Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
J Mol Cell Cardiol ; 193: 25-35, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38768805

RESUMEN

The YTH-domain family (YTHDF) of RNA binding proteins can control gene expression at the post-transcriptional level by regulating mRNAs with N6-methyladenosine (m6A) modifications. Despite the established importance of m6A in the heart, the cardiac role of specific m6A-binding proteins remains unclear. Here, we characterized the function of YTHDF1 in cardiomyocytes using a newly generated cardiac-restricted mouse model. Deletion of YTHDF1 in adult cardiomyocytes led to hypertrophy, fibrosis, and dysfunction. Using mass spectrometry, we identified the necessity of YTHDF1 for the expression of cardiomyocyte membrane raft proteins. Specifically, YTHDF1 bound to m6A-modified Caveolin 1 (Cav1) mRNA and favored its translation. We further demonstrated that YTHDF1 regulates downstream ERK signaling. Altogether, our findings highlight a novel role for YTHDF1 as a post-transcriptional regulator of caveolar proteins which is necessary for the maintenance of cardiac function.

2.
3.
Circ Res ; 132(4): 519-540, 2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36795845

RESUMEN

During cardiac development and morphogenesis, cardiac progenitor cells differentiate into cardiomyocytes that expand in number and size to generate the fully formed heart. Much is known about the factors that regulate initial differentiation of cardiomyocytes, and there is ongoing research to identify how these fetal and immature cardiomyocytes develop into fully functioning, mature cells. Accumulating evidence indicates that maturation limits proliferation and conversely proliferation occurs rarely in cardiomyocytes of the adult myocardium. We term this oppositional interplay the proliferation-maturation dichotomy. Here we review the factors that are involved in this interplay and discuss how a better understanding of the proliferation-maturation dichotomy could advance the utility of human induced pluripotent stem cell-derived cardiomyocytes for modeling in 3-dimensional engineered cardiac tissues to obtain truly adult-level function.


Asunto(s)
Células Madre Pluripotentes Inducidas , Ingeniería de Tejidos , Humanos , Ingeniería de Tejidos/métodos , Miocitos Cardíacos/fisiología , Miocardio , Diferenciación Celular/fisiología , Proliferación Celular
5.
Stem Cell Reports ; 17(9): 2005-2022, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-35931076

RESUMEN

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) provide great opportunities for mechanistic dissection of human cardiac pathophysiology; however, hiPSC-CMs remain immature relative to the adult heart. To identify novel signaling pathways driving the maturation process during heart development, we analyzed published transcriptional and epigenetic datasets from hiPSC-CMs and prenatal and postnatal human hearts. These analyses revealed that several components of the MAPK and PI3K-AKT pathways are downregulated in the postnatal heart. Here, we show that dual inhibition of these pathways for only 5 days significantly enhances the maturation of day 30 hiPSC-CMs in many domains: hypertrophy, multinucleation, metabolism, T-tubule density, calcium handling, and electrophysiology, many equivalent to day 60 hiPSC-CMs. These data indicate that the MAPK/PI3K/AKT pathways are involved in cardiomyocyte maturation and provide proof of concept for the manipulation of key signaling pathways for optimal hiPSC-CM maturation, a critical aspect of faithful in vitro modeling of cardiac pathologies and subsequent drug discovery.


Asunto(s)
Células Madre Pluripotentes Inducidas , Diferenciación Celular/fisiología , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Recién Nacido , Miocitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
7.
Front Cell Dev Biol ; 10: 806564, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35663393

RESUMEN

The heart is one of the least regenerative organs. This is in large part due to the inability of adult mammalian cardiomyocytes to proliferate and divide. In recent years, a number of small molecules and molecular targets have been identified to stimulate cardiomyocyte proliferation, including p38 inhibition, YAP-Tead activation, fibroblast growth factor 1 and Neuregulin 1. Despite these exciting initial findings, a therapeutic approach to enhance cardiomyocyte proliferation in vivo is still lacking. We hypothesized that a more comprehensive in vitro validation using live-cell imaging and assessment of the proliferative effects on various cardiomyocyte sources might identify the most potent proliferative stimuli. Here, we used previously published stimuli to determine their proliferative effect on cardiomyocytes from different species and isolated from different developmental timepoints. Although all stimuli enhanced DNA synthesis and Histone H3 phosphorylation in neonatal rat ventricular cardiomyocytes to similar degrees, these effects varied substantially in mouse cardiomyocytes and human iPSC-derived cardiomyocytes. Our results highlight p21 inhibition and Yap-Tead activation as potent proliferative strategies to induce cultured cardiomyocyte cell cycle activity across mouse, rat and human cardiomyocytes.

8.
Circ Res ; 129(12): 1086-1101, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34645281

RESUMEN

RATIONALE: The initial hypertrophy response to cardiac pressure overload is considered compensatory, but with sustained stress, it eventually leads to heart failure. Recently, a role for recruited macrophages in determining the transition from compensated to decompensated hypertrophy has been established. However, whether cardiac resident immune cells influence the early phase of hypertrophy development has not been established. OBJECTIVE: To assess the role of cardiac immune cells in the early hypertrophy response to cardiac pressure overload induced by transverse aortic constriction (TAC). METHODS AND RESULTS: We performed cytometry by time-of-flight to determine the identity and abundance of immune cells in the heart at 1 and 4 weeks after TAC. We observed a substantial increase in cardiac macrophages 1 week after TAC. We then conducted Cite-Seq single-cell RNA sequencing of cardiac immune cells isolated from 4 sham and 6 TAC hearts. We identified 12 clusters of monocytes and macrophages, categorized as either resident or recruited macrophages, that showed remarkable changes in their abundance between sham and TAC conditions. To determine the role of cardiac resident macrophages early in the response to a hypertrophic stimulus, we used a blocking antibody against macrophage colony-stimulating factor 1 receptor (CD115). As blocking CD115 initially depletes all macrophages, we allowed the replenishment of recruited macrophages by monocytes before performing TAC. This preferential depletion of resident macrophages resulted in enhanced fibrosis and a blunted angiogenesis response to TAC. Macrophage depletion in CCR2 (C-C chemokine receptor type 2) knockout mice showed that aggravated fibrosis was primarily caused by the recruitment of monocyte-derived macrophages. Finally, 6 weeks after TAC these early events lead to depressed cardiac function and enhanced fibrosis, despite complete restoration of cardiac immune cells. CONCLUSIONS: Cardiac resident macrophages are a heterogeneous population of immune cells with key roles in stimulating angiogenesis and inhibiting fibrosis in response to cardiac pressure overload.


Asunto(s)
Cardiomegalia/metabolismo , Macrófagos/metabolismo , Neovascularización Fisiológica , Animales , Cardiomegalia/patología , Células Cultivadas , Fibrosis , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Receptores CCR2/genética , Receptores CCR2/metabolismo , Análisis de la Célula Individual , Transcriptoma
9.
Nat Commun ; 12(1): 4808, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34376683

RESUMEN

Myocardial regeneration is restricted to early postnatal life, when mammalian cardiomyocytes still retain the ability to proliferate. The molecular cues that induce cell cycle arrest of neonatal cardiomyocytes towards terminally differentiated adult heart muscle cells remain obscure. Here we report that the miR-106b~25 cluster is higher expressed in the early postnatal myocardium and decreases in expression towards adulthood, especially under conditions of overload, and orchestrates the transition of cardiomyocyte hyperplasia towards cell cycle arrest and hypertrophy by virtue of its targetome. In line, gene delivery of miR-106b~25 to the mouse heart provokes cardiomyocyte proliferation by targeting a network of negative cell cycle regulators including E2f5, Cdkn1c, Ccne1 and Wee1. Conversely, gene-targeted miR-106b~25 null mice display spontaneous hypertrophic remodeling and exaggerated remodeling to overload by derepression of the prohypertrophic transcription factors Hand2 and Mef2d. Taking advantage of the regulatory function of miR-106b~25 on cardiomyocyte hyperplasia and hypertrophy, viral gene delivery of miR-106b~25 provokes nearly complete regeneration of the adult myocardium after ischemic injury. Our data demonstrate that exploitation of conserved molecular programs can enhance the regenerative capacity of the injured heart.


Asunto(s)
MicroARNs/genética , Infarto del Miocardio/genética , Miocitos Cardíacos/metabolismo , Regeneración/genética , Animales , Animales Recién Nacidos , Cardiomegalia/genética , Células Cultivadas , Ecocardiografía , Regulación de la Expresión Génica , Humanos , Hiperplasia/genética , Ratones , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Methods Mol Biol ; 2158: 323-336, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32857384

RESUMEN

Genetic lineage tracing is accomplished using bi-transgenic mice, where one allele is altered to express Cre recombinase, and another allele encodes a Cre-dependent genetic reporter protein. Once Cre is activated (constitutive or in response to tamoxifen), the marker gene-expressing cells become indelibly labeled by the reporter protein. Therefore, daughter cells derived from labeled cells are permanently labeled even if the marker gene that drove Cre recombinase expression is no longer expressed in these cells. This system is commonly used to label putative progenitor cells and determine the fate of their progeny. Here, we describe the use of c-kit-based genetic lineage-tracing mouse line as an example and discuss caveats for performing these types of experiments.


Asunto(s)
Linaje de la Célula/genética , Rastreo Celular/métodos , Células Madre/química , Células Madre/metabolismo , Animales , Expresión Génica , Genes Reporteros , Ligamiento Genético , Proteínas Fluorescentes Verdes/genética , Integrasas/genética , Integrasas/metabolismo , Ratones , Ratones Transgénicos , Modelos Animales , Miocitos Cardíacos/química , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-kit/análisis , Proteínas Proto-Oncogénicas c-kit/genética , Células Madre/citología , Tamoxifeno/farmacología
11.
J Vis Exp ; (164)2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-33104059

RESUMEN

The adult mammalian heart is composed of various cell types including cardiomyocytes, endothelial cells and fibroblasts. Since it is difficult to reliably identify nuclei of cardiomyocytes on histological sections, many groups rely on isolating viable cardiomyocytes prior to fixation to perform immunostaining. However, these live cardiomyocyte isolation techniques require optimization to maximize the yield, viability and quality of the samples, with inherent fluctuations from sample to sample despite maximum optimization. Here, we report a reproducible protocol, involving fixation prior to enzymatic digestion of the heart, which leads to maximum yield while preserving the in vivo morphology of individual cardiomyocytes. We further developed an automated analysis platform to determine the number of nuclei and DNA content per nucleus for individual cardiomyocytes. After exposing the chest cavity, the heart was arrested in diastole by perfusion with 60 mM KCl in PBS. Next, the heart was fixed in 4% paraformaldehyde (PFA) solution, and then digested with 60 mg/mL collagenase solution. After digestions, cells were singularized by trituration, and the cardiomyocyte fraction was enriched via differential centrifugation. Isolated cardiomyocytes were stained for Troponin T and α-actinin to assess purity of the obtained population. Furthermore, we developed an image analysis platform to determine cardiomyocyte nucleation and ploidy status following DAPI staining. Image based ploidy assessments led to consistent and reproducible results. Thus, with this protocol, it is possible to preserve native morphology of individual cardiomyocytes to allow immunocytochemistry and DNA content analysis while achieving maximum yield.


Asunto(s)
Separación Celular/métodos , Miocardio/citología , Miocitos Cardíacos/citología , Ploidias , Fijación del Tejido , Animales , Núcleo Celular/metabolismo , ADN/metabolismo , Imagenología Tridimensional , Inmunohistoquímica , Perfusión , Programas Informáticos , Soluciones , Coloración y Etiquetado
12.
Curr Heart Fail Rep ; 17(5): 225-233, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32686010

RESUMEN

PURPOSE OF REVIEW: The loss of contractile function after heart injury remains one of the major healthcare issues of our time. One strategy to deal with this problem would be to increase the number of cardiomyocytes to enhance cardiac function. In the last couple of years, reactivation of cardiomyocyte proliferation has repeatedly demonstrated to aid in functional recovery after cardiac injury. RECENT FINDINGS: The Tgf-ß superfamily plays key roles during development of the heart and populating the embryonic heart with cardiomyocytes. In this review, we discuss the role of Tgf-ß signaling in regulating cardiomyocyte proliferation during development and in the setting of cardiac regeneration. Although various pathways to induce cardiomyocyte proliferation have been established, the extent to which cardiomyocyte proliferation requires or involves activation of the Tgf-ß superfamily is not entirely clear. More research is needed to better understand cross-talk between pathways that regulate cardiomyocyte proliferation.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proliferación Celular , Insuficiencia Cardíaca/diagnóstico , Humanos , Miocitos Cardíacos/patología , Transducción de Señal
13.
FASEB J ; 34(4): 5642-5657, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32100368

RESUMEN

The adult mammalian heart has a limited regenerative capacity. Therefore, identification of endogenous cells and mechanisms that contribute to cardiac regeneration is essential for the development of targeted therapies. The side population (SP) phenotype has been used to enrich for stem cells throughout the body; however, SP cells isolated from the heart have been studied exclusively in cell culture or after transplantation, limiting our understanding of their function in vivo. We generated a new Abcg2-driven lineage-tracing mouse model with efficient labeling of SP cells. Labeled SP cells give rise to terminally differentiated cells in bone marrow and intestines. In the heart, labeled SP cells give rise to lineage-traced cardiomyocytes under homeostatic conditions with an increase in this contribution following cardiac injury. Instead of differentiating into cardiomyocytes like proposed cardiac progenitor cells, cardiac SP cells fuse with preexisting cardiomyocytes to stimulate cardiomyocyte cell cycle reentry. Our study is the first to show that fusion between cardiomyocytes and non-cardiomyocytes, identified by the SP phenotype, contribute to endogenous cardiac regeneration by triggering cardiomyocyte cell cycle reentry in the adult mammalian heart.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/fisiología , Diferenciación Celular , Isquemia Miocárdica/patología , Miocitos Cardíacos/citología , Regeneración , Células de Población Lateral/citología , Animales , Trasplante de Médula Ósea , Linaje de la Célula , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Noqueados , Isquemia Miocárdica/terapia , Miocitos Cardíacos/metabolismo , Células de Población Lateral/metabolismo
15.
Biotechnol Bioeng ; 116(9): 2353-2363, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31038193

RESUMEN

Local delivery of viral vectors can enhance the efficacy of therapies by selectively affecting necessary tissues and reducing the required vector dose. Pluronic F127 is a thermosensitive polymer that undergoes a solution-gelation (sol-gel) transition as temperature increases and can deliver vectors without damaging them. While pluronics can be spread over large areas, such as the surface of an organ, before gelation, they lack sufficient adhesivity to remain attached to some tissues, such as the surface of the heart or mucosal surfaces. Here, we utilized blends of pluronic F127 and polycarbophil (PCB), a mucoadhesive agent, to provide the necessary adhesivity for local delivery of viral vectors to the cardiac muscle. The effects of PCB concentration on adhesive properties, sol-gel temperature transition and cytocompatibility were evaluated. Rheological studies showed that PCB decreased the sol-gel transition temperature at concentrations >1% and increased the adhesive properties of the gel. Furthermore, these gels were able to deliver viral vectors and transduce cells in vitro and in vivo in a neonatal mouse apical resection model. These gels could be a useful platform for delivering viral vectors over the surface of organs where increased adhesivity is required.


Asunto(s)
Resinas Acrílicas , Técnicas de Transferencia de Gen , Vectores Genéticos , Miocardio/metabolismo , Poloxámero , Adhesivos Tisulares , Virus , Resinas Acrílicas/química , Resinas Acrílicas/farmacología , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Ensayo de Materiales , Poloxámero/química , Poloxámero/farmacología , Adhesivos Tisulares/química , Adhesivos Tisulares/farmacología
16.
Circulation ; 139(4): 533-545, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30586742

RESUMEN

BACKGROUND: N6-Methyladenosine (m6A) methylation is the most prevalent internal posttranscriptional modification on mammalian mRNA. The role of m6A mRNA methylation in the heart is not known. METHODS: To determine the role of m6A methylation in the heart, we isolated primary cardiomyocytes and performed m6A immunoprecipitation followed by RNA sequencing. We then generated genetic tools to modulate m6A levels in cardiomyocytes by manipulating the levels of the m6A RNA methylase methyltransferase-like 3 (METTL3) both in culture and in vivo. We generated cardiac-restricted gain- and loss-of-function mouse models to allow assessment of the METTL3-m6A pathway in cardiac homeostasis and function. RESULTS: We measured the level of m6A methylation on cardiomyocyte mRNA, and found a significant increase in response to hypertrophic stimulation, suggesting a potential role for m6A methylation in the development of cardiomyocyte hypertrophy. Analysis of m6A methylation showed significant enrichment in genes that regulate kinases and intracellular signaling pathways. Inhibition of METTL3 completely abrogated the ability of cardiomyocytes to undergo hypertrophy when stimulated to grow, whereas increased expression of the m6A RNA methylase METTL3 was sufficient to promote cardiomyocyte hypertrophy both in vitro and in vivo. Finally, cardiac-specific METTL3 knockout mice exhibit morphological and functional signs of heart failure with aging and stress, showing the necessity of RNA methylation for the maintenance of cardiac homeostasis. CONCLUSIONS: Our study identified METTL3-mediated methylation of mRNA on N6-adenosines as a dynamic modification that is enhanced in response to hypertrophic stimuli and is necessary for a normal hypertrophic response in cardiomyocytes. Enhanced m6A RNA methylation results in compensated cardiac hypertrophy, whereas diminished m6A drives eccentric cardiomyocyte remodeling and dysfunction, highlighting the critical importance of this novel stress-response mechanism in the heart for maintaining normal cardiac function.


Asunto(s)
Adenosina/análogos & derivados , Hipertrofia Ventricular Izquierda/enzimología , Metiltransferasas/metabolismo , Miocitos Cardíacos/enzimología , Función Ventricular Izquierda , Remodelación Ventricular , Adenosina/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Metiltransferasas/deficiencia , Metiltransferasas/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/patología , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Transducción de Señal
18.
Nat Commun ; 9(1): 4237, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30315164

RESUMEN

The mammalian heart has a limited regenerative capacity and typically progresses to heart failure following injury. Here, we defined a hedgehog (HH)-Gli1-Mycn network for cardiomyocyte proliferation and heart regeneration from amphibians to mammals. Using a genome-wide screen, we verified that HH signaling was essential for heart regeneration in the injured newt. Next, pharmacological and genetic loss- and gain-of-function of HH signaling demonstrated the essential requirement for HH signaling in the neonatal, adolescent, and adult mouse heart regeneration, and in the proliferation of hiPSC-derived cardiomyocytes. Fate-mapping and molecular biological studies revealed that HH signaling, via a HH-Gli1-Mycn network, contributed to heart regeneration by inducing proliferation of pre-existing cardiomyocytes and not by de novo cardiomyogenesis. Further, Mycn mRNA transfection experiments recapitulated the effects of HH signaling and promoted adult cardiomyocyte proliferation. These studies defined an evolutionarily conserved function of HH signaling that may serve as a platform for human regenerative therapies.


Asunto(s)
Corazón/fisiología , Proteínas Hedgehog/metabolismo , Proteína Proto-Oncogénica N-Myc/metabolismo , Regeneración/fisiología , Salamandridae/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Animales , Proliferación Celular/genética , Proliferación Celular/fisiología , Proteínas Hedgehog/genética , Humanos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Proteína Proto-Oncogénica N-Myc/genética , Regeneración/genética , Salamandridae/fisiología , Transducción de Señal , Proteína con Dedos de Zinc GLI1/genética
19.
Circ Heart Fail ; 11(8): e004867, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30354563

RESUMEN

BACKGROUND: Advances in radiotherapy for thoracic cancers have resulted in improvement of survival. However, radiation exposure to the heart can induce cardiotoxicity. No therapy is currently available to inhibit these untoward effects. We examined whether a small tetrapeptide, N-acetyl-Ser-Asp-Lys-Pro (Ac-SDKP), can counteract radiation-induced cardiotoxicity by inhibiting macrophage-dependent inflammatory and fibrotic pathways. METHODS AND RESULTS: After characterizing a rat model of cardiac irradiation with magnetic resonance imaging protocols, we examined the effects of Ac-SDKP in radiation-induced cardiomyopathy. We treated rats with Ac-SDKP for 18 weeks. We then compared myocardial contractile function and extracellular matrix by cardiac magnetic resonance imaging and the extent of inflammation, fibrosis, and Mac-2 (galectin-3) release by tissue analyses. Because Mac-2 is a crucial macrophage-derived mediator of fibrosis, we performed studies to determine Mac-2 synthesis by macrophages in response to radiation, and change in profibrotic responses by Mac-2 gene depleted cardiac fibroblasts after radiation. Cardiac irradiation diminished myocardial contractile velocities and enhanced extracellular matrix deposition. This was accompanied by macrophage infiltration, fibrosis, cardiomyocyte apoptosis, and cardiac Mac-2 expression. Ac-SDKP strongly inhibited these detrimental effects. Ac-SDKP migrated into the perinuclear cytoplasm of the macrophages and inhibited radiation-induced Mac-2 release. Cardiac fibroblasts lacking the Mac-2 gene showed reduced transforming growth factor ß1, collagen I, and collagen III expression after radiation exposure. CONCLUSIONS: Our study identifies novel cardioprotective effects of Ac-SDKP in a model of cardiac irradiation. These protective effects are exerted by inhibiting inflammation, fibrosis, and reducing macrophage activation. This study shows a therapeutic potential of this endogenously released peptide to counteract radiation-induced cardiomyopathy.


Asunto(s)
Cardiomiopatías/prevención & control , Macrófagos/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Oligopéptidos/farmacología , Traumatismos por Radiación/prevención & control , Protectores contra Radiación/farmacología , Animales , Apoptosis/efectos de los fármacos , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Cardiotoxicidad , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Femenino , Fibrosis , Galectina 3/genética , Galectina 3/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Células RAW 264.7 , Traumatismos por Radiación/diagnóstico por imagen , Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/patología , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/metabolismo , Función Ventricular Izquierda/efectos de los fármacos
20.
Sci Rep ; 8(1): 6657, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29703991

RESUMEN

The recent development of mass cytometry has allowed simultaneous detection of 40 or more unique parameters from individual single cells. While similar to flow cytometry, which is based on detection of fluorophores, one key distinguishing feature of mass cytometry is the detection of atomic masses of lanthanides by mass spectrometry in a mass cytometer. Its superior mass resolution results in lack of signal overlap, thereby allowing multiparametric detection of molecular features in each single cell greater than that of flow cytometry, which is limited to 20 parameters. Unfortunately, most detection in mass cytometry relies on lanthanide-tagged antibodies, which is ideal to detect proteins, but not other types of molecular features. To further expand the repertoire of molecular features that are detectable by mass cytometry, we developed a lanthanide-chelated, azide-containing probe that allows click-chemistry mediated labeling of target molecules. Following incorporation of the thymidine analog 5-ethynyl-2'-deoxyuridine (EdU) during DNA synthesis in S-phase of the cell cycle, we demonstrate that the probe introduced here, tagged with Terbium-159 (159Tb), reacts via copper-catalyzed azide-alkyne Huisgen cycloaddition (click-chemistry) with Edu. Thus, detection of 159Tb makes it possible to measure DNA synthesis in single cells using mass cytometry. The approach introduced here shows similar sensitivity (true positive rate) to other methods used to measure DNA synthesis in single cells by mass cytometry and is compatible with the parallel antibody-based detection of other parameters in single cells. Due to its universal nature, the use of click-chemistry in mass cytometry expands the types of molecular targets that can be monitored by mass cytometry.


Asunto(s)
Química Clic/métodos , Técnicas Citológicas/métodos , Indicadores y Reactivos/síntesis química , Espectrometría de Masas/métodos , Análisis de la Célula Individual/métodos , Azidas/metabolismo , Quelantes/metabolismo , Células HeLa , Humanos , Elementos de la Serie de los Lantanoides/metabolismo , Coloración y Etiquetado/métodos , Terbio/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...