Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Physiol Genomics ; 56(7): 469-482, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38525531

RESUMEN

Single-cell technologies such as flow cytometry and single-cell RNA sequencing have allowed for comprehensive characterization of the kidney cellulome. However, there is a disparity in the various protocols for preparing kidney single-cell suspensions. We aimed to address this limitation by characterizing kidney cellular heterogeneity using three previously published single-cell preparation protocols. Single-cell suspensions were prepared from male and female C57BL/6 kidneys using the following kidney tissue dissociation protocols: a scRNAseq protocol (P1), a multi-tissue digestion kit from Miltenyi Biotec (P2), and a protocol established in our laboratory (P3). Following dissociation, flow cytometry was used to identify known major cell types including leukocytes (myeloid and lymphoid), vascular cells (smooth muscle and endothelial), nephron epithelial cells (intercalating, principal, proximal, and distal tubule cells), podocytes, and fibroblasts. Of the protocols tested, P2 yielded significantly less leukocytes and type B intercalating cells compared with the other techniques. P1 and P3 produced similar yields for most cell types; however, endothelial and myeloid-derived cells were significantly enriched using P1. Significant sex differences were detected in only two cell types: granulocytes (increased in males) and smooth muscle cells (increased in females). Future single-cell studies that aim to enrich specific kidney cell types may benefit from this comparative analysis.NEW & NOTEWORTHY This study is the first to evaluate published single-cell suspension preparation protocols and their ability to produce high-quality cellular yields from the mouse kidney. Three single-cell digestion protocols were compared and each produced significant differences in kidney cellular heterogeneity. These findings highlight the importance of the digestion protocol when using single-cell technologies. This study may help future single-cell science research by guiding researchers to choose protocols that enrich certain cell types of interest.


Asunto(s)
Riñón , Ratones Endogámicos C57BL , Análisis de la Célula Individual , Animales , Análisis de la Célula Individual/métodos , Femenino , Masculino , Ratones , Riñón/metabolismo , Riñón/citología , Citometría de Flujo/métodos , Células Endoteliales/metabolismo , Células Endoteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/citología
2.
Basic Res Cardiol ; 118(1): 11, 2023 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-36988733

RESUMEN

Coronary microvascular dysfunction (CMD) is associated with cardiac dysfunction and predictive of cardiac mortality in obesity, especially in females. Clinical data further support that CMD associates with development of heart failure with preserved ejection fraction and that mineralocorticoid receptor (MR) antagonism may be more efficacious in obese female, versus male, HFpEF patients. Accordingly, we examined the impact of smooth muscle cell (SMC)-specific MR deletion on obesity-associated coronary and cardiac diastolic dysfunction in female mice. Obesity was induced in female mice via western diet (WD) feeding alongside littermates fed standard diet. Global MR blockade with spironolactone prevented coronary and cardiac dysfunction in obese females and specific deletion of SMC-MR was sufficient to prevent obesity-associated coronary and cardiac diastolic dysfunction. Cardiac gene expression profiling suggested reduced cardiac inflammation in WD-fed mice with SMC-MR deletion independent of blood pressure, aortic stiffening, and cardiac hypertrophy. Further mechanistic studies utilizing single-cell RNA sequencing of non-cardiomyocyte cell populations revealed novel impacts of SMC-MR deletion on the cardiac cellulome in obese mice. Specifically, WD feeding induced inflammatory gene signatures in non-myocyte populations including B/T cells, macrophages, and endothelium as well as increased coronary VCAM-1 protein expression, independent of cardiac fibrosis, that was prevented by SMC-MR deletion. Further, SMC-MR deletion induced a basal reduction in cardiac mast cells and prevented WD-induced cardiac pro-inflammatory chemokine expression and leukocyte recruitment. These data reveal a central role for SMC-MR signaling in obesity-associated coronary and cardiac dysfunction, thus supporting the emerging paradigm of a vascular origin of cardiac dysfunction in obesity.


Asunto(s)
Cardiomiopatías , Insuficiencia Cardíaca , Masculino , Femenino , Ratones , Animales , Ratones Obesos , Insuficiencia Cardíaca/complicaciones , Multiómica , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Volumen Sistólico , Antagonistas de Receptores de Mineralocorticoides/farmacología , Obesidad/metabolismo
3.
Circulation ; 142(15): 1448-1463, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-32795101

RESUMEN

BACKGROUND: Cardiac fibrosis is a key antecedent to many types of cardiac dysfunction including heart failure. Physiological factors leading to cardiac fibrosis have been recognized for decades. However, the specific cellular and molecular mediators that drive cardiac fibrosis, and the relative effect of disparate cell populations on cardiac fibrosis, remain unclear. METHODS: We developed a novel cardiac single-cell transcriptomic strategy to characterize the cardiac cellulome, the network of cells that forms the heart. This method was used to profile the cardiac cellular ecosystem in response to 2 weeks of continuous administration of angiotensin II, a profibrotic stimulus that drives pathological cardiac remodeling. RESULTS: Our analysis provides a comprehensive map of the cardiac cellular landscape uncovering multiple cell populations that contribute to pathological remodeling of the extracellular matrix of the heart. Two phenotypically distinct fibroblast populations, Fibroblast-Cilp and Fibroblast-Thbs4, emerged after induction of tissue stress to promote fibrosis in the absence of smooth muscle actin-expressing myofibroblasts, a key profibrotic cell population. After angiotensin II treatment, Fibroblast-Cilp develops as the most abundant fibroblast subpopulation and the predominant fibrogenic cell type. Mapping intercellular communication networks within the heart, we identified key intercellular trophic relationships and shifts in cellular communication after angiotensin II treatment that promote the development of a profibrotic cellular microenvironment. Furthermore, the cellular responses to angiotensin II and the relative abundance of fibrogenic cells were sexually dimorphic. CONCLUSIONS: These results offer a valuable resource for exploring the cardiac cellular landscape in health and after chronic cardiovascular stress. These data provide insights into the cellular and molecular mechanisms that promote pathological remodeling of the mammalian heart, highlighting early transcriptional changes that precede chronic cardiac fibrosis.


Asunto(s)
Cardiomegalia/metabolismo , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Miocardio/metabolismo , Análisis de la Célula Individual , Estrés Fisiológico , Animales , Cardiomegalia/patología , Fibroblastos/patología , Fibrosis , Ratones , Miocardio/patología , Pirofosfatasas/metabolismo , Trombospondinas/metabolismo
4.
Cardiovasc Diabetol ; 20(1): 116, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34074290

RESUMEN

BACKGROUND: Diabetes is associated with a significantly elevated risk of cardiovascular disease and its specific pathophysiology remains unclear. Recent studies have changed our understanding of cardiac cellularity, with cellular changes accompanying diabetes yet to be examined in detail. This study aims to characterise the changes in the cardiac cellular landscape in murine diabetes to identify potential cellular protagonists in the diabetic heart. METHODS: Diabetes was induced in male FVB/N mice by low-dose streptozotocin and a high-fat diet for 26-weeks. Cardiac function was measured by echocardiography at endpoint. Flow cytometry was performed on cardiac ventricles as well as blood, spleen, and bone-marrow at endpoint from non-diabetic and diabetic mice. To validate flow cytometry results, immunofluorescence staining was conducted on left-ventricles of age-matched mice. RESULTS: Mice with diabetes exhibited hyperglycaemia and impaired glucose tolerance at endpoint. Echocardiography revealed reduced E:A and e':a' ratios in diabetic mice indicating diastolic dysfunction. Systolic function was not different between the experimental groups. Detailed examination of cardiac cellularity found resident mesenchymal cells (RMCs) were elevated as a result of diabetes, due to a marked increase in cardiac fibroblasts, while smooth muscle cells were reduced in proportion. Moreover, we found increased levels of Ly6Chi monocytes in both the heart and in the blood. Consistent with this, the proportion of bone-marrow haematopoietic stem cells were increased in diabetic mice. CONCLUSIONS: Murine diabetes results in distinct changes in cardiac cellularity. These changes-in particular increased levels of fibroblasts-offer a framework for understanding how cardiac cellularity changes in diabetes. The results also point to new cellular mechanisms in this context, which may further aid in development of pharmacotherapies to allay the progression of cardiomyopathy associated with diabetes.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Cardiomiopatías Diabéticas/etiología , Fibroblastos/patología , Miocardio/patología , Disfunción Ventricular Izquierda/etiología , Función Ventricular Izquierda , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/fisiopatología , Diástole , Dieta Alta en Grasa , Fibroblastos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Masculino , Ratones , Monocitos/metabolismo , Monocitos/patología , Miocardio/metabolismo , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Estreptozocina , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
5.
Biochem Soc Trans ; 48(6): 2483-2493, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33259583

RESUMEN

Single-cell transcriptomics enables inference of context-dependent phenotypes of individual cells and determination of cellular diversity of complex tissues. Cardiac fibrosis is a leading factor in the development of heart failure and a major cause of morbidity and mortality worldwide with no effective treatment. Single-cell RNA-sequencing (scRNA-seq) offers a promising new platform to identify new cellular and molecular protagonists that may drive cardiac fibrosis and development of heart failure. This review will summarize the application scRNA-seq for understanding cardiac fibrosis and development of heart failure. We will also discuss some key considerations in interpreting scRNA-seq data and some of its limitations.


Asunto(s)
Secuencia de Bases , Corazón/fisiología , Miocardio/metabolismo , Transcriptoma , Animales , Biología Computacional , Fibroblastos/metabolismo , Fibrosis/fisiopatología , Insuficiencia Cardíaca/fisiopatología , Homeostasis , Humanos , Ratones , Miofibroblastos/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual
6.
Semin Cell Dev Biol ; 61: 71-79, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27521522

RESUMEN

Identification of the key ingredients and essential processes required to achieve perfect tissue regeneration in humans has so far remained elusive. Injury in vertebrates induces an obligatory wound response that will precede or overlap any regeneration specific program or scarring outcome. This process shapes the cellular and molecular landscape of the tissue, influencing the success of endogenous repair pathways or for potential clinical intervention. The involvement of immune cells is also required for aspects of development extending beyond the initial inflammatory phase of wounding. It has now become clear from amphibian, fish and mammalian models of tissue injury that the type of immune response and the profile of immune cells attending the site of injury can act as the gatekeepers that determine wound repair quality. The heterogeneity among innate and adaptive immune cell populations, along with the developmental origin of these cells, form key ingredients affecting the potential for downstream repair and the suppression of fibrosis. Cell-to-cell interactions between immune cells, such as macrophages and T cells, with stem cells and mesenchymal cells are critically important for shaping this process and these exchanges, are in turn influenced by the type of injury, tissue location and developmental stage of the organism. Developmentally, mouse cardiac regeneration is restricted to early stages of postnatal life where the balance of innate to adaptive immune cells may be poised towards regeneration. In the injured adult mouse liver, specific macrophage subsets improve repair while other bone marrow derived cells can exacerbate injury. Other studies using genetically diverse mice have shown enhanced regeneration in certain strains, restricted to specific tissues. This enhanced repair is linked with expression of genes such as Insulin-like Growth Factor- 1 (IGF-1) and activin (Act 1), that both play important roles in shaping the immune system. Immune cells are now appreciated to have powerful influences on critical cell types required for regeneration success. The winning recipe for tissue regeneration is likely to be found ultimately by identifying the genetic elements and specific cell populations that limit or allow intrinsic potential. This will be essential for developing therapeutic strategies for tissue regeneration in humans.


Asunto(s)
Sistema Inmunológico/fisiología , Regeneración/fisiología , Animales , Evolución Biológica , Humanos , Inmunidad Celular , Inmunidad Innata , Cicatrización de Heridas
8.
Circ Res ; 118(3): 400-9, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26635390

RESUMEN

RATIONALE: Accurate knowledge of the cellular composition of the heart is essential to fully understand the changes that occur during pathogenesis and to devise strategies for tissue engineering and regeneration. OBJECTIVE: To examine the relative frequency of cardiac endothelial cells, hematopoietic-derived cells, and fibroblasts in the mouse and human heart. METHODS AND RESULTS: Using a combination of genetic tools and cellular markers, we examined the occurrence of the most prominent cell types in the adult mouse heart. Immunohistochemistry revealed that endothelial cells constitute >60%, hematopoietic-derived cells 5% to 10%, and fibroblasts <20% of the nonmyocytes in the heart. A refined cell isolation protocol and an improved flow cytometry approach provided an independent means of determining the relative abundance of nonmyocytes. High-dimensional analysis and unsupervised clustering of cell populations confirmed that endothelial cells are the most abundant cell population. Interestingly, fibroblast numbers are smaller than previously estimated, and 2 commonly assigned fibroblast markers, Sca-1 and CD90, under-represent fibroblast numbers. We also describe an alternative fibroblast surface marker that more accurately identifies the resident cardiac fibroblast population. CONCLUSIONS: This new perspective on the abundance of different cell types in the heart demonstrates that fibroblasts comprise a relatively minor population. By contrast, endothelial cells constitute the majority of noncardiomyocytes and are likely to play a greater role in physiological function and response to injury than previously appreciated.


Asunto(s)
Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Corazón , Células Madre Hematopoyéticas/metabolismo , Adulto , Animales , Biomarcadores/metabolismo , Recuento de Células , Diferenciación Celular , Linaje de la Célula , Separación Celular/métodos , Femenino , Citometría de Flujo , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Inmunohistoquímica , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Fenotipo
9.
Adv Exp Med Biol ; 1003: 105-118, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28667556

RESUMEN

Macrophages are principally recognized as an important cell type for removal of tissue debris and as sentinels for tissue damage and foreign antigens. However, macrophages also participate in a diverse range of biological processes including angiogenesis, fibrosis, immune modulation, cell survival, and stem cell mobilization. Cardiac tissue macrophages (cTMs) are a heterogeneous population of phagocytic cells with distinct ontogenetic, phenotypic, and functional characteristics. While our understanding of cTMs has increased substantially over the last 5 years, large gaps in our knowledge regarding the cell biology of cTMs exist, in particular, the development of their unique phenotype and their roles in cardiac homeostasis and tissue stress. This review aims to discuss the current knowledge regarding cTMs and identify key questions that must be addressed to gain a better understanding of the role of cTMs in tissue development, homeostasis, and disease.


Asunto(s)
Enfermedades Cardiovasculares/inmunología , Macrófagos/inmunología , Miocardio/inmunología , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Diferenciación Celular , Linaje de la Célula , Homeostasis , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Miocardio/metabolismo , Miocardio/patología , Fenotipo , Transducción de Señal
11.
Circ Res ; 114(9): 1422-34, 2014 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-24650916

RESUMEN

RATIONALE: Cardiac fibroblasts are critical to proper heart function through multiple interactions with the myocardial compartment, but appreciation of their contribution has suffered from incomplete characterization and lack of cell-specific markers. OBJECTIVE: To generate an unbiased comparative gene expression profile of the cardiac fibroblast pool, identify and characterize the role of key genes in cardiac fibroblast function, and determine their contribution to myocardial development and regeneration. METHODS AND RESULTS: High-throughput cell surface and intracellular profiling of cardiac and tail fibroblasts identified canonical mesenchymal stem cell and a surprising number of cardiogenic genes, some expressed at higher levels than in whole heart. While genetically marked fibroblasts contributed heterogeneously to interstitial but not cardiomyocyte compartments in infarcted hearts, fibroblast-restricted depletion of one highly expressed cardiogenic marker, T-box 20, caused marked myocardial dysmorphology and perturbations in scar formation on myocardial infarction. CONCLUSIONS: The surprising transcriptional identity of cardiac fibroblasts, the adoption of cardiogenic gene programs, and direct contribution to cardiac development and repair provoke alternative interpretations for studies on more specialized cardiac progenitors, offering a novel perspective for reinterpreting cardiac regenerative therapies.


Asunto(s)
Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/genética , Miocardio/metabolismo , Regeneración/genética , Animales , Biomarcadores/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/patología , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN no Traducido/genética , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética
12.
Mol Cell Neurosci ; 68: 82-91, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25936601

RESUMEN

Spinal cord injury (SCI) frequently leads to a permanent functional impairment as a result of the initial injury followed by secondary injury mechanism, which is characterised by increased inflammation, glial scarring and neuronal cell death. Finding drugs that may reduce inflammatory cell invasion and activation to reduce glial scarring and increase neuronal survival is of major importance for improving the outcome after SCI. In the present study, we examined the effect of rapamycin, an mTORC1 inhibitor and an inducer of autophagy, on recovery from spinal cord injury. Autophagy, a process that facilitates the degradation of cytoplasmic proteins, is also important for maintenance of neuronal homeostasis and plays a major role in neurodegeneration after neurotrauma. We examined rapamycin effects on the inflammatory response, glial scar formation, neuronal survival and regeneration in vivo using spinal cord hemisection model in mice, and in vitro using primary cortical neurons and human astrocytes. We show that a single injection of rapamycin, inhibited p62/SQSTM1, a marker of autophagy, inhibited mTORC1 downstream effector p70S6K, reduced macrophage/neutrophil infiltration into the lesion site, microglia activation and secretion of TNFα. Rapamycin inhibited astrocyte proliferation and reduced the number of GFAP expressing cells at the lesion site. Finally, it increased neuronal survival and axonogenesis towards the lesion site. Our study shows that rapamycin treatment increased significantly p-Akt levels at the lesion site following SCI. Similarly, rapamycin treatment of neurons and astrocytes induced p-Akt elevation under stress conditions. Together, these findings indicate that rapamycin is a promising candidate for treatment of acute SCI condition and may be a useful therapeutic agent.


Asunto(s)
Inmunosupresores/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/etiología , Sirolimus/uso terapéutico , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/patología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Antígeno CD11b/metabolismo , Recuento de Células , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Proteína 3 Similar a ELAV/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Ratas , Factores de Tiempo
13.
Proc Natl Acad Sci U S A ; 110(23): 9415-20, 2013 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-23690624

RESUMEN

The failure to replace damaged body parts in adult mammals results from a muted growth response and fibrotic scarring. Although infiltrating immune cells play a major role in determining the variable outcome of mammalian wound repair, little is known about the modulation of immune cell signaling in efficiently regenerating species such as the salamander, which can regrow complete body structures as adults. Here we present a comprehensive analysis of immune signaling during limb regeneration in axolotl, an aquatic salamander, and reveal a temporally defined requirement for macrophage infiltration in the regenerative process. Although many features of mammalian cytokine/chemokine signaling are retained in the axolotl, they are more dynamically deployed, with simultaneous induction of inflammatory and anti-inflammatory markers within the first 24 h after limb amputation. Systemic macrophage depletion during this period resulted in wound closure but permanent failure of limb regeneration, associated with extensive fibrosis and disregulation of extracellular matrix component gene expression. Full limb regenerative capacity of failed stumps was restored by reamputation once endogenous macrophage populations had been replenished. Promotion of a regeneration-permissive environment by identification of macrophage-derived therapeutic molecules may therefore aid in the regeneration of damaged body parts in adult mammals.


Asunto(s)
Ambystoma mexicanum/fisiología , Extremidades/fisiología , Regulación de la Expresión Génica/fisiología , Macrófagos/fisiología , Regeneración/fisiología , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Citocinas/inmunología , Matriz Extracelular/metabolismo , Citometría de Flujo , Fluorescencia , Técnicas Histológicas , Inmunohistoquímica , Macrófagos/inmunología , Células Mieloides/inmunología , Fagocitosis/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/inmunología , Cicatrización de Heridas/fisiología
15.
Hypertension ; 81(4): 738-751, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38318714

RESUMEN

Aortic diseases such as atherosclerosis, aortic aneurysms, and aortic stiffening are significant complications that can have significant impact on end-stage cardiovascular disease. With limited pharmacological therapeutic strategies that target the structural changes in the aorta, surgical intervention remains the only option for some patients with these diseases. Although there have been significant contributions to our understanding of the cellular architecture of the diseased aorta, particularly in the context of atherosclerosis, furthering our insight into the cellular drivers of disease is required. The major cell types of the aorta are well defined; however, the advent of single-cell RNA sequencing provides unrivaled insights into the cellular heterogeneity of each aortic cell type and the inferred biological processes associated with each cell in health and disease. This review discusses previous concepts that have now been enhanced with recent advances made by single-cell RNA sequencing with a focus on aortic cellular heterogeneity.


Asunto(s)
Enfermedades de la Aorta , Aterosclerosis , Humanos , ARN , Aorta/metabolismo , Enfermedades de la Aorta/genética , Perfilación de la Expresión Génica , Aterosclerosis/genética , Aterosclerosis/metabolismo
16.
iScience ; 26(10): 107759, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37736052

RESUMEN

Diabetes is associated with a significantly elevated risk of heart failure. However, despite extensive efforts to characterize the phenotype of the diabetic heart, the molecular and cellular protagonists that underpin cardiac pathological remodeling in diabetes remain unclear, with a notable paucity of data regarding the impact of diabetes on non-myocytes within the heart. Here we aimed to define key differences in cardiac non-myocytes between spontaneously type-2 diabetic (db/db) and healthy control (db/h) mouse hearts. Single-cell transcriptomic analysis revealed a concerted diabetes-induced cellular response contributing to cardiac remodeling. These included cell-specific activation of gene programs relating to fibroblast hyperplasia and cell migration, and dysregulation of pathways involving vascular homeostasis and protein folding. This work offers a new perspective for understanding the cellular mediators of diabetes-induced cardiac pathology, and pathways that may be targeted to address the cardiac complications associated with diabetes.

17.
Chromosoma ; 119(2): 167-76, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19904548

RESUMEN

Chromosomal and telomeric reprogramming was assessed in intraspecies hybrids obtained by fusion of embryonic stem (ES) cells and mouse embryonic fibroblasts. Evaluation of the ploidy of ES-somatic hybrids revealed that 21 of 59 clones had a tetraploid DNA profile while the remaining clones showed deviations from the expected profile of fusion between two diploid cells. Microsatellite polymerase chain reaction analysis of four of these clones demonstrated no random loss of somatic chromosome pairs in the ES-somatic cell hybrids. Pluripotential of ES-somatic hybrids was assessed by gene expression analysis, antibody staining for Oct4 and SSEA-1 and teratoma formation containing derivatives of the three germ layers. Reprogramming of telomeric maintenance was observed with ES-somatic hybrids showing high telomerase activity and increased telomere lengths. However, we detected no significant increase in the expression of the three critical telomerase subunits: telomerase reverse transcriptase (TERT), telomerase RNA component (TERC), and dyskerin. This indicates that activation of telomerase and telomere maintenance is not reliant on changes in gene expression of TERT, TERC, and dyskerin following ES-somatic cell fusion or sister chromatid recombination and may arise through elimination of negative regulation of telomerase activity. This is the first demonstration of telomere lengthening following cell fusion and offers a new model for studying and identifying new regulators of telomere maintenance.


Asunto(s)
Células Madre Embrionarias/citología , Fibroblastos/citología , Telómero/genética , Animales , Técnicas de Cultivo de Célula , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Fusión Celular , Línea Celular , Segregación Cromosómica , Células Madre Embrionarias/fisiología , Fibroblastos/fisiología , Regulación de la Expresión Génica , Células Híbridas , Antígeno Lewis X/metabolismo , Ratones , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Ploidias , ARN/biosíntesis , ARN/genética , Telomerasa/biosíntesis , Telomerasa/genética
18.
Cardiovasc Res ; 117(10): 2252-2262, 2021 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-32941598

RESUMEN

AIMS: Sex differences have been consistently identified in cardiac physiology and incidence of cardiac disease. However, the underlying biological causes for the differences remain unclear. We sought to characterize the cardiac non-myocyte cellular landscape in female and male hearts to determine whether cellular proportion of the heart is sex-dependent and whether endocrine factors modulate the cardiac cell proportions. METHODS AND RESULTS: Utilizing high-dimensional flow cytometry and immunofluorescence imaging, we found significant sex-specific differences in cellular composition of the heart in adult and juvenile mice, that develops postnatally. Removal of systemic gonadal hormones by gonadectomy results in rapid sex-specific changes in cardiac non-myocyte cellular proportions including alteration in resident mesenchymal cell and leucocyte populations, indicating gonadal hormones and their downstream targets regulate cardiac cellular composition. The ectopic reintroduction of oestrogen and testosterone to female and male mice, respectively, reverses many of these gonadectomy-induced compositional changes. CONCLUSION: This work shows that the constituent cell types of the mouse heart are hormone-dependent and that the cardiac cellular landscapes are distinct in females and males, remain plastic, and can be rapidly modulated by endocrine factors. These observations have implications for strategies aiming to therapeutically alter cardiac cellular heterogeneity and underscore the importance of considering biological sex for studies examining cardiac physiology and stress responses.


Asunto(s)
Estradiol/metabolismo , Miocardio/metabolismo , Testosterona/metabolismo , Factores de Edad , Animales , Separación Celular , Estradiol/farmacología , Terapia de Reemplazo de Estrógeno , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Masculino , Ratones Endogámicos C57BL , Miocardio/citología , Orquiectomía , Ovariectomía , RNA-Seq , Caracteres Sexuales , Análisis de la Célula Individual , Testosterona/farmacología , Transcriptoma
19.
STAR Protoc ; 2(4): 100866, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34647038

RESUMEN

This protocol features parallel isolation of myocytes and non-myocytes from murine hearts. It was designed with considerations for (1) time required to extract cardiac cells, (2) cell viability, and (3) protocol scalability. Here, a peristaltic pump and 3D-printed elements are combined to perfuse the heart with enzymes to dissociate cells. Myocytes and non-myocytes extracted using this protocol are separated by centrifugation and/or fluorescence-activated cell sorting for use in downstream applications including single-cell omics or other bio-molecular analyses. For complete details on the use and execution of this protocol, please refer to McLellan et al. (2020).


Asunto(s)
Separación Celular/métodos , Miocardio/citología , Miocitos Cardíacos , Análisis de la Célula Individual/métodos , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Genómica , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/clasificación , Miocitos Cardíacos/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA