Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(14)2022 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-35887248

RESUMEN

The cellular heterogeneity of the tumor environment of breast cancer (BC) is extremely complex and includes different actors such as neoplastic, stromal, and immunosuppressive cells, which contribute to the chemical and mechanical modification of the environment surrounding the tumor-exasperating immune-escaping mechanisms. In addition to molecular signals that make the tumor microenvironment (TME) unacceptable for the penetrance of the immune system, the physical properties of tumoral extracellular matrix (tECM) also have carved out a fundamental role in the processes of the protection of the tumor niche. Tumor-associated macrophages (TAMs), with an M2 immunosuppressive phenotype, are important determinants for the establishment of a tumor phenotype excluded from T cells. NF-κB transcription factors orchestrate innate immunity and represent the common thread between inflammation and cancer. Many studies have focused on canonical activation of NF-κB; however, activation of non-canonical signaling predicts poor survival and resistance to therapy. In this scenario, we demonstrated the existence of an unusual association of NF-κB components in TAMs that determines the deposition of HSPG2 that affects the stiffness of tECM. These results highlight a new mechanism counterbalanced between physical factors and a new perspective of mechano-pathology to be targeted to counteract immune evasion in BC.


Asunto(s)
FN-kappa B , Neoplasias , Humanos , Macrófagos , Neoplasias/patología , Microambiente Tumoral , Macrófagos Asociados a Tumores
2.
Int J Mol Sci ; 23(17)2022 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-36076927

RESUMEN

Before entering human clinical studies to evaluate their safety and effectiveness, new drugs and novel medical treatments are subject to extensive animal testing that are expensive and time-consuming. By contrast, advanced technologies enable the development of animal-free models that allow the efficacy of innovative therapies to be studied without sacrificing animals, while providing helpful information and details. We report on the powerful combination of 3D bioprinting (3DB) and photo-thermal therapy (PTT) applications. To this end, we realize a 3DB construct consisting of glioblastoma U87-MG cells in a 3D geometry, incorporating biomimetic keratin-coated gold nanoparticles (Ker-AuNPs) as a photo-thermal agent. The resulting plasmonic 3DB structures exhibit a homogeneous cell distribution throughout the entire volume while promoting the localization of Ker-AuNPs within the cells. A 3D immunofluorescence assay and transmission electron microscopy (TEM) confirm the uniform distribution of fluorescent-labeled Ker-AuNPs in the volume and their capability to enter the cells. Laser-assisted (λ = 532 nm) PTT experiments demonstrate the extraordinary ability of Ker-AuNPs to generate heating, producing the highest temperature rise of about 16 °C in less than 2 min.


Asunto(s)
Glioblastoma , Hipertermia Inducida , Nanopartículas del Metal , Terapia Fototérmica , Materiales Biomiméticos , Glioblastoma/terapia , Oro/química , Humanos , Queratinas/química , Nanopartículas del Metal/química , Terapia Fototérmica/métodos
3.
Int J Mol Sci ; 22(11)2021 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-34070750

RESUMEN

The immune system is a fine modulator of the tumor biology supporting or inhibiting its progression, growth, invasion and conveys the pharmacological treatment effect. Tumors, on their side, have developed escaping mechanisms from the immune system action ranging from the direct secretion of biochemical signals to an indirect reaction, in which the cellular actors of the tumor microenvironment (TME) collaborate to mechanically condition the extracellular matrix (ECM) making it inhospitable to immune cells. TME is composed of several cell lines besides cancer cells, including tumor-associated macrophages, cancer-associated fibroblasts, CD4+ and CD8+ lymphocytes, and innate immunity cells. These populations interface with each other to prepare a conservative response, capable of evading the defense mechanisms implemented by the host's immune system. The presence or absence, in particular, of cytotoxic CD8+ cells in the vicinity of the main tumor mass, is able to predict, respectively, the success or failure of drug therapy. Among various mechanisms of immunescaping, in this study, we characterized the modulation of the phenotypic profile of CD4+ and CD8+ cells in resting and activated states, in response to the mechanical pressure exerted by a three-dimensional in vitro system, able to recapitulate the rheological and stiffness properties of the tumor ECM.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Matriz Extracelular/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Escape del Tumor , Microambiente Tumoral/inmunología , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Fibroblastos Asociados al Cáncer/inmunología , Fibroblastos Asociados al Cáncer/patología , Técnicas de Cultivo de Célula , Módulo de Elasticidad , Matriz Extracelular/química , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Humanos , Hidrogeles/química , Interferón gamma/genética , Interferón gamma/inmunología , Activación de Linfocitos , Mecanotransducción Celular , Modelos Biológicos , FN-kappa B/genética , FN-kappa B/inmunología , Fenotipo , Cultivo Primario de Células , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Reología , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/inmunología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/inmunología , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral/genética , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/patología
4.
Biofabrication ; 16(4)2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38986455

RESUMEN

Over the past three decades, cell therapy development has fallen short of expectations, with many cellular sources demonstrating a 'Janus effect' and raising safety concerns. Extracellular vesicles (EVs), supported by advanced technologies, present a promising avenue in regenerative medicine, offering benefits such as immune tolerance and avoidance of negative aspects associated with cell transplants. Our previous research showcased enhanced and organized subcutaneous vascularization using three-dimensional bioprinted patches containing HUVEC-derived EVs in immunodeficient animal models. In this context, stress conditions on the cells of origin further boosted the EVs' neoangiogenic potential. Since neovascularization is the first regenerative target requiring restoration, the present study aims to complement our previous work by employing an injectable gelatin methacrylate (GelMA) hydrogel functionalized with HUVEC-derived EVs in a pathological condition of acute myocardial infarction. This bioactive hydrogel resulted in reduced fibrosis, improved contractility, and promoted angiogenesis, showing promise in countering tissue deterioration and addressing vascular deficits. Moreover, the molecular characterization of EVs through miRNome and proteomic analyses further supports their potential as bio-additives for hydrogel functionalization. This cell-free approach mitigates immune rejection and oncogenic risks, offering innovative therapeutic advantages.


Asunto(s)
Vesículas Extracelulares , Células Endoteliales de la Vena Umbilical Humana , Hidrogeles , Infarto del Miocardio , Neovascularización Fisiológica , Humanos , Animales , Infarto del Miocardio/terapia , Infarto del Miocardio/patología , Hidrogeles/química , Neovascularización Fisiológica/efectos de los fármacos , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/química , Vesículas Extracelulares/trasplante , Metacrilatos/química , Gelatina/química , Inyecciones , Masculino
5.
Biofabrication ; 15(4)2023 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-37473749

RESUMEN

In this work, we present an innovative, high-throughput rotary wet-spinning biofabrication method for manufacturing cellularized constructs composed of highly-aligned hydrogel fibers. The platform is supported by an innovative microfluidic printing head (MPH) bearing a crosslinking bath microtank with a co-axial nozzle placed at the bottom of it for the immediate gelation of extruded core/shell fibers. After a thorough characterization and optimization of the new MPH and the fiber deposition parameters, we demonstrate the suitability of the proposed system for thein vitroengineering of functional myo-substitutes. The samples produced through the described approach were first characterizedin vitroand then used as a substrate to ascertain the effects of electro-mechanical stimulation on myogenic maturation. Of note, we found a characteristic gene expression modulation of fast (MyH1), intermediate (MyH2), and slow (MyH7) twitching myosin heavy chain isoforms, depending on the applied stimulation protocol. This feature should be further investigated in the future to biofabricate engineered myo-substitutes with specific functionalities.


Asunto(s)
Bioimpresión , Hidrogeles , Hidrogeles/química , Desarrollo de Músculos/genética , Microfluídica , Bioimpresión/métodos , Impresión Tridimensional , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
6.
FEBS J ; 290(18): 4440-4464, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37166453

RESUMEN

Astrocytes are essential players in development and functions, being particularly relevant as regulators of brain energy metabolism, ionic homeostasis and synaptic transmission. They are also the major source of l-serine in the brain, which is synthesized from the glycolytic intermediate 3-phosphoglycerate through the phosphorylated pathway. l-Serine is the precursor of the two main co-agonists of the N-methyl-d-aspartate receptor, glycine and d-serine. Strikingly, dysfunctions in both l- and d-serine metabolism are associated with neurological and psychiatric disorders. Here, we exploited a differentiation protocol, based on the generation of human mature astrocytes from neural stem cells, and investigated the modification of the proteomic and metabolomic profile during the differentiation process. We show that differentiated astrocytes are more similar to mature rather than to reactive ones, and that axogenesis and pyrimidine metabolism increase up to 30 days along with the folate cycle and sphingolipid metabolism. Consistent with the proliferation and cellular maturation processes that are taking place, also the intracellular levels of l-serine, glycine, threonine, l- and d-aspartate (which level is unexpectedly higher than that of d-serine) show the same biosynthetic time course. A significant utilization of l-serine from the medium is apparent while glycine is first consumed and then released with a peak at 30 days, parallel to its intracellular level. These results underline how metabolism changes during astrocyte differentiation, highlight that d-serine synthesis is restricted in differentiated astrocytes and provide a valuable model for developing potential novel therapeutic approaches to address brain diseases, especially the ones related to serine metabolism alterations.


Asunto(s)
Astrocitos , Células Madre Pluripotentes Inducidas , Humanos , Astrocitos/metabolismo , Serina/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Proteómica , Diferenciación Celular , Receptores de N-Metil-D-Aspartato/genética , Glicina/farmacología , Glicina/metabolismo
7.
Cells ; 12(5)2023 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-36899835

RESUMEN

AL amyloidosis is caused by the misfolding of immunoglobulin light chains leading to an impaired function of tissues and organs in which they accumulate. Due to the paucity of -omics profiles from undissected samples, few studies have addressed amyloid-related damage system wide. To fill this gap, we evaluated proteome changes in the abdominal subcutaneous adipose tissue of patients affected by the AL isotypes κ and λ. Through our retrospective analysis based on graph theory, we have herein deduced new insights representing a step forward from the pioneering proteomic investigations previously published by our group. ECM/cytoskeleton, oxidative stress and proteostasis were confirmed as leading processes. In this scenario, some proteins, including glutathione peroxidase 1 (GPX1), tubulins and the TRiC complex, were classified as biologically and topologically relevant. These and other results overlap with those already reported for other amyloidoses, supporting the hypothesis that amyloidogenic proteins could induce similar mechanisms independently of the main fibril precursor and of the target tissues/organs. Of course, further studies based on larger patient cohorts and different tissues/organs will be essential, which would be a key point that would allow for a more robust selection of the main molecular players and a more accurate correlation with clinical aspects.


Asunto(s)
Amiloidosis de Cadenas Ligeras de las Inmunoglobulinas , Humanos , Proteómica/métodos , Estudios Retrospectivos , Biopsia , Grasa Subcutánea/metabolismo
8.
Proc Natl Acad Sci U S A ; 106(37): 15885-90, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19717420

RESUMEN

Primitive cells capable of generating small resistance arterioles and capillary structures in the injured myocardium have been identified repeatedly. However, these cells do not form large conductive coronary arteries that would have important implications in the management of the ischemic heart. In the current study, we determined whether the human heart possesses a class of progenitor cells that regulates the growth of endothelial cells (ECs) and smooth muscle cells (SMCs) and vasculogenesis. The expression of vascular endothelial growth-factor receptor 2 (KDR) was used, together with the stem cell antigen c-kit, to isolate and expand a resident coronary vascular progenitor cell (VPC) from human myocardial samples. Structurally, vascular niches composed of c-kit-KDR-positive VPCs were identified within the walls of coronary vessels. The VPCs were connected by gap junctions to ECs, SMCs, and fibroblasts that operate as supporting cells. In vitro, VPCs were self-renewing and clonogenic and differentiated predominantly into ECs and SMCs and partly into cardiomyocytes. To establish the functional import of VPCs, a critical stenosis was created in immunosuppressed dogs, and tagged human VPCs were injected in proximity to the constricted artery. One month later, there was an increase in coronary blood flow (CBF) distal to the stenotic artery, resulting in functional improvement of the ischemic myocardium. Regenerated large, intermediate, and small human coronary arteries and capillaries were found. In conclusion, the human heart contains a pool of VPCs that can be implemented clinically to form functionally competent coronary vessels and improve CBF in patients with ischemic cardiomyopathy.


Asunto(s)
Células Madre Adultas/citología , Vasos Coronarios/citología , Actinas/metabolismo , Células Madre Adultas/fisiología , Células Madre Adultas/trasplante , Animales , Diferenciación Celular , Proliferación Celular , Separación Celular , Conexina 43/metabolismo , Circulación Coronaria , Estenosis Coronaria/patología , Estenosis Coronaria/fisiopatología , Estenosis Coronaria/terapia , Vasos Coronarios/fisiopatología , Perros , Células Endoteliales/citología , Células Endoteliales/fisiología , Perfilación de la Expresión Génica , Humanos , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/terapia , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/fisiología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Regeneración/genética , Regeneración/fisiología , Trasplante Heterólogo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
Nutrients ; 14(13)2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-35807868

RESUMEN

The evaluation of probiotics' efficacy in treating irritable bowel syndrome is supported by an increasing number of clinical studies based on a heterogeneous approach of products tested and the patient cohort involved. Although the role of gut microbiota dysbiosis in IBS pathogenesis and the beneficial contribution of probiotics were demonstrated, a tool to discriminate symptom-specific strains and a personalised medicine protocol are still lacking. Thus, this study employs, for the first time, a method that combines the preferred reporting items for systematic reviews and meta-analysis and multi-criteria decision analysis methods in a structured decision-making tool to analyze the efficacy of probiotic mix, in order to identify the most effective formulation and to discriminate which probiotics are more efficient in treating different symptoms. The PRISMA methodology resulted in a qualitative and quantitative analysis of 104 clinical studies from 2011 to 2021, revealing a prevalence of Lactobacillus rhamnosus, Lactobacillus acidophilus, and Bifidobacterium animalis subsp. lactis. MCDA analysis showed that formulations based on Lactobacillus rhamnosus and Lactobacillus acidophilus have the highest efficacy, especially on quality of life, bloating, and abdominal pain. This methodological approach could become more specific by modelling clinical studies according to the age and gender of patients and probiotic strain.


Asunto(s)
Bifidobacterium animalis , Síndrome del Colon Irritable , Lacticaseibacillus rhamnosus , Probióticos , Técnicas de Apoyo para la Decisión , Humanos , Síndrome del Colon Irritable/tratamiento farmacológico , Lactobacillus acidophilus , Probióticos/uso terapéutico , Calidad de Vida
10.
Cardiovasc Res ; 118(8): 1872-1884, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34254111

RESUMEN

Alterations in the DMD gene, which codes for the protein dystrophin, cause forms of dystrophinopathies such as Duchenne muscular dystrophy, an X-linked disease. Cardiomyopathy linked to DMD mutations is becoming the leading cause of death in patients with dystrophinopathy. Since phenotypic pathophysiological mechanisms are not fully understood, the improvement and development of new disease models, considering their relative advantages and disadvantages, is essential. The application of genetic engineering approaches on induced pluripotent stem cells, such as gene-editing technology, enables the development of physiologically relevant human cell models for in vitro dystrophinopathy studies. The combination of induced pluripotent stem cells-derived cardiovascular cell types and 3D bioprinting technologies hold great promise for the study of dystrophin-linked cardiomyopathy. This combined approach enables the assessment of responses to physical or chemical stimuli, and the influence of pharmaceutical approaches. The critical objective of in vitro microphysiological systems is to more accurately reproduce the microenvironment observed in vivo. Ground-breaking methodology involving the connection of multiple microphysiological systems comprised of different tissues would represent a move toward precision body-on-chip disease modelling could lead to a critical expansion in what is known about inter-organ responses to disease and novel therapies that have the potential to replace animal models. In this review, we will focus on the generation, development, and application of current cellular, animal, and potential for bio-printed models, in the study of the pathophysiological mechanisms underlying dystrophin-linked cardiomyopathy in the direction of personalized medicine.


Asunto(s)
Cardiomiopatías , Células Madre Pluripotentes Inducidas , Distrofia Muscular de Duchenne , Animales , Cardiomiopatías/genética , Cardiomiopatías/terapia , Distrofina/genética , Distrofina/metabolismo , Corazón , Células Madre Pluripotentes Inducidas/metabolismo , Distrofia Muscular de Duchenne/genética
11.
Cells ; 11(18)2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36139463

RESUMEN

Abscisic acid (ABA) regulates plant responses to stress, partly via NO. In mammals, ABA stimulates NO production by innate immune cells and keratinocytes, glucose uptake and mitochondrial respiration by skeletal myocytes and improves blood glucose homeostasis through its receptors LANCL1 and LANCL2. We hypothesized a role for the ABA-LANCL1/2 system in cardiomyocyte protection from hypoxia via NO. The effect of ABA and of the silencing or overexpression of LANCL1 and LANCL2 were investigated in H9c2 rat cardiomyoblasts under normoxia or hypoxia/reoxygenation. In H9c2, hypoxia induced ABA release, and ABA stimulated NO production. ABA increased the survival of H9c2 to hypoxia, and L-NAME, an inhibitor of NO synthase (NOS), abrogated this effect. ABA also increased glucose uptake and NADPH levels and increased phosphorylation of Akt, AMPK and eNOS. Overexpression or silencing of LANCL1/2 significantly increased or decreased, respectively, transcription, expression and phosphorylation of AMPK, Akt and eNOS; transcription of NAMPT, Sirt1 and the arginine transporter. The mitochondrial proton gradient and cell vitality increased in LANCL1/2-overexpressing vs. -silenced cells after hypoxia/reoxygenation, and L-NAME abrogated this difference. These results implicate the ABA-LANCL1/2 hormone-receptor system in NO-mediated cardiomyocyte protection against hypoxia.


Asunto(s)
Ácido Abscísico , Miocitos Cardíacos , Proteínas Quinasas Activadas por AMP/metabolismo , Ácido Abscísico/metabolismo , Animales , Glucemia/metabolismo , Hipoxia de la Célula , Hormonas/metabolismo , Proteínas de la Membrana/metabolismo , Miocitos Cardíacos/metabolismo , NADP/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptores Acoplados a Proteínas G , Sirtuina 1/metabolismo
12.
Proc Natl Acad Sci U S A ; 105(5): 1668-73, 2008 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-18216245

RESUMEN

Coronary artery disease is the most common cause of cardiac failure in the Western world, and to date there is no alternative to bypass surgery for severe coronary atherosclerosis. We report that c-kit-positive cardiac progenitor cells (CPCs) activated with insulin-like growth factor 1 and hepatocyte growth factor before their injection in proximity of the site of occlusion of the left coronary artery in rats, engrafted within the host myocardium forming temporary niches. Subsequently, CPCs divided and differentiated into endothelial cells and smooth muscle cells and, to a lesser extent, into cardiomyocytes. The acquisition of vascular lineages appeared to be mediated by the up-regulation of hypoxia-inducible factor 1alpha, which promoted the synthesis and secretion of stromal-derived factor 1 from hypoxic coronary vessels. Stromal-derived factor 1 was critical in the conversion of CPCs to the vascular fate. CPCs formed conductive and intermediate-sized coronary arteries together with resistance arterioles and capillaries. The new vessels were connected with the primary coronary circulation, and this increase in vascularization more than doubled myocardial blood flow in the infarcted myocardium. This beneficial effect, together with myocardial regeneration attenuated postinfarction dilated myopathy, reduced infarct size and improved function. In conclusion, locally delivered activated CPCs generate de novo coronary vasculature and may be implemented clinically for restoration of blood supply to the ischemic myocardium.


Asunto(s)
Vasos Coronarios/fisiología , Mioblastos Cardíacos/fisiología , Neovascularización Fisiológica , Regeneración , Células Madre/fisiología , Animales , Diferenciación Celular , Quimiocina CXCL12/metabolismo , Vasos Coronarios/citología , Células Endoteliales/citología , Femenino , Factor de Crecimiento de Hepatocito/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Mioblastos Cardíacos/efectos de los fármacos , Mioblastos Cardíacos/trasplante , Isquemia Miocárdica/metabolismo , Miocitos del Músculo Liso/citología , Proteínas Proto-Oncogénicas c-kit/análisis , Ratas , Ratas Endogámicas F344 , Trasplante de Células Madre , Células Madre/química , Células Madre/efectos de los fármacos
13.
Stem Cell Res ; 55: 102487, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34419748

RESUMEN

Emery-Dreifuss muscular dystrophy type 1 (EDMD1) is a rare genetic disease caused by mutations in the EMD gene coding for a nuclear envelope protein emerin. We generated and characterized induced pluripotent stem cells (iPSCs) from two EDMD1 patients bearing a mutation c.del153C and from one healthy donor. That mutation leads to generation of premature STOP codon. Established iPSCs are very valuable tool for disease pathogenesis investigation and for the development of new therapeutic methods after differentiation to cardiac or muscle cells. Obtained iPSCs show the proper morphology, pluripotency markers expression, normal karyotype and potential to differentiate into three germ layers.


Asunto(s)
Células Madre Pluripotentes Inducidas , Distrofia Muscular de Emery-Dreifuss , Diferenciación Celular , Células Cultivadas , Células Clonales , Humanos , Distrofia Muscular de Emery-Dreifuss/genética , Mutación
14.
Biomolecules ; 11(4)2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33917623

RESUMEN

The Cdkn2a locus is one of the most studied tumor suppressor loci in the context of several cancer types. However, in the last years, its expression has also been linked to terminal differentiation and the activation of the senescence program in different cellular subtypes. Knock-out (KO) of the entire locus enhances the capability of stem cells to proliferate in some tissues and respond to severe physiological and non-physiological damages in different organs, including the heart. Emery-Dreifuss muscular dystrophy (EDMD) is characterized by severe contractures and muscle loss at the level of skeletal muscles of the elbows, ankles and neck, and by dilated cardiomyopathy. We have recently demonstrated, using the LMNA Δ8-11 murine model of Emery-Dreifuss muscular dystrophy (EDMD), that dystrophic muscle stem cells prematurely express non-lineage-specific genes early on during postnatal growth, leading to rapid exhaustion of the muscle stem cell pool. Knock-out of the Cdkn2a locus in EDMD dystrophic mice partially restores muscle stem cell properties. In the present study, we describe the cardiac phenotype of the LMNA Δ8-11 mouse model and functionally characterize the effects of KO of the Cdkn2a locus on heart functions and life expectancy.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Distrofia Muscular de Emery-Dreifuss/patología , Animales , Apoptosis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Modelos Animales de Enfermedad , Sitios Genéticos , Genotipo , Lamina Tipo A/deficiencia , Lamina Tipo A/genética , Longevidad , Ratones , Ratones Noqueados , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/mortalidad , Miocardio/citología , Miocardio/metabolismo , Miocardio/patología , Fenotipo , Células Madre/citología , Células Madre/metabolismo , Tasa de Supervivencia
15.
Front Bioeng Biotechnol ; 9: 732130, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34604190

RESUMEN

In the last decades, biomedical research has significantly boomed in the academia and industrial sectors, and it is expected to continue to grow at a rapid pace in the future. An in-depth analysis of such growth is not trivial, given the intrinsic multidisciplinary nature of biomedical research. Nevertheless, technological advances are among the main factors which have enabled such progress. In this review, we discuss the contribution of two state-of-the-art technologies-namely biofabrication and organ-on-a-chip-in a selection of biomedical research areas. We start by providing an overview of these technologies and their capacities in fabricating advanced in vitro tissue/organ models. We then analyze their impact on addressing a range of current biomedical challenges. Ultimately, we speculate about their future developments by integrating these technologies with other cutting-edge research fields such as artificial intelligence and big data analysis.

16.
Biofabrication ; 13(3)2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33434889

RESUMEN

Extracellular vesicles (EVs) have become a key tool in the biotechnological landscape due to their well-documented ability to mediate intercellular communication. This feature has been explored and is under constant investigation by researchers, who have demonstrated the important role of EVs in several research fields ranging from oncology to immunology and diagnostics to regenerative medicine. Unfortunately, there are still some limitations to overcome before clinical application, including the inability to confine the EVs to strategically defined sites of interest to avoid side effects. In this study, for the first time, EV application is supported by 3D bioprinting technology to develop a new strategy for applying the angiogenic cargo of human umbilical vein endothelial cell-derived EVs in regenerative medicine. EVs, derived from human endothelial cells and grown under different stressed conditions, were collected and used as bioadditives for the formulation of advanced bioinks. Afterin vivosubcutaneous implantation, we demonstrated that the bioprinted 3D structures, loaded with EVs, supported the formation of a new functional vasculaturein situ, consisting of blood-perfused microvessels recapitulating the printed pattern. The results obtained in this study favour the development of new therapeutic approaches for critical clinical conditions, such as the need for prompt revascularization of ischaemic tissues, which represent the fundamental substrate for advanced regenerative medicine applications.


Asunto(s)
Bioimpresión , Vesículas Extracelulares , Impresión Tridimensional , Comunicación Celular , Células Endoteliales de la Vena Umbilical Humana , Humanos , Medicina Regenerativa
17.
Dis Model Mech ; 14(12)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34796900

RESUMEN

Large-animal models for Duchenne muscular dystrophy (DMD) are crucial for the evaluation of diagnostic procedures and treatment strategies. Pigs cloned from male cells lacking DMD exon 52 (DMDΔ52) exhibit molecular, clinical and pathological hallmarks of DMD, but die before sexual maturity and cannot be propagated by breeding. Therefore, we generated female DMD+/- carriers. A single founder animal had 11 litters with 29 DMDY/-, 34 DMD+/- as well as 36 male and 29 female wild-type offspring. Breeding with F1 and F2 DMD+/- carriers resulted in an additional 114 DMDY/- piglets. With intensive neonatal management, the majority survived for 3-4 months, providing statistically relevant cohorts for experimental studies. Pathological investigations and proteome studies of skeletal muscles and myocardium confirmed the resemblance to human disease mechanisms. Importantly, DMDY/- pigs displayed progressive myocardial fibrosis and increased expression of connexin-43, associated with significantly reduced left ventricular ejection fraction, at 3 months. Furthermore, behavioral tests provided evidence for impaired cognitive ability. Our breeding cohort of DMDΔ52 pigs and standardized tissue repositories provide important resources for studying DMD disease mechanisms and for testing novel treatment strategies.


Asunto(s)
Cardiomiopatías , Distrofia Muscular de Duchenne , Animales , Cardiomiopatías/patología , Femenino , Humanos , Masculino , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/patología , Volumen Sistólico , Porcinos , Función Ventricular Izquierda
18.
Circ Res ; 102(5): 597-606, 2008 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-18202313

RESUMEN

Heart failure is the leading cause of death in the elderly, but whether this is the result of a primary aging myopathy dictated by depletion of the cardiac progenitor cell (CPC) pool is unknown. Similarly, whether current lifespan reflects the ineluctable genetic clock or heart failure interferes with the genetically determined fate of the organ and organism is an important question. We have identified that chronological age leads to telomeric shortening in CPCs, which by necessity generate a differentiated progeny that rapidly acquires the senescent phenotype conditioning organ aging. CPC aging is mediated by attenuation of the insulin-like growth factor-1/insulin-like growth factor-1 receptor and hepatocyte growth factor/c-Met systems, which do not counteract any longer the CPC renin-angiotensin system, resulting in cellular senescence, growth arrest, and apoptosis. However, pulse-chase 5-bromodeoxyuridine-labeling assay revealed that the senescent heart contains functionally competent CPCs that have the properties of stem cells. This subset of telomerase-competent CPCs have long telomeres and, following activation, migrate to the regions of damage, where they generate a population of young cardiomyocytes, reversing partly the aging myopathy. The senescent heart phenotype and heart failure are corrected to some extent, leading to prolongation of maximum lifespan.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Insuficiencia Cardíaca/terapia , Factor de Crecimiento de Hepatocito/uso terapéutico , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Miocitos Cardíacos/efectos de los fármacos , Células Madre Adultas/metabolismo , Células Madre Adultas/patología , Envejecimiento/patología , Animales , Antígenos de Diferenciación/biosíntesis , Apoptosis/efectos de los fármacos , Recuento de Células , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Corazón/efectos de los fármacos , Corazón/crecimiento & desarrollo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Masculino , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fenotipo , Ratas , Ratas Endogámicas F344 , Receptores de Factores de Crecimiento/metabolismo , Regeneración/efectos de los fármacos , Tasa de Supervivencia , Telómero/metabolismo
19.
Cells ; 9(6)2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32585911

RESUMEN

The recent advances, offered by cell therapy in the regenerative medicine field, offer a revolutionary potential for the development of innovative cures to restore compromised physiological functions or organs. Adult myogenic precursors, such as myoblasts or satellite cells, possess a marked regenerative capacity, but the exploitation of this potential still encounters significant challenges in clinical application, due to low rate of proliferation in vitro, as well as a reduced self-renewal capacity. In this scenario, induced pluripotent stem cells (iPSCs) can offer not only an inexhaustible source of cells for regenerative therapeutic approaches, but also a valuable alternative for in vitro modeling of patient-specific diseases. In this study we established a reliable protocol to induce the myogenic differentiation of iPSCs, generated from pericytes and fibroblasts, exploiting skeletal muscle-derived extracellular vesicles (EVs), in combination with chemically defined factors. This genetic integration-free approach generates functional skeletal myotubes maintaining the engraftment ability in vivo. Our results demonstrate evidence that EVs can act as biological "shuttles" to deliver specific bioactive molecules for a successful transgene-free differentiation offering new opportunities for disease modeling and regenerative approaches.


Asunto(s)
Vesículas Extracelulares/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Desarrollo de Músculos/fisiología , Músculo Esquelético/metabolismo , Adulto , Animales , Diferenciación Celular , Voluntarios Sanos , Humanos , Masculino , Ratones , Adulto Joven
20.
J Clin Invest ; 130(5): 2408-2421, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31999646

RESUMEN

Lamin A is a component of the inner nuclear membrane that, together with epigenetic factors, organizes the genome in higher order structures required for transcriptional control. Mutations in the lamin A/C gene cause several diseases belonging to the class of laminopathies, including muscular dystrophies. Nevertheless, molecular mechanisms involved in the pathogenesis of lamin A-dependent dystrophies are still largely unknown. The polycomb group (PcG) of proteins are epigenetic repressors and lamin A interactors, primarily involved in the maintenance of cell identity. Using a murine model of Emery-Dreifuss muscular dystrophy (EDMD), we show here that lamin A loss deregulated PcG positioning in muscle satellite stem cells, leading to derepression of non-muscle-specific genes and p16INK4a, a senescence driver encoded in the Cdkn2a locus. This aberrant transcriptional program caused impairment in self-renewal, loss of cell identity, and premature exhaustion of the quiescent satellite cell pool. Genetic ablation of the Cdkn2a locus restored muscle stem cell properties in lamin A/C-null dystrophic mice. Our findings establish a direct link between lamin A and PcG epigenetic silencing and indicate that lamin A-dependent muscular dystrophy can be ascribed to intrinsic epigenetic dysfunctions of muscle stem cells.


Asunto(s)
Epigénesis Genética , Lamina Tipo A/biosíntesis , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Emery-Dreifuss/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Lamina Tipo A/genética , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patología , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/patología , Proteínas del Grupo Polycomb/genética , Proteínas Represoras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA