Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Hum Mol Genet ; 31(4): 499-509, 2022 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-34505136

RESUMEN

Limb-girdle muscular dystrophy R3 (LGMDR3) is caused by mutations in the SGCA gene coding for α-sarcoglycan (SG). Together with ß- γ- and δ-SG, α-SG forms a tetramer embedded in the dystrophin associated protein complex crucial for protecting the sarcolemma from mechanical stresses elicited by muscle contraction. Most LGMDR3 cases are due to missense mutations, which result in non-properly folded, even though potentially functional α-SG. These mutants are prematurely discarded by the cell quality control. Lacking one subunit, the SG-complex is disrupted. The resulting loss of function leads to sarcolemma instability, muscle fiber damage and progressive limb muscle weakness. LGMDR3 is severely disabling and, unfortunately, still incurable. Here, we propose the use of small molecules, belonging to the class of cystic fibrosis transmembrane regulator (CFTR) correctors, for recovering mutants of α-SG defective in folding and trafficking. Specifically, CFTR corrector C17 successfully rerouted the SG-complex containing the human R98H-α-SG to the sarcolemma of hind-limb muscles of a novel LGMDR3 murine model. Notably, the muscle force of the treated model animals was fully recovered. To our knowledge, this is the first time that a compound designated for cystic fibrosis is successfully tested in a muscular dystrophy and may represent a novel paradigm of treatment for LGMDR3 as well as different other indications in which a potentially functional protein is prematurely discarded as folding-defective. Furthermore, the use of small molecules for recovering the endogenous mutated SG has an evident advantage over complex procedures such as gene or cell transfer.


Asunto(s)
Fibrosis Quística , Distrofia Muscular de Cinturas , Distrofias Musculares , Animales , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Ratones , Músculo Esquelético/metabolismo , Distrofias Musculares/metabolismo , Distrofia Muscular de Cinturas/genética , Sarcoglicanos/genética , Sarcoglicanos/metabolismo
2.
Purinergic Signal ; 20(1): 83-89, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37074620

RESUMEN

ATP is a ubiquitous extracellular messenger released in a wide number of pathophysiological conditions. ATP is known to be present in minute amounts in the extracellular space in healthy tissues and in the blood, and to modulate a multiplicity of cell responses. Cell culture systems are widely used to explore purinergic signaling. We show here that currently used fetal bovine sera contain ATP in the 300-1300 pmol/L range. Serum ATP is associated with albumin as well as with microparticle/microvesicle fraction. Serum microparticles/microvesicles affect in vitro cell responses due to their content of miRNAs, growth factors, and other bioactive molecules. ATP is likely to be one of these bioactive factors found in a variable amount in sera of different commercial sources. ATP in serum supports ATP-dependent biochemical reactions such as the hexokinase-dependent phosphorylation of glucose to glucose 6-phosphate, and affects purinergic signaling. These findings show that cells growing in vitro in serum-supplemented media are exposed to varying levels of extracellular ATP, and thus to varying degrees of purinergic stimulation.


Asunto(s)
Espacio Extracelular , Albúmina Sérica Bovina , Células Cultivadas , Espacio Extracelular/metabolismo , Adenosina Trifosfato/metabolismo , Glucosa
3.
FASEB J ; 36(3): e22140, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35107852

RESUMEN

Few studies explored the role of microRNAs (miRNAs) in the post-transcriptional regulation of glycolytic proteins and downstream effectors in ovarian cancer cells. We recently showed that the functional activation of the cytoskeletal regulator FAK in endothelial cells is fostered by the glycolytic enhancer 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3). We tested the hypothesis that miR-206 and mir-26b, emerging onco-suppressors targeting PFKFB3 in estrogen-dependent tumors, would regulate proliferation and migration of serous epithelial ovarian cancer (EOC) cells via common glycolytic proteins, i.e., GLUT1 and PFKFB3, and downstream FAK. PFKFB3 was overexpressed in SKOV3, and its pharmacological inhibition with 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) significantly reduced cell proliferation and motility. Both miR-206 and miR-26b directly targeted PFKFB3 as evaluated by a luciferase reporter assay. However, endogenous levels of miR-26b were higher than those of miR-206, which was barely detectable in SKOV3 as well as OVCAR5 and CAOV3 cells. Accordingly, only the anti-miR-26b inhibitor concentration-dependently increased PFKFB3 levels. While miR-206 overexpression impaired proliferation and migration by downregulating PFKFB3 levels, the decreased PFKFB3 protein levels related to miR-26 overexpression had no functional consequences in all EOC cell lines. Finally, consistent with the migration outcome, exogenous miR-206 and miR-26b induced opposite effects on the levels of total FAK and of its phosphorylated form at Tyr576/577. 3PO did not prevent miR-26b-induced SKOV3 migration. Overall, these results support the inverse relation between endogenous miRNA levels and their tumor-suppressive effects and suggest that restoring miR-206 expression represents a potential dual anti-PFKFB3/FAK strategy to control ovarian cancer progression.


Asunto(s)
Movimiento Celular/genética , Proliferación Celular/genética , Regulación hacia Abajo/genética , Quinasa 1 de Adhesión Focal/genética , MicroARNs/genética , Neoplasias Ováricas/genética , Fosfofructoquinasa-2/genética , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/patología , Línea Celular , Línea Celular Tumoral , Femenino , Regulación de la Expresión Génica/genética , Glucólisis/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neoplasias Ováricas/patología
4.
Int J Mol Sci ; 24(16)2023 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-37628888

RESUMEN

Sarcoglycanopathies, also known as limb girdle muscular dystrophy 3-6, are rare muscular dystrophies characterized, although heterogeneous, by high disability, with patients often wheelchair-bound by late adolescence and frequently developing respiratory and cardiac problems. These diseases are currently incurable, emphasizing the importance of effective treatment strategies and the necessity of animal models for drug screening and therapeutic verification. Using the CRISPR/Cas9 genome editing technique, we generated and characterized δ-sarcoglycan and ß-sarcoglycan knockout zebrafish lines, which presented a progressive disease phenotype that worsened from a mild larval stage to distinct myopathic features in adulthood. By subjecting the knockout larvae to a viscous swimming medium, we were able to anticipate disease onset. The δ-SG knockout line was further exploited to demonstrate that a δ-SG missense mutant is a substrate for endoplasmic reticulum-associated degradation (ERAD), indicating premature degradation due to protein folding defects. In conclusion, our study underscores the utility of zebrafish in modeling sarcoglycanopathies through either gene knockout or future knock-in techniques. These novel zebrafish lines will not only enhance our understanding of the disease's pathogenic mechanisms, but will also serve as powerful tools for phenotype-based drug screening, ultimately contributing to the development of a cure for sarcoglycanopathies.


Asunto(s)
Distrofia Muscular de Cinturas , Sarcoglicanopatías , Animales , Degradación Asociada con el Retículo Endoplásmico , Pez Cebra/genética , Evaluación Preclínica de Medicamentos , Larva
5.
Int J Mol Sci ; 23(20)2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-36293223

RESUMEN

Congenital pseudomyotonia in cattle (PMT) is a rare skeletal muscle disorder, clinically characterized by stiffness and by delayed muscle relaxation after exercise. Muscle relaxation impairment is due to defective content of the Sarco(endo)plasmic Reticulum Ca2+ ATPase isoform 1 (SERCA1) protein, caused by missense mutations in the ATP2A1 gene. PMT represents the only mammalian model of human Brody myopathy. In the Romagnola breed, two missense variants occurring in the same allele were described, leading to Gly211Val and Gly286Val (G211V/G286V) substitutions. In this study, we analyzed the consequences of G211V and G286V mutations. Results support that the reduced amount of SERCA1 is a consequence of the G211V mutation, the G286V mutation almost being benign and the ubiquitin-proteasome system (UPS) being involved. After blocking the proteasome using a proteasome inhibitor, we found that the G211V mutant accumulates in cells at levels comparable to those of WT SERCA1. Our conclusion is that G211/286V mutations presumably originate in a folding-defective SERCA1 protein, recognized and diverted to degradation by UPS, although still catalytically functional, and that the main role is played by G211V mutation. Rescue of mutated SERCA1 to the sarcoplasmic reticulum membrane can re-establish resting cytosolic Ca2+ concentration and prevent the appearance of pathological signs, paving the way for a possible therapeutic approach against Brody disease.


Asunto(s)
Síndrome de Isaacs , Bovinos , Humanos , Animales , Síndrome de Isaacs/genética , Síndrome de Isaacs/veterinaria , Síndrome de Isaacs/patología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Complejo de la Endopetidasa Proteasomal/genética , Inhibidores de Proteasoma , Estrés del Retículo Endoplásmico , Retículo Sarcoplasmático/genética , Mutación , Ubiquitina/genética , Músculo Esquelético/patología , Mamíferos
6.
FASEB J ; 34(9): 12768-12784, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32757462

RESUMEN

Few studies have explored the mechanisms coupling estrogen signals to metabolic demand in endothelial cells. We recently showed that 17ß-estradiol (E2) triggers angiogenesis via the membrane G-protein coupled estrogen receptor (GPER) and the key glycolytic protein PFKFB3 as a downstream effector. We herein investigated whether estrogenic agents regulate the stability and/or degradation of glycolytic proteins in human umbilical vein endothelial cells (HUVECs). Similarly to E2, the GPER selective agonist G1 rapidly increased PFKFB3 protein amounts, without affecting mRNA levels. In the presence of cycloheximide, E2 and G1 treatment counteracted PFKFB3 degradation over time, whereas E2-induced PFKFB3 stabilization was abolished by the GPER antagonist G15. Inhibitors of selective SCF E3 ubiquitin ligase (SMER-3) and proteasome (MG132) rapidly increased PFKFB3 protein levels. Accordingly, ubiquitin-bound PFKFB3 was lower in E2- or G1-treated HUVECs. Both agents increased deubiquitinase USP19 levels through GPER signaling. Notably, USP 19 siRNA decreased PFKFB3 levels and abolished E2- and G1-mediated HUVEC tubularization. Finally, E2 and G1 treatments rapidly enhanced glucose transporter GLUT1 levels via GPER independent of transcriptional activation. These findings provide new evidence on mechanisms coupling estrogen signals with the glycolytic program in endothelium and unravel the role of USP19 as a target of the pro-angiogenic effect of estrogenic agents.


Asunto(s)
Endopeptidasas/metabolismo , Estradiol/farmacología , Transportador de Glucosa de Tipo 1/metabolismo , Fosfofructoquinasa-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos
7.
Hum Mol Genet ; 27(6): 969-984, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29351619

RESUMEN

Limb-girdle muscular dystrophy type 2D (LGMD2D) is a rare autosomal-recessive disease, affecting striated muscle, due to mutation of SGCA, the gene coding for α-sarcoglycan. Nowadays, more than 50 different SGCA missense mutations have been reported. They are supposed to impact folding and trafficking of α-sarcoglycan because the defective polypeptide, although potentially functional, is recognized and disposed of by the quality control of the cell. The secondary reduction of α-sarcoglycan partners, ß-, γ- and δ-sarcoglycan, disrupts a key membrane complex that, associated to dystrophin, contributes to assure sarcolemma stability during muscle contraction. The complex deficiency is responsible for muscle wasting and the development of a severe form of dystrophy. Here, we show that the application of small molecules developed to rescue ΔF508-CFTR trafficking, and known as CFTR correctors, also improved the maturation of several α-sarcoglycan mutants that were consequently rescued at the plasma membrane. Remarkably, in myotubes from a patient with LGMD2D, treatment with CFTR correctors induced the proper re-localization of the whole sarcoglycan complex, with a consequent reduction of sarcolemma fragility. Although the mechanism of action of CFTR correctors on defective α-sarcoglycan needs further investigation, this is the first report showing a quantitative and functional recovery of the sarcoglycan-complex in human pathologic samples, upon small molecule treatment. It represents the proof of principle of a pharmacological strategy that acts on the sarcoglycan maturation process and we believe it has a great potential to develop as a cure for most of the patients with LGMD2D.


Asunto(s)
Sarcoglicanopatías/tratamiento farmacológico , Sarcoglicanos/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células HEK293 , Humanos , Contracción Muscular , Músculo Esquelético/metabolismo , Músculo Estriado/metabolismo , Mutación Missense , Prueba de Estudio Conceptual , Sarcoglicanopatías/genética , Sarcoglicanopatías/metabolismo , Sarcoglicanos/genética
8.
Int J Mol Sci ; 21(5)2020 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-32155735

RESUMEN

Sarcoglycanopathies are rare limb girdle muscular dystrophies, still incurable, even though symptomatic treatments may slow down the disease progression. Most of the disease-causing defects are missense mutations leading to a folding defective protein, promptly removed by the cell's quality control, even if possibly functional. Recently, we repurposed small molecules screened for cystic fibrosis as potential therapeutics in sarcoglycanopathy. Indeed, cystic fibrosis transmembrane regulator (CFTR) correctors successfully recovered the defective sarcoglycan-complex in vitro. Our aim was to test the combined administration of some CFTR correctors with C17, the most effective on sarcoglycans identified so far, and evaluate the stability of the rescued sarcoglycan-complex. We treated differentiated myogenic cells from both sarcoglycanopathy and healthy donors, evaluating the global rescue and the sarcolemma localization of the mutated protein, by biotinylation assays and western blot analyses. We observed the additive/synergistic action of some compounds, gathering the first ideas on possible mechanism/s of action. Our data also suggest that a defective α-sarcoglycan is competent for assembly into the complex that, if helped in cell traffic, can successfully reach the sarcolemma. In conclusion, our results strengthen the idea that CFTR correctors, acting probably as proteostasis modulators, have the potential to progress as therapeutics for sarcoglycanopathies caused by missense mutations.


Asunto(s)
Aminopiridinas/farmacología , Benzodioxoles/farmacología , Fibras Musculares Esqueléticas/efectos de los fármacos , Mutación , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Sarcoglicanopatías/tratamiento farmacológico , Sarcoglicanos/metabolismo , Fibrosis Quística , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Combinación de Medicamentos , Células HEK293 , Humanos , Fibras Musculares Esqueléticas/metabolismo , Sarcoglicanopatías/genética , Sarcoglicanopatías/metabolismo , Sarcoglicanopatías/patología , Sarcoglicanos/genética
9.
Hum Mol Genet ; 23(14): 3746-58, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24565866

RESUMEN

Many membrane and secretory proteins that fail to pass quality control in the endoplasmic reticulum (ER) are dislocated into the cytosol and degraded by the proteasome. In applying rigid rules, however, quality control sometimes discharges proteins that, even though defective, retain their function. The unnecessary removal of such proteins represents the pathogenetic hallmark of diverse genetic diseases, in the case of ΔF508 mutant of cystic fibrosis transmembrane conductance regulator being probably the best known example. Recently, the inappropriate proteasomal degradation of skeletal muscle sarcoglycans (α, ß, γ and δ) with missense mutation has been proposed to be at the bases of mild-to-severe forms of limb girdle muscular dystrophy (LGMD) known as type 2D, 2E, 2C and 2F, respectively. The quality control pathway responsible for sarcoglycan mutant disposal, however, is so far unexplored. Here we reveal key components of the degradative route of V247M α-sarcoglycan mutant, the second most frequently reported mutation in LGMD-2D. The disclosure of the pathway, which is led by the E3 ligases HRD1 and RFP2, permits to identify new potential druggable targets of a disease for which no effective therapy is at present available. Notably, we show that the pharmacological inhibition of HRD1 activity rescues the expression of V247-α-sarcoglycan both in a heterologous cell model and in myotubes derived from a LGMD-2D patient carrying the L31P/V247M mutations. This represents the first evidence that the activity of E3 ligases, the enzymes in charge of mutant fate, can be eligible for drug interventions to treat sarcoglycanopathy.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Sarcoglicanopatías/metabolismo , Sarcoglicanos/genética , Sarcoglicanos/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células Cultivadas , Retículo Endoplásmico/metabolismo , Inhibidores Enzimáticos/farmacología , Células HEK293 , Humanos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Mutación Missense , Sarcoglicanopatías/genética , Sarcoglicanopatías/patología , Transducción de Señal/efectos de los fármacos , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitinación
10.
J Biol Chem ; 289(48): 33073-82, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25288803

RESUMEN

A missense mutation in ATP2A1 gene, encoding sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA1) protein, causes Chianina cattle congenital pseudomyotonia, an exercise-induced impairment of muscle relaxation. Skeletal muscles of affected cattle are characterized by a selective reduction of SERCA1 in sarcoplasmic reticulum membranes. In this study, we provide evidence that the ubiquitin proteasome system is involved in the reduced density of mutated SERCA1. The treatment with MG132, an inhibitor of ubiquitin proteasome system, rescues the expression level and membrane localization of the SERCA1 mutant in a heterologous cellular model. Cells co-transfected with the Ca(2+)-sensitive probe aequorin show that the rescued SERCA1 mutant exhibits the same ability of wild type to maintain Ca(2+) homeostasis within cells. These data have been confirmed by those obtained ex vivo on adult skeletal muscle fibers from a biopsy from a pseudomyotonia-affected subject. Our data show that the mutation generates a protein most likely corrupted in proper folding but not in catalytic activity. Rescue of mutated SERCA1 to sarcoplasmic reticulum membrane can re-establish resting cytosolic Ca(2+) concentration and prevent the appearance of pathological signs of cattle pseudomyotonia.


Asunto(s)
Enfermedades de los Bovinos/enzimología , Síndrome de Isaacs/enzimología , Síndrome de Isaacs/veterinaria , Proteínas Musculares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Retículo Sarcoplasmático/enzimología , Ubiquitina/metabolismo , Animales , Calcio/metabolismo , Bovinos , Enfermedades de los Bovinos/genética , Enfermedades de los Bovinos/patología , Cricetinae , Células HEK293 , Humanos , Síndrome de Isaacs/genética , Síndrome de Isaacs/patología , Leupeptinas/farmacología , Proteínas Musculares/genética , Mutación , Complejo de la Endopetidasa Proteasomal/genética , Inhibidores de Proteasoma/farmacología , Pliegue de Proteína/efectos de los fármacos , Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/patología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Ubiquitina/genética
11.
Curr Opin Pharmacol ; 76: 102459, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38713975

RESUMEN

Sarcoglycanopathies are rare autosomal recessive diseases belonging to the family of limb-girdle muscular dystrophies. They are caused by mutations in the genes coding for α-, ß-, γ-, and δ-sarcoglycan. The mutations impair the assembly of a key structural complex, which normally protects the sarcolemma of striated muscle from contraction-derived stress. Although heterogeneous, sarcoglycanopathies are characterized by progressive muscle degeneration, increased serum creatine kinase levels, loss of ambulation often during adolescence, and variable cardio-respiratory impairment. Genetic defects can impair sarcoglycan synthesis or produce a protein that is defective in folding. There is currently no effective treatment available; however, both gene replacement strategy and small molecule-based approaches show great promise and have entered or are starting to enter clinical trials.


Asunto(s)
Sarcoglicanopatías , Sarcoglicanos , Humanos , Sarcoglicanopatías/terapia , Sarcoglicanopatías/genética , Sarcoglicanopatías/metabolismo , Animales , Sarcoglicanos/genética , Sarcoglicanos/metabolismo , Terapia Genética/métodos , Mutación
12.
ACS Med Chem Lett ; 14(8): 1049-1053, 2023 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-37583821

RESUMEN

4'-Methyl-4,5'-bithiazoles were previously identified as cystic fibrosis transmembrane regulator (CFTR) correctors, thus being able to correct folding defective mutants of the channel regulating chloride transport through the membrane. Additionally, bithiazole derivative C17 was reported to recover α-sarcoglycan in vitro and in vivo. We report here the synthesis of two new derivatives of C17, in which the two sides of the bithiazole scaffold were modified. The synthesized compounds and the corresponding precursors were tested in myogenic cells to evaluate the expression of α-sarcoglycan. The results highlighted that both substitutions of the bithiazole scaffold are important to achieve the maximum recovery of the α-sarcoglycan mutant. Nonetheless, partial preservation of the activity was observed. Accordingly, this paves the way to further derivatizations/optimization and target fishing studies, which were preliminarily performed in this study as a proof of concept, allowing the investigation of the molecular mechanisms leading to the α-sarcoglycan correction.

13.
J Gen Physiol ; 154(11)2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-36149386

RESUMEN

Inositol 1,4,5-trisphosphate receptors (IP3Rs) are enriched at postsynaptic membrane compartments of the neuromuscular junction (NMJ), surrounding the subsynaptic nuclei and close to nicotinic acetylcholine receptors (nAChRs) of the motor endplate. At the endplate level, it has been proposed that nerve-dependent electrical activity might trigger IP3-associated, local Ca2+ signals not only involved in excitation-transcription (ET) coupling but also crucial to the development and stabilization of the NMJ itself. The present study was undertaken to examine whether denervation affects the subsynaptic IP3R distribution in skeletal muscles and which are the underlying mechanisms. Fluorescence microscopy, carried out on in vivo denervated muscles (following sciatectomy) and in vitro denervated skeletal muscle fibers from flexor digitorum brevis (FDB), indicates that denervation causes a reduction in the subsynaptic IP3R1-stained region, and such a decrease appears to be determined by the lack of muscle electrical activity, as judged by partial reversal upon field electrical stimulation of in vitro denervated skeletal muscle fibers.


Asunto(s)
Calcio , Receptores Nicotínicos , Calcio/metabolismo , Inositol , Receptores de Inositol 1,4,5-Trifosfato , Músculo Esquelético/metabolismo , Unión Neuromuscular
14.
NPJ Regen Med ; 7(1): 25, 2022 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-35468920

RESUMEN

The production of skeletal muscle constructs useful for replacing large defects in vivo, such as in congenital diaphragmatic hernia (CDH), is still considered a challenge. The standard application of prosthetic material presents major limitations, such as hernia recurrences in a remarkable number of CDH patients. With this work, we developed a tissue engineering approach based on decellularized diaphragmatic muscle and human cells for the in vitro generation of diaphragmatic-like tissues as a proof-of-concept of a new option for the surgical treatment of large diaphragm defects. A customized bioreactor for diaphragmatic muscle was designed to control mechanical stimulation and promote radial stretching during the construct engineering. In vitro tests demonstrated that both ECM remodeling and fibroblast overgrowth were positively influenced by the bioreactor culture. Mechanically stimulated constructs also increased tissue maturation, with the formation of new oriented and aligned muscle fibers. Moreover, after in vivo orthotopic implantation in a surgical CDH mouse model, mechanically stimulated muscles maintained the presence of human cells within myofibers and hernia recurrence did not occur, suggesting the value of this approach for treating diaphragm defects.

15.
Plant Cell Physiol ; 52(12): 2225-35, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22025557

RESUMEN

Cell-penetrating peptides are short cationic peptides with the property of translocating across the plasma membrane and transferring macromolecules otherwise unable to permeate cell membranes. We investigated the potential ability of the protein transduction domain derived from amino acids 47-57 of the human immunodeficiency virus type 1 (HIV-1) TAT (transactivator of transcription) protein to be used as a nanocarrier for the delivery of aequorin, a Ca(2+)-sensitive photoprotein widely used as a reliable Ca(2+) reporter in cell populations. The TAT peptide, either covalently linked to apoaequorin or ionically bound to plasmids encoding differentially targeted aequorin, was supplied to plant suspension-cultured cells. The TAT-aequorin fusion protein was found to be rapidly and effectively translocated into plant cells. The chimeric molecule was internalized in fully active biological form and at levels suitable to monitor intracellular Ca(2+) concentrations. Plant cells incubated for just 5 min with TAT-aequorin responded to different environmental stimuli with the expected Ca(2+) signatures. On the other hand, TAT-mediated plasmid internalization did not provide the necessary level of transformation efficiency to allow calibration of luminescence signals into Ca(2+) concentration values. These results indicate that TAT-mediated aequorin transduction is a promising alternative to traditional plant transformation methods to monitor intracellular Ca(2+) dynamics rapidly and effectively in plant cells.


Asunto(s)
Aequorina/metabolismo , Calcio/metabolismo , Células Vegetales/metabolismo , Transducción Genética/métodos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Western Blotting , Supervivencia Celular , ADN/genética , Daucus carota/citología , Endocitosis , Humanos , Espacio Intracelular/metabolismo , Luminiscencia , Microscopía Fluorescente , Nanoestructuras , Plásmidos/genética , Transporte de Proteínas , Protoplastos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Glycine max/citología , Glycine max/metabolismo
16.
Mol Cell Biochem ; 351(1-2): 183-96, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21308481

RESUMEN

Evidence shows that extracellular ATP signals influence myogenesis, regeneration and physiology of skeletal muscle. Present work was aimed at characterizing the extracellular ATP signaling system of skeletal muscle C2C12 cells during differentiation. We show that mechanical and electrical stimulation produces substantial release of ATP from differentiated myotubes, but not from proliferating myoblasts. Extracellular ATP-hydrolyzing activity is low in myoblasts and high in myotubes, consistent with the increased expression of extracellular enzymes during differentiation. Stimulation of cells with extracellular nucleotides produces substantial Ca(2+) transients, whose amplitude and shape changed during differentiation. Consistently, C2C12 cells express several P2X and P2Y receptors, whose level changes along with maturation stages. Supplementation with either ATP or UTP stimulates proliferation of C2C12 myoblasts, whereas excessive doses were cytotoxic. The data indicate that skeletal muscle development is accompanied by major functional changes in extracellular ATP signaling.


Asunto(s)
Adenosina Trifosfato/metabolismo , Diferenciación Celular , Proliferación Celular , Músculo Esquelético/metabolismo , Transducción de Señal , Animales , Secuencia de Bases , Western Blotting , Línea Celular , Cartilla de ADN , Ratones , Músculo Esquelético/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Zebrafish ; 18(3): 231-234, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33877911

RESUMEN

The study of swimming behavior is an important part of fish biology research and the swim tunnel is used to study swimming performance as well as metabolism of fish. In this investigation, we have developed a user-friendly, automated, modular, and low-cost swim tunnel that permits to study the performance of one or more fish separately, as well as a small group of individuals. To validate our swim tunnel, we assessed swimming activity of four different species (zebrafish, medaka, guppy, and cavefish) recording reliable data of swimming behavior and performance. Because swimming behavior has been recently used in different fields from physiology to ecotoxicology, our setup could help researchers with a low-cost solution.


Asunto(s)
Peces/fisiología , Natación , Animales , Oryzias/fisiología , Poecilia/fisiología , Natación/fisiología , Pez Cebra/fisiología
18.
Am J Physiol Cell Physiol ; 298(3): C550-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20042733

RESUMEN

Sphingosine 1-phosphate (S1P) is a bioactive lipid known to control cell growth that was recently shown to act as a trophic factor for skeletal muscle, reducing the progress of denervation atrophy. The aim of this work was to investigate whether S1P is involved in skeletal muscle fiber recovery (regeneration) after myotoxic injury induced by bupivacaine. The postnatal ability of skeletal muscle to grow and regenerate is dependent on resident stem cells called satellite cells. Immunofluorescence analysis demonstrated that S1P-specific receptors S1P(1) and S1P(3) are expressed by quiescent satellite cells. Soleus muscles undergoing regeneration following injury induced by intramuscular injection of bupivacaine exhibited enhanced expression of S1P(1) receptor, while S1P(3) expression progressively decreased to adult levels. S1P(2) receptor was absent in quiescent cells but was transiently expressed in the early regenerating phases only. Administration of S1P (50 microM) at the moment of myotoxic injury caused a significant increase of the mean cross-sectional area of regenerating fibers in both rat and mouse. In separate experiments designed to test the trophic effects of S1P, neutralization of endogenous circulating S1P by intraperitoneal administration of anti-S1P antibody attenuated fiber growth. Use of selective modulators of S1P receptors indicated that S1P(1) receptor negatively and S1P(3) receptor positively modulate the early phases of regeneration, whereas S1P(2) receptor appears to be less important. The present results show that S1P signaling participates in the regenerative processes of skeletal muscle.


Asunto(s)
Lisofosfolípidos/metabolismo , Desarrollo de Músculos , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Regeneración , Células Satélite del Músculo Esquelético/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Animales , Bupivacaína , Membrana Celular/metabolismo , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Inyecciones Intramusculares , Lisofosfolípidos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Desarrollo de Músculos/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/fisiopatología , Ratas , Ratas Wistar , Receptores de Lisoesfingolípidos/efectos de los fármacos , Receptores de Lisoesfingolípidos/metabolismo , Regeneración/efectos de los fármacos , Células Satélite del Músculo Esquelético/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Esfingosina/administración & dosificación , Esfingosina/metabolismo , Factores de Tiempo
19.
Expert Rev Mol Med ; 11: e28, 2009 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-19781108

RESUMEN

Sarcoglycanopathies are a group of autosomal recessive muscle-wasting disorders caused by genetic defects in one of four cell membrane glycoproteins, alpha-, beta-, gamma- or delta-sarcoglycan. These four sarcoglycans form a subcomplex that is closely linked to the major dystrophin-associated protein complex, which is essential for membrane integrity during muscle contraction and provides a scaffold for important signalling molecules. Proper assembly, trafficking and targeting of the sarcoglycan complex is of vital importance, and mutations that severely perturb tetramer formation and localisation result in sarcoglycanopathy. Gene defects in one sarcoglycan cause the absence or reduced concentration of the other subunits. Most genetic defects generate mutated proteins that are degraded through the cell's quality control system; however, in many cases, conformational modifications do not affect the function of the protein, yet it is recognised as misfolded and prematurely degraded. Recent evidence shows that misfolded sarcoglycans could be rescued to the cell membrane by assisting their maturation along the ER secretory pathway. This review summarises the etiopathogenesis of sarcoglycanopathies and highlights the quality control machinery as a potential pharmacological target for therapy of these genetic disorders.


Asunto(s)
Complejo de Proteínas Asociado a la Distrofina/metabolismo , Retículo Endoplásmico/metabolismo , Músculo Esquelético/metabolismo , Distrofias Musculares/metabolismo , Sarcoglicanos/metabolismo , Secuencia de Aminoácidos , Animales , Membrana Celular/metabolismo , Complejo de Proteínas Asociado a la Distrofina/química , Humanos , Datos de Secuencia Molecular , Distrofias Musculares/genética , Distrofias Musculares/terapia , Mutación Missense/genética , Mutación Missense/fisiología , Transporte de Proteínas/fisiología , Sarcoglicanos/genética
20.
Am J Pathol ; 173(1): 170-81, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18535179

RESUMEN

Sarcoglycanopathies are progressive muscle-wasting disorders caused by genetic defects of four proteins, alpha-, beta-, gamma-, and delta-sarcoglycan, which are elements of a key transmembrane complex of striated muscle. The proper assembly of the sarcoglycan complex represents a critical issue of sarcoglycanopathies, as several mutations severely perturb tetramer formation. Misfolded proteins are generally degraded through the cell's quality-control system; however, this can also lead to the removal of some functional polypeptides. To explore whether it is possible to rescue sarcoglycan mutants by preventing their degradation, we generated a heterologous cell system, based on human embryonic kidney (HEK) 293 cells, constitutively expressing three (beta, gamma, and delta) of the four sarcoglycans. In these betagammadelta-HEK cells, the lack of alpha-sarcoglycan prevented complex formation and cell surface localization, wheras the presence of alpha-sarcoglycan allowed maturation and targeting of the tetramer. As in muscles of sarcoglycanopathy patients, transfection of betagammadelta-HEK cells with disease-causing alpha-sarcoglycan mutants led to dramatic reduction of the mutated proteins and the absence of the complex from the cell surface. Proteasomal inhibition reduced the degradation of mutants and facilitated the assembly and targeting of the sarcoglycan complex to the plasma membrane. These data provide important insights for the potential development of pharmacological therapies for sarcoglycanopathies.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Isoformas de Proteínas/metabolismo , Sarcoglicanos/metabolismo , Western Blotting , Línea Celular , Inhibidores de Cisteína Proteinasa/farmacología , Electroforesis en Gel de Poliacrilamida , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Leupeptinas/farmacología , Microscopía Confocal , Mutagénesis Sitio-Dirigida , Mutación , Pliegue de Proteína , Isoformas de Proteínas/química , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Sarcoglicanos/química , Sarcoglicanos/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA