Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Pharm Educ ; 88(8): 100730, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38852678

RESUMEN

Over the past 2 decades, the Academy has witnessed an increase in new colleges and schools of pharmacy and, simultaneously, a decrease in student applications, resulting in a decline in enrollment across most institutions. Although the number of students pursuing a Doctor of Pharmacy degree has been dropping, the Academy is responsible for bolstering recruitment to effectively prepare a robust pharmacy workforce to care for our ever-growing and complex patient populations. The 2023-2024 Student Affairs Committee (SAC) was convened to explore new ideas, develop innovative strategies, and gather supportive resources that can be utilized by colleges and schools of pharmacy to attract students to the pharmacy profession. The SAC was charged with developing a framework for a video mini-series that utilizes the art of storytelling to promote the pharmacy profession to prospective students. Secondarily, the SAC was charged with developing a plan to engage with students who apply but do not ultimately get accepted into nonpharmacy health professions programs and consider recommendations for targeting pharmacy technicians to pursue a PharmD degree. To accomplish this work, we created videos and proposed other innovative tools and flexible pathways to assist in recruiting students into the pharmacy profession. We also conducted a literature and website review, engaged in professional networking across the Academy, and proposed best practices to enhance student recruitment. In addition, we offered 8 recommendations to the American Association of Colleges of Pharmacy and 7 suggestions to colleges and schools of pharmacy to attract students to the pharmacy profession.

2.
Life Sci Alliance ; 7(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38373797

RESUMEN

Skeletal muscle development is a highly ordered process orchestrated transcriptionally by the myogenic regulatory factors. However, the downstream molecular mechanisms of myogenic regulatory factor functions in myogenesis are not fully understood. Here, we identified the RNA-binding protein Musashi2 (Msi2) as a myogenin target gene and a post-transcriptional regulator of myoblast differentiation. Msi2 knockdown in murine myoblasts blocked differentiation without affecting the expression of MyoD or myogenin. Msi2 overexpression was also sufficient to promote myoblast differentiation and myocyte fusion. Msi2 loss attenuated autophagosome formation via down-regulation of the autophagic protein MAPL1LC3/ATG8 (LC3) at the early phase of myoblast differentiation. Moreover, forced activation of autophagy effectively suppressed the differentiation defects incurred by Msi2 loss. Consistent with its functions in myoblasts in vitro, mice deficient for Msi2 exhibited smaller limb skeletal muscles, poorer exercise performance, and muscle fiber-type switching in vivo. Collectively, our study demonstrates that Msi2 is a novel regulator of mammalian myogenesis and establishes a new functional link between muscular development and autophagy regulation.


Asunto(s)
Desarrollo de Músculos , Músculo Esquelético , Animales , Ratones , Miogenina/genética , Miogenina/metabolismo , Músculo Esquelético/metabolismo , Desarrollo de Músculos/genética , Autofagia/genética , Proteínas de Unión al ARN/genética , Mamíferos/metabolismo
3.
Hum Mol Genet ; 31(5): 718-732, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-34581784

RESUMEN

In Duchenne muscular dystrophy (DMD), mutations in dystrophin result in a loss of the dystrophin-glycoprotein complex (DGC) at the myofiber membrane, which functions to connect the extracellular matrix with the intracellular actin cytoskeleton. The dystroglycan subcomplex interacts with dystrophin and spans the sarcolemma where its extensive carbohydrates (matriglycan and CT2 glycan) directly interact with the extracellular matrix. In the current manuscript, we show that sarcospan overexpression enhances the laminin-binding capacity of dystroglycan in DMD muscle by increasing matriglycan glycosylation of α-dystroglycan. Furthermore, we find that this modification is not affected by loss of Galgt2, a glycotransferase, which catalyzes the CT2 glycan. Our findings reveal that the matriglycan carbohydrates, and not the CT2 glycan, are necessary for sarcospan-mediated amelioration of DMD. Overexpression of Galgt2 in the DMD mdx murine model prevents muscle pathology by increasing CT2 modified α-dystroglycan. Galgt2 also increases expression of utrophin, which compensates for the loss of dystrophin in DMD muscle. We found that combined loss of Galgt2 and dystrophin reduced utrophin expression; however, it did not interfere with sarcospan rescue of disease. These data reveal a partial dependence of sarcospan on Galgt2 for utrophin upregulation. In addition, sarcospan alters the cross-talk between the adhesion complexes by decreasing the association of integrin ß1D with dystroglycan complexes. In conclusion, sarcospan functions to re-wire the cell to matrix connections by strengthening the cellular adhesion and signaling, which, in turn, increases the resilience of the myofiber membrane.


Asunto(s)
Distrofina , Distrofia Muscular de Duchenne , Animales , Carbohidratos , Distroglicanos/genética , Distroglicanos/metabolismo , Distrofina/genética , Distrofina/metabolismo , Laminina/genética , Laminina/metabolismo , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Utrofina/genética , Utrofina/metabolismo
4.
Exp Physiol ; 105(10): 1767-1777, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32833332

RESUMEN

NEW FINDINGS: What is the central question of this study? Does fukutin deficiency in skeletal muscle cause mitochondrial dysfunction, and if so, can AMP-activated protein kinase (AMPK) stimulation via 5-aminoimidazole-4-carboxamide ribonucleotide attenuate this through regulation of mitochondrial biogenesis and autophagy? What is the main finding and its importance? Mitochondrial dysfunction is associated with fukutin deficiency and AMPK stimulation may benefit muscle contractility to a greater extent than mitochondrial function. ABSTRACT: Disruptions in the dystrophin-glycoprotein complex (DGC) are clearly the primary basis underlying various forms of muscular dystrophies and dystroglycanopathies, but the cellular consequences of DGC disruption are still being investigated. Mitochondrial abnormalities are becoming an apparent consequence and contributor to dystrophy disease pathology. Herein, we demonstrate that muscle-specific deletion of the fukutin gene (Myf5/fktn-KO mice (Fktn KO)), a model of secondary dystroglycanopathy, results in ∼30% lower muscle strength (P < 0.001) and 16% lower mitochondrial respiratory function (P = 0.002) compared to healthy littermate controls (LM). We also observed ∼80% lower expression of the gene for peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) (P = 0.004), a primary transcription factor for mitochondrial biogenesis, in Fktn KO mice that likely contributes to the mitochondrial defects. PGC-1α is post-translationally regulated via phosphorylation by AMP-activated protein kinase (AMPK). Treatment with the AMPK agonist 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) failed to rescue mitochondrial deficits in Fktn KO mice (P = 0.458) but did have beneficial (∼30% greater) effects on recovery of muscle contractility following injury in both LM and Fktn KO mice compared to saline treatment (P = 0.006). The beneficial effects of AMPK stimulation via AICAR on muscle contractile function may be partially explained by AMPK's other role of regulating skeletal muscle autophagy, a cellular process critical for clearance of damaged and/or dysfunctional organelles. Two primary conclusions can be drawn from this data: (1) fukutin deletion produces intrinsic muscular metabolic defects that likely contribute to dystroglycanopathy disease pathology, and (2) AICAR treatment accelerates recovery of muscle contractile function following injury suggesting AMPK signalling as a possible target for therapeutic strategies.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Condicionamiento Físico Animal/fisiología , Ribonucleótidos/farmacología , Transferasas/deficiencia , Proteínas Quinasas Activadas por AMP/metabolismo , Aminoimidazol Carboxamida/farmacología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Enfermedades Mitocondriales/fisiopatología , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Fuerza Muscular/efectos de los fármacos , Fuerza Muscular/fisiología , Músculo Esquelético/fisiopatología , Distrofias Musculares/metabolismo , Distrofias Musculares/fisiopatología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
5.
Nat Commun ; 10(1): 2943, 2019 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-31270333

RESUMEN

Mutations exclusively in equilibrative nucleoside transporter 3 (ENT3), the only intracellular nucleoside transporter within the solute carrier 29 (SLC29) gene family, cause an expanding spectrum of human genetic disorders (e.g., H syndrome, PHID syndrome, and SHML/RDD syndrome). Here, we identify adult stem cell deficits that drive ENT3-related abnormalities in mice. ENT3 deficiency alters hematopoietic and mesenchymal stem cell fates; the former leads to stem cell exhaustion, and the latter leads to breaches of mesodermal tissue integrity. The molecular pathogenesis stems from the loss of lysosomal adenosine transport, which impedes autophagy-regulated stem cell differentiation programs via misregulation of the AMPK-mTOR-ULK axis. Furthermore, mass spectrometry-based metabolomics and bioenergetics studies identify defects in fatty acid utilization, and alterations in mitochondrial bioenergetics can additionally propel stem cell deficits. Genetic, pharmacologic and stem cell interventions ameliorate ENT3-disease pathologies and extend the lifespan of ENT3-deficient mice. These findings delineate a primary pathogenic basis for the development of ENT3 spectrum disorders and offer critical mechanistic insights into treating human ENT3-related disorders.


Asunto(s)
Células Madre Adultas/metabolismo , Proteínas de Transporte de Nucleósidos/metabolismo , Adenosina/metabolismo , Adenilato Quinasa/metabolismo , Células Madre Adultas/ultraestructura , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Autofagia , Transporte Biológico , Diferenciación Celular , Autorrenovación de las Células , Metabolismo Energético , Ácidos Grasos/metabolismo , Células HEK293 , Humanos , Metabolismo de los Lípidos , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Fenotipo , Ribonucleótidos/farmacología , Transducción de Señal , Análisis de Supervivencia , Serina-Treonina Quinasas TOR/metabolismo
6.
JCI Insight ; 52019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31039133

RESUMEN

In the current preclinical study, we demonstrate the therapeutic potential of sarcospan (SSPN) overexpression to alleviate cardiomyopathy associated with Duchenne muscular dystrophy (DMD) utilizing dystrophin-deficient mdx mice with utrophin haploinsufficiency that more accurately represent the severe disease course of human DMD. SSPN interacts with dystrophin, the DMD disease gene product, and its autosomal paralog utrophin, which is upregulated in DMD as a partial compensatory mechanism. SSPN transgenic mice have enhanced abundance of fully glycosylated α-dystroglycan, which may further protect dystrophin-deficient cardiac membranes. Baseline echocardiography reveals SSPN improves systolic function and hypertrophic indices in mdx and mdx:utr-heterozygous mice. Assessment of SSPN transgenic mdx mice by hemodynamic pressure-volume methods highlights enhanced systolic performance compared to mdx controls. SSPN restores cardiac sarcolemma stability, the primary defect in DMD disease, reduces fibrotic response and improves contractile function. We demonstrate that SSPN ameliorates more advanced cardiac disease in the context of diminished sarcolemma expression of utrophin and ß1D integrin that mitigate disease severity and partially restores responsiveness to ß-adrenergic stimulation. Overall, our current and previous findings suggest SSPN overexpression in DMD mouse models positively impacts skeletal, pulmonary and cardiac performance by addressing the stability of proteins at the sarcolemma that protect the heart from injury, supporting SSPN and membrane stabilization as a therapeutic target for DMD.


Asunto(s)
Cardiomiopatías/terapia , Terapia Genética/métodos , Proteínas de la Membrana/genética , Distrofia Muscular de Duchenne/complicaciones , Proteínas de Neoplasias/genética , Sarcolema/patología , Animales , Cardiomiopatías/diagnóstico , Cardiomiopatías/etiología , Modelos Animales de Enfermedad , Distrofina/genética , Ecocardiografía , Femenino , Humanos , Integrina beta1 , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Contracción Muscular/genética , Músculo Esquelético/citología , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Miocardio/citología , Miocardio/patología , Proteínas de Neoplasias/metabolismo , Estabilidad Proteica , Utrofina/metabolismo
7.
J Tissue Eng Regen Med ; 12(8): 1867-1876, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29774991

RESUMEN

Duchenne muscular dystrophy is a severe muscle wasting disease due to the absence of the dystrophin protein from the muscle cell membrane, which renders the muscle susceptible to continuous damage. In Duchenne muscular dystrophy patients, muscle weakness, together with cycles of degeneration/regeneration and replacement with noncontractile tissue, limit mobility and lifespan. Because the loss of dystrophin results in loss of polarity and a reduction in the number of self-renewing satellite cells, it is postulated that these patients could achieve an improved quality of life if delivered cells could restore satellite cell function. In this study, we used both an established myotoxic injury model in wild-type (WT) mice and mdx mice alone (spontaneous muscle damage). Single (SC) and aggregated (AGG) mesenchymal stem cells (MSCs) were injected into the gastrocnemius muscles 4 hr after injury (WT mice). The recovery of peak isometric torque was longitudinally assessed over 5 weeks, with earlier takedowns for histological assessment of healing (fibre cross-section area and central nucleation) and MSC retention. AGG-treated WT mice had significantly greater torque recovery at Day 14 than SC or saline-treated mice and a greater CSA at Day 10, compared with SC/saline. AGG-treated mdx mice had a greater peak isometric torque compared with SC/saline. In vitro immunomodulatory factor secretion of AGG-MSCs was higher than SC-MSCs for all tested growth factors with the largest difference observed in hepatocyte growth factor. Future studies are necessary to pair immunomodulatory factor secretion with functional attributes, to better predict the potential therapeutic value of MSC treatment modalities.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Músculo Esquelético/fisiología , Distrofia Muscular de Duchenne , Regeneración , Animales , Agregación Celular , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/terapia
8.
J Clin Invest ; 128(6): 2339-2355, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29533927

RESUMEN

The remarkable regeneration capability of skeletal muscle depends on the coordinated proliferation and differentiation of satellite cells (SCs). The self-renewal of SCs is critical for long-term maintenance of muscle regeneration potential. Hypoxia profoundly affects the proliferation, differentiation, and self-renewal of cultured myoblasts. However, the physiological relevance of hypoxia and hypoxia signaling in SCs in vivo remains largely unknown. Here, we demonstrate that SCs are in an intrinsic hypoxic state in vivo and express hypoxia-inducible factor 2A (HIF2A). HIF2A promotes the stemness and long-term homeostatic maintenance of SCs by maintaining their quiescence, increasing their self-renewal, and blocking their myogenic differentiation. HIF2A stabilization in SCs cultured under normoxia augments their engraftment potential in regenerative muscle. Conversely, HIF2A ablation leads to the depletion of SCs and their consequent regenerative failure in the long-term. In contrast, transient pharmacological inhibition of HIF2A accelerates muscle regeneration by increasing SC proliferation and differentiation. Mechanistically, HIF2A induces the quiescence and self-renewal of SCs by binding the promoter of the Spry1 gene and activating Spry1 expression. These findings suggest that HIF2A is a pivotal mediator of hypoxia signaling in SCs and may be therapeutically targeted to improve muscle regeneration.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proliferación Celular , Músculo Esquelético/fisiología , Regeneración , Células Satélite del Músculo Esquelético/metabolismo , Transducción de Señal , Animales , Diferenciación Celular , Hipoxia de la Célula , Ratones , Ratones Transgénicos , Desarrollo de Músculos , Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/citología
9.
Bioorg Med Chem ; 26(6): 1167-1173, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28911855

RESUMEN

Although EGFR is a highly sought-after drug target, inhibitor resistance remains a challenge. As an alternative strategy for kinase inhibition, we sought to explore whether allosteric activation mechanisms could effectively be disrupted. The kinase domain of EGFR forms an atypical asymmetric dimer via head-to-tail interactions and serves as a requisite for kinase activation. The kinase dimer interface is primarily formed by the H-helix derived from one kinase monomer and the small lobe of the second monomer. We hypothesized that a peptide designed to resemble the binding surface of the H-helix may serve as an effective disruptor of EGFR dimerization and activation. A library of constrained peptides was designed to mimic the H-helix of the kinase domain and interface side chains were optimized using molecular modeling. Peptides were constrained using peptide "stapling" to structurally reinforce an alpha-helical conformation. Peptide stapling was demonstrated to notably enhance cell permeation of an H-helix derived peptide termed EHBI2. Using cell-based assays, EHBI2 was further shown to significantly reduce EGFR activity as measured by EGFR phosphorylation and phosphorylation of the downstream signaling substrate Akt. To our knowledge, this is the first H-helix-based compound targeting the asymmetric interface of the kinase domain that can successfully inhibit EGFR activation and signaling. This study presents a novel, alternative targeting site for allosteric inhibition of EGFR.


Asunto(s)
Receptores ErbB/metabolismo , Péptidos/química , Inhibidores de Proteínas Quinasas/química , Regulación Alostérica , Línea Celular Tumoral , Dimerización , Receptores ErbB/química , Humanos , Microscopía Fluorescente , Péptidos/síntesis química , Péptidos/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/metabolismo , Estructura Secundaria de Proteína
10.
Hum Mol Genet ; 26(10): 1952-1965, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28334834

RESUMEN

Limb Girdle Muscular Dystrophies type 2I (LGMD2I), a recessive autosomal muscular dystrophy, is caused by mutations in the Fukutin Related Protein (FKRP) gene. It has been proposed that FKRP, a ribitol-5-phosphate transferase, is a participant in α-dystroglycan (αDG) glycosylation, which is important to ensure the cell/matrix anchor of muscle fibers. A LGMD2I knock-in mouse model was generated to express the most frequent mutation (L276I) encountered in patients. The expression of FKRP was not altered neither at transcriptional nor at translational levels, but its function was impacted since abnormal glycosylation of αDG was observed. Skeletal muscles were functionally impaired from 2 months of age and a moderate dystrophic pattern was evident starting from 6 months of age. Gene transfer with a rAAV2/9 vector expressing Fkrp restored biochemical defects, corrected the histological abnormalities and improved the resistance to eccentric stress in the mouse model. However, injection of high doses of the vector induced a decrease of αDG glycosylation and laminin binding, even in WT animals. Finally, intravenous injection of the rAAV-Fkrp vector into a dystroglycanopathy mouse model due to Fukutin (Fktn) knock-out indicated a dose-dependent toxicity. These data suggest requirement for a control of FKRP expression in muscles.


Asunto(s)
Distrofia Muscular de Cinturas/terapia , Proteínas/genética , Proteínas/uso terapéutico , Animales , Modelos Animales de Enfermedad , Distroglicanos/metabolismo , Expresión Génica , Regulación de la Expresión Génica/genética , Terapia Genética/métodos , Glicosilación , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofia Muscular de Cinturas/genética , Mutación , Pentosafosfatos/metabolismo , Pentosiltransferasa , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteínas/metabolismo , Transferasas
11.
Glycobiology ; 26(12): 1284-1296, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27496765

RESUMEN

Both LARGE1 (formerly LARGE) and its paralog LARGE2 are bifunctional glycosyltransferases with xylosy- and glucuronyltransferase activities, and are capable of synthesizing polymers composed of a repeating disaccharide [-3Xylα1,3GlcAß1-]. Post-translational modification of the O-mannosyl glycan of α-dystroglycan (α-DG) with the polysaccharide is essential for it to act as a receptor for ligands in the extracellular matrix (ECM), and both LARGE paralogs contribute to the modification in vivo. LARGE1 and LARGE2 have different tissue distribution profiles and enzymatic properties; however, the functional difference of the homologs remains to be determined, and α-DG is the only known substrate for the modification by LARGE1 or LARGE2. Here we show that LARGE2 can modify proteoglycans (PGs) with the laminin-binding glycan. We found that overexpression of LARGE2, but not LARGE1, mediates the functional modification on the surface of DG-/-, Pomt1-/- and Fktn-/- embryonic stem cells. We identified a heparan sulfate-PG glypican-4 as a substrate for the LARGE2-dependent modification by affinity purification and subsequent mass spectrometric analysis. Furthermore, we showed that LARGE2 could modify several additional PGs with the laminin-binding glycan, most likely within the glycosaminoglycan (GAG)-protein linkage region. Our results indicate that LARGE2 can modify PGs with the GAG-like polysaccharide composed of xylose and glucuronic acid to confer laminin binding. Thus, LARGE2 may play a differential role in stabilizing the basement membrane and modifying its functions by augmenting the interactions between laminin globular domain-containing ECM proteins and PGs.


Asunto(s)
Glicosiltransferasas/metabolismo , Laminina/metabolismo , Proteoglicanos/metabolismo , Animales , Sitios de Unión , Células Madre Embrionarias/metabolismo , Glicosilación , Glicosiltransferasas/química , Laminina/química , Ratones , Ratones Noqueados , Proteoglicanos/química
12.
Artículo en Inglés | MEDLINE | ID: mdl-27430020

RESUMEN

Myosin heavy chain isoforms are an important component defining fiber type specific properties in skeletal muscle, such as oxidative versus glycolytic metabolism, rate of contraction, and fatigability. While the molecular mechanisms that underlie specification of the different fiber types are becoming clearer, how this programming becomes disrupted in muscular dystrophy and the functional consequences of fiber type changes in disease are not fully resolved. Fiber type changes in disease, with specific focus on muscular dystrophies caused by defects in the dystrophin glycoprotein complex, are discussed.

13.
Skelet Muscle ; 6: 20, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27257474

RESUMEN

BACKGROUND: Secondary dystroglycanopathies are a subset of muscular dystrophy caused by abnormal glycosylation of α-dystroglycan (αDG). Loss of αDG functional glycosylation prevents it from binding to laminin and other extracellular matrix receptors, causing muscular dystrophy. Mutations in a number of genes, including FKTN (fukutin), disrupt αDG glycosylation. METHODS: We analyzed conditional Fktn knockout (Fktn KO) muscle for levels of mTOR signaling pathway proteins by Western blot. Two cohorts of Myf5-cre/Fktn KO mice were treated with the mammalian target of rapamycin (mTOR) inhibitor rapamycin (RAPA) for 4 weeks and evaluated for changes in functional and histopathological features. RESULTS: Muscle from 17- to 25-week-old fukutin-deficient mice has activated mTOR signaling. However, in tamoxifen-inducible Fktn KO mice, factors related to Akt/mTOR signaling were unchanged before the onset of dystrophic pathology, suggesting that Akt/mTOR signaling pathway abnormalities occur after the onset of disease pathology and are not causative in early dystroglycanopathy development. To determine any pharmacological benefit of targeting mTOR signaling, we administered RAPA daily for 4 weeks to Myf5/Fktn KO mice to inhibit mTORC1. RAPA treatment reduced fibrosis, inflammation, activity-induced damage, and central nucleation, and increased muscle fiber size in Myf5/Fktn KO mice compared to controls. RAPA-treated KO mice also produced significantly higher torque at the conclusion of dosing. CONCLUSIONS: These findings validate a misregulation of mTOR signaling in dystrophic dystroglycanopathy skeletal muscle and suggest that such signaling molecules may be relevant targets to delay and/or reduce disease burden in dystrophic patients.


Asunto(s)
Distroglicanos/metabolismo , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular Animal/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Fenómenos Biomecánicos , Modelos Animales de Enfermedad , Regulación hacia Abajo , Estimulación Eléctrica , Femenino , Predisposición Genética a la Enfermedad , Glicosilación , Masculino , Ratones Noqueados , Contracción Muscular , Fuerza Muscular , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Distrofia Muscular Animal/fisiopatología , Factor 5 Regulador Miogénico/deficiencia , Factor 5 Regulador Miogénico/genética , Fenotipo , Procesamiento Proteico-Postraduccional , Proteínas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo , Torque , Transferasas
14.
Am J Physiol Cell Physiol ; 311(2): C190-200, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27281480

RESUMEN

The primary objective of this study was to determine whether alterations in mitochondria affect recovery of skeletal muscle strength and mitochondrial enzyme activity following myotoxic injury. 3-Methyladenine (3-MA) was administered daily (15 mg/kg) to blunt autophagy, and the creatine analog guanidionpropionic acid (ß-GPA) was administered daily (1% in chow) to enhance oxidative capacity. Male C57BL/6 mice were randomly assigned to nontreatment (Con, n = 6), 3-MA-treated (n = 6), and ß-GPA-treated (n = 8) groups for 10 wk. Mice were euthanized at 14 days after myotoxic injury for assessment of mitochondrial remodeling during regeneration and its association with the recovery of muscle strength. Expression of several autophagy-related proteins, e.g., phosphorylated Ulk1 (∼2- to 4-fold, P < 0.049) was greater in injured than uninjured muscles, indicating a relationship between muscle regeneration/remodeling and autophagy. By 14 days postinjury, recovery of muscle strength (18% less, P = 0.03) and mitochondrial enzyme (e.g., citrate synthase) activity (22% less, P = 0.049) were significantly lower in 3-MA-treated than Con mice, suggesting that the autophagy process plays an important role during muscle regeneration. In contrast, muscle regeneration was nearly complete in ß-GPA-treated mice, i.e., muscle strength recovered to 93% of baseline vs. 78% for Con mice. Remarkably, 14 days allowed sufficient time for a near-complete recovery of mitochondrial function in ß-GPA-treated mice (e.g., no difference in citrate synthase activity between injured and uninjured, P = 0.49), indicating a robust mitochondrial remodeling process during muscle regeneration. In conclusion, autophagy is likely activated following muscle injury and appears to play an important role in functional muscle regeneration.


Asunto(s)
Autofagia/fisiología , Mitocondrias Musculares/fisiología , Músculo Esquelético/fisiología , Recuperación de la Función/fisiología , Regeneración/fisiología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Autofagia/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Musculares/efectos de los fármacos , Fuerza Muscular/efectos de los fármacos , Fuerza Muscular/fisiología , Músculo Esquelético/efectos de los fármacos , Enfermedades Musculares/tratamiento farmacológico , Enfermedades Musculares/fisiopatología , Recuperación de la Función/efectos de los fármacos , Regeneración/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
15.
PLoS One ; 11(1): e0147049, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26751696

RESUMEN

Glycosylated α-dystroglycan provides an essential link between extracellular matrix proteins, like laminin, and the cellular cytoskeleton via the dystrophin-glycoprotein complex. In secondary dystroglycanopathy muscular dystrophy, glycosylation abnormalities disrupt a complex O-mannose glycan necessary for muscle structural integrity and signaling. Fktn-deficient dystroglycanopathy mice develop moderate to severe muscular dystrophy with skeletal muscle developmental and/or regeneration defects. To gain insight into the role of glycosylated α-dystroglycan in these processes, we performed muscle fiber typing in young (2, 4 and 8 week old) and regenerated muscle. In mice with Fktn disruption during skeletal muscle specification (Myf5/Fktn KO), newly regenerated fibers (embryonic myosin heavy chain positive) peaked at 4 weeks old, while total regenerated fibers (centrally nucleated) were highest at 8 weeks old in tibialis anterior (TA) and iliopsoas, indicating peak degeneration/regeneration activity around 4 weeks of age. In contrast, mature fiber type specification at 2, 4 and 8 weeks old was relatively unchanged. Fourteen days after necrotic toxin-induced injury, there was a divergence in muscle fiber types between Myf5/Fktn KO (skeletal-muscle specific) and whole animal knockout induced with tamoxifen post-development (Tam/Fktn KO) despite equivalent time after gene deletion. Notably, Tam/Fktn KO retained higher levels of embryonic myosin heavy chain expression after injury, suggesting a delay or abnormality in differentiation programs. In mature fiber type specification post-injury, there were significant interactions between genotype and toxin parameters for type 1, 2a, and 2x fibers, and a difference between Myf5/Fktn and Tam/Fktn study groups in type 2b fibers. These data suggest that functionally glycosylated α-dystroglycan has a unique role in muscle regeneration and may influence fiber type specification post-injury.


Asunto(s)
Distroglicanos/genética , Fibras Musculares Esqueléticas/fisiología , Distrofia Muscular Animal/genética , Proteínas/genética , Regeneración , Animales , Cardiotoxinas/química , Modelos Animales de Enfermedad , Distroglicanos/metabolismo , Exones , Eliminación de Gen , Genotipo , Glicosilación , Laminina/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/fisiología , Distrofia Muscular Animal/metabolismo , Necrosis , Transducción de Señal , Transferasas
16.
J Vis Exp ; (118)2016 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-28060301

RESUMEN

With this method, consecutive cryosections are collected to enable both microscopy applications for tissue histology and enrichment of RNA for gene expression using adjacent regions from a single mouse skeletal muscle. Typically, it is challenging to achieve adequate homogenization of small skeletal muscle samples because buffer volumes may be too low for efficient grinding applications, yet without sufficient mechanical disruption, the dense tissue architecture of muscle limits penetration of buffer reagents, ultimately causing low RNA yield. By following the protocol reported here, 30 µm sections are collected and pooled allowing cryosectioning and subsequent needle homogenization to mechanically disrupt the muscle, increasing the surface area exposed for buffer penetration. The primary limitations of the technique are that it requires a cryostat, and it is relatively low throughput. However, high-quality RNA can be obtained from small samples of pooled muscle cryosections, making this method accessible for many different skeletal muscles and other tissues. Furthermore, this technique enables matched analyses (e.g., tissue histopathology and gene expression) from adjacent regions of a single skeletal muscle so that measurements can be directly compared across applications to reduce experimental uncertainty and to reduce replicative animal experiments necessary to source a small tissue for multiple applications.


Asunto(s)
Crioultramicrotomía , Músculo Esquelético/patología , Animales , Ratones , ARN
17.
PLoS One ; 10(9): e0139489, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26418018

RESUMEN

Although microRNAs (miRNAs) are small, non-protein-coding entities, they have important roles in post-transcriptional regulation of most of the human genome. These small entities generate fine-tuning adjustments in the expression of mRNA, which can mildly or massively affect the abundance of proteins. Previously, we found that the expression of miR-30c-2-3p is induced by lysophosphatidic acid and has an important role in the regulation of cell proliferation in ovarian cancer cells. The goal here is to confirm that ATF3 mRNA is a target of miR-30c-2-3p silencing, thereby further establishing the functional role of miR-30c-2-3p. Using a combination of bioinformatics, qRT-PCR, immunoblotting and luciferase assays, we uncovered a regulatory pathway between miR-30c-2-3p and the expression of the transcription factor, ATF3. Lysophosphatidic acids triggers the expression of both miR-30c-2-3p and ATF3, which peak at 1 h and are absent 8 h post stimulation in SKOV-3 and OVCAR-3 serous ovarian cancer cells. The 3´-untranslated region (3´-UTR) of ATF3 was a predicted, putative target for miR-30c-2-3p, which we confirmed as a bona-fide interaction using a luciferase reporter assay. Specific mutations introduced into the predicted site of interaction between miR-30c-2-3p and the 3´-UTR of ATF3 alleviated the suppression of the luciferase signal. Furthermore, the presence of anti-miR-30c-2-3p enhanced ATF3 mRNA and protein after lysophosphatidic acid stimulation. Thus, the data suggest that after the expression of ATF3 and miR-30c-2-3p are elicited by lysophosphatidic acid, subsequently miR-30c-2-3p negatively regulates the expression of ATF3 through post-transcriptional silencing, which prevents further ATF3-related outcomes as a consequence of lysophosphatidic acid signaling.


Asunto(s)
Factor de Transcripción Activador 3/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Lisofosfolípidos/farmacología , MicroARNs/genética , Transcripción Genética/efectos de los fármacos , Regiones no Traducidas 3'/genética , Factor de Transcripción Activador 3/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Immunoblotting , Microscopía Fluorescente , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Cancer Lett ; 369(1): 175-83, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26319900

RESUMEN

The regulator of G protein signaling 10 (RGS10) protein is a GTPase activating protein that accelerates the hydrolysis of GTP and therefore canonically inactivates G proteins, ultimately terminating signaling. Rheb is a small GTPase protein that shuttles between its GDP- and GTP-bound forms to activate mTOR. Since RGS10 suppression augments ovarian cancer cell viability, we sought to elucidate the molecular mechanism. Following RGS10 suppression in serum-free conditions, phosphorylation of mTOR, the eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1), p70S6K and S6 Ribosomal Protein appear. Furthermore, suppressing RGS10 increases activated Rheb, suggesting RGS10 antagonizes mTOR signaling via the small G-protein. The effects of RGS10 suppression are enhanced after stimulating cells with the growth factor, lysophosphatidic acid, and reduced with mTOR inhibitors, temsirolimus and INK-128. Suppression of RGS10 leads to an increase in cell proliferation, even in the presence of etoposide. In summary, the RGS10 suppression increases Rheb-GTP and mTOR signaling in ovarian cancer cells. Our results suggest that RGS10 could serve in a novel, and previously unknown, role by accelerating the hydrolysis of GTP from Rheb in ovarian cancer cells.


Asunto(s)
Proteínas de Unión al GTP Monoméricas/metabolismo , Neuropéptidos/metabolismo , Neoplasias Ováricas/metabolismo , Proteínas RGS/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Femenino , Humanos , Neoplasias Ováricas/patología , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas RGS/genética , Proteína Homóloga de Ras Enriquecida en el Cerebro , Transducción de Señal
19.
Bioorg Med Chem ; 23(12): 2761-6, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25840798

RESUMEN

The epidermal growth factor receptor (EGFR) dimerization arm is a key feature that stabilizes dimerization of the extracellular receptor, thereby mediating activation of the tyrosine kinase domain. Peptides mimicking this ß-loop feature can disrupt dimer formation and kinase activation, yet these peptides lack structural constraints or contain redox sensitive disulfide bonds which may limit their stability in physiological environments. Selenylsulfide bonds are a promising alternative to disulfide bonds as they maintain much of the same structural and chemical behavior, yet they are inherently less prone to reduction. Herein, we describe the synthesis, stability and activity of selenylsulfide-bridged dimerization arm mimics. The synthesis was accomplished using an Fmoc-based strategy along with C-terminal labeling for improved overall yield. This selenylsulfide-bridged peptide displayed both proteolytic stability and structural stability even under reducing conditions, demonstrating the potential application of the selenylsulfide bond to generate redox stable ß-loop peptides for disruption of protein-protein interactions.


Asunto(s)
Receptores ErbB/metabolismo , Péptidos/química , Peptidomiméticos/química , Multimerización de Proteína/efectos de los fármacos , Selenio/química , Sulfuros/química , Secuencia de Aminoácidos , Animales , Línea Celular , Diseño de Fármacos , Receptores ErbB/química , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/síntesis química , Péptidos/farmacología , Peptidomiméticos/síntesis química , Peptidomiméticos/farmacología , Conformación Proteica/efectos de los fármacos , Mapas de Interacción de Proteínas/efectos de los fármacos , Estabilidad Proteica , Selenio/farmacología , Sulfuros/síntesis química , Sulfuros/farmacología
20.
PLoS One ; 10(3): e0118796, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25790232

RESUMEN

The epidermal growth factor receptor (EGFR) is overexpressed in multiple carcinomas and is the focus of a variety of targeted therapies. Here we report the design of peptide-based compounds that mimic the EGFR dimerization arm and inhibit allosteric activation of EGFR. These peptides are modified to contain a triazolyl bridge between the peptide strands to constrain the EGFR dimerization arm ß-loop. In this study, we demonstrate that these peptides have significantly improved proteolytic stability over the non-modified peptide sequence, and their inhibitory effects are dependent on the number of the methylene units and orientation of the introduced triazolyl bridge. We identified a peptide, EDA2, which downregulates receptor phosphorylation and dimerization and reduces cell viability. This is the first example of a biologically active triazolyl-bridged peptide targeting the EGFR dimerization interface that effectively downregulates EGFR activation.


Asunto(s)
Regulación Alostérica/fisiología , Carcinoma/metabolismo , Ectodisplasinas/biosíntesis , Receptores ErbB/química , Modelos Moleculares , Ingeniería de Proteínas/métodos , Dimerización , Ectodisplasinas/metabolismo , Humanos , Simulación de Dinámica Molecular , Triazoles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...