomal, ribosome-associated, and RNA-binding proteins in the heart, and impairment of cardiac VCP activity resulted in aggregation of large ribosomal subunit proteins. These data identify multifactorial functions and diverse mechanisms whereby VCP regulates cardiomyocyte protein and RNA quality control that are critical for cardiac homeostasis, suggesting how human VCP mutations negatively affect the heart.


Asunto(s)
Cardiomiopatías/patología , Corazón/fisiología , Miocardio/metabolismo , Proteína que Contiene Valosina/metabolismo , Animales , Cardiomiopatías/metabolismo , Células Cultivadas , Degradación Asociada con el Retículo Endoplásmico , Laminas/metabolismo , Ratones , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteínas Nucleares/metabolismo , Subunidades de Proteína/metabolismo , Proteínas de Unión al ARN/metabolismo , Ratas , Proteínas Ribosómicas/metabolismo , Ubiquitinación , Proteína que Contiene Valosina/genética
18.
Nat Commun ; 10(1): 76, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30622267

RESUMEN

Thrombospondins (Thbs) are a family of five secreted matricellular glycoproteins in vertebrates that broadly affect cell-matrix interaction. While Thbs4 is known to protect striated muscle from disease by enhancing sarcolemmal stability through increased integrin and dystroglycan attachment complexes, here we show that Thbs3 antithetically promotes sarcolemmal destabilization by reducing integrin function, augmenting disease-induced decompensation. Deletion of Thbs3 in mice enhances integrin membrane expression and membrane stability, protecting the heart from disease stimuli. Transgene-mediated overexpression of α7ß1D integrin in the heart ameliorates the disease predisposing effects of Thbs3 by augmenting sarcolemmal stability. Mechanistically, we show that mutating Thbs3 to contain the conserved RGD integrin binding domain normally found in Thbs4 and Thbs5 now rescues the defective expression of integrins on the sarcolemma. Thus, Thbs proteins mediate the intracellular processing of integrin plasma membrane attachment complexes to regulate the dynamics of cellular remodeling and membrane stability.


Asunto(s)
Cardiomiopatías/patología , Integrinas/metabolismo , Sarcolema/patología , Trombospondinas/metabolismo , Animales , Células COS , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/etiología , Células Cultivadas , Chlorocebus aethiops , Modelos Animales de Enfermedad , Distroglicanos/metabolismo , Ecocardiografía , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Miocitos Cardíacos , Cultivo Primario de Células , Dominios y Motivos de Interacción de Proteínas/genética , Ratas , Ratas Sprague-Dawley , Sarcolema/metabolismo , Trombospondinas/genética
19.
BMC Health Serv Res ; 20(1): 2, 2019 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-31888611

RESUMEN

BACKGROUND: Clinical supervision is recommended for allied health professionals for the purpose of supporting them in their professional role, continued professional development and ensuring patient safety and high quality care. The aim of this mixed methods study was to explore allied health professionals' perceptions about the aspects of clinical supervision that can facilitate effective clinical supervision. METHODS: Individual semi-structured interviews were conducted on a purposive sample of 38 allied health professionals working in a metropolitan public hospital. Qualitative analysis was completed using an interpretive description approach. To enable triangulation of qualitative data, a quantitative descriptive survey of clinical supervision effectiveness was also conducted using the Manchester Clinical Supervision Scale (MCSS-26). RESULTS: Three main themes emerged from qualitative analysis: Allied health professionals reported that clinical supervision was most effective when their professional development was the focus of clinical supervision; the supervisor possessed the skills and attributes required to facilitate a constructive supervisory relationship; and the organisation provided an environment that facilitated this relationship together with their own professional development. Three subthemes also emerged within each of the main themes: the importance of the supervisory relationship; prioritisation of clinical supervision relative to other professional duties; and flexibility of supervision models, processes and approaches to clinical supervision. The mean MCSS-26 score was 79.2 (95%CI 73.7 to 84.3) with scores ranging from 44 to 100. MCSS-26 results converged with the qualitative findings with participants reporting an overall positive experience with clinical supervision. CONCLUSIONS: The factors identified by allied health professionals that influenced the effectiveness of their clinical supervision were mostly consistent among the professions. However, allied health professionals reported using models of clinical supervision that best suited their profession's role and learning style. This highlighted the need for flexible approaches to allied health clinical supervision that should be reflected in clinical supervision policies and guidelines. Many of the identified factors that influence the effectiveness of clinical supervision of allied health professionals can be influenced by health organisations.


Asunto(s)
Técnicos Medios en Salud , Administración de Personal en Hospitales , Actitud del Personal de Salud , Encuestas de Atención de la Salud , Humanos , Entrevistas como Asunto , Rol Profesional , Investigación Cualitativa , Desarrollo de Personal
20.
J Am Heart Assoc ; 7(20): e010013, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30371263

RESUMEN

Background Transforming growth factor beta ( TGF -ß) is an important cytokine in mediating the cardiac fibrosis that often accompanies pathogenic cardiac remodeling. Cardiomyocyte-specific expression of a mutant αB-crystallin (Cry ABR120G), which causes human desmin-related cardiomyopathy, results in significant cardiac fibrosis. During onset of fibrosis, fibroblasts are activated to the so-called myofibroblast state and TGF -ß binding mediates an essential signaling pathway underlying this process. Here, we test the hypothesis that fibroblast-based TGF -ß signaling can result in significant cardiac fibrosis in a disease model of cardiac proteotoxicity that has an exclusive cardiomyocyte-based etiology. Methods and Results Against the background of cardiomyocyte-restricted expression of Cry ABR120G, we have partially ablated TGF -ß signaling in cardiac myofibroblasts to observe whether cardiac fibrosis is reduced despite the ongoing pathogenic stimulus of Cry ABR120G production. Transgenic Cry ABR120G mice were crossed with mice containing a floxed allele of TGF -ß receptor 2 ( Tgfbr2 f/f). The double transgenic animals were subsequently crossed to another transgenic line in which Cre expression was driven from the periostin locus ( Postn) so that Tgfbr2 would be ablated with myofibroblast conversion. Structural and functional assays were then used to determine whether general fibrosis was affected and cardiac function rescued in Cry ABR120G mice lacking Tgfbr2 in the myofibroblasts. Ablation of myofibroblast specific TGF -ß signaling led to decreased morbidity in a proteotoxic disease resulting from cardiomyocyte autonomous expression of Cry ABR120G. Cardiac fibrosis was decreased and hypertrophy was also significantly attenuated, with a significant improvement in survival probability over time, even though the primary proteotoxic insult continued. Conclusions Myofibroblast-targeted knockdown of Tgfbr2 signaling resulted in reduced fibrosis and improved cardiac function, leading to improved probability of survival.


Asunto(s)
Miocardio/patología , Miofibroblastos/fisiología , Factor de Crecimiento Transformador beta/fisiología , Análisis de Varianza , Animales , Cardiomiopatías/patología , Modelos Animales de Enfermedad , Femenino , Fibroblastos/fisiología , Fibrosis/etiología , Cardiopatías/patología , Masculino , Ratones Transgénicos , Distrofias Musculares/patología , Miocitos Cardíacos/fisiología , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal/fisiología , Cadena B de alfa-Cristalina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...