Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Stem Cells ; 40(10): 932-948, 2022 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-35896368

RESUMEN

Adipose-derived cells (ADCs) from white adipose tissue are promising stem cell candidates because of their large regenerative reserves and the potential for cardiac regeneration. However, given the heterogeneity of ADC and its unsolved mechanisms of cardiac acquisition, ADC-cardiac transition efficiency remains low. In this study, we explored the heterogeneity of ADCs and the cellular kinetics of 39,432 single-cell transcriptomes along the leukemia inhibitory factor (LIF)-induced ADC-cardiac transition. We identified distinct ADC subpopulations that reacted differentially to LIF when entering the cardiomyogenic program, further demonstrating that ADC-myogenesis is time-dependent and initiates from transient changes in nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. At later stages, pseudotime analysis of ADCs navigated a trajectory with 2 branches corresponding to activated myofibroblast or cardiomyocyte-like cells. Our findings offer a high-resolution dissection of ADC heterogeneity and cell fate during ADC-cardiac transition, thus providing new insights into potential cardiac stem cells.


Asunto(s)
Miocitos Cardíacos , Factor 2 Relacionado con NF-E2 , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/farmacología , RNA-Seq , Diferenciación Celular/genética
2.
Sci Rep ; 12(1): 5614, 2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35379860

RESUMEN

Multipotent cells derived from white adipose tissue have been shown to differentiate into multiple lineages including neurogenic lineages. However, the high innervation of brown adipose tissue by the sympathetic nervous system suggest it might be a better source of neural precursor cells. To investigate potential differences between white and brown progenitors, we cultured white and brown dedifferentiated fat (wDFAT and brDFAT) cells from mouse and human adipose tissue and compared marker expression of neural precursors, and neuronal and glial cells, using fluorescence-activated cell sorting, bright-field imaging, immunofluorescence, and RNA analysis by qPCR. The results showed that both wDFAT and brDFAT cells had the capacity to generate neuronal and glial-like cells under neurogenic conditions. However, the brDFAT cells exhibited enhanced propensity for neurogenic differentiation. The neurogenic cells were at least in part derived from Adiponectin-expressing cells. TdTomato-expressing cells derived from Adiponectin (Adipoq) Cre ERT2 -tdTomato flox/flox mice gave rise to individual cells and cell clusters with neurogenic characteristics. Moreover, human brDFAT cells demonstrated a similar ability to undergo neurogenic differentiation after treatment with neurogenic medium, as assessed by immunofluorescence and qPCR. Together, our results support that brDFAT cells have ability to undergo neurogenic differentiation.


Asunto(s)
Tejido Adiposo Pardo , Células-Madre Neurales , Tejido Adiposo Blanco , Animales , Diferenciación Celular/fisiología , Ratones , Neurogénesis
3.
Circ Res ; 127(10): 1288-1305, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-32854559

RESUMEN

RATIONALE: The BMPs (bone morphogenetic proteins) are essential morphogens in angiogenesis and vascular development. Disruption of BMP signaling can trigger cardiovascular diseases, such as arteriovenous malformations. OBJECTIVE: A computational model predicted that BMP4 and BMP9 and their inhibitors MGP (matrix gamma-carboxyglutamic acid [Gla] protein) and CV2 (crossveinless-2) would form a regulatory system consisting of negative feedback loops with time delays and that BMP9 would trigger oscillatory expression of the 2 inhibitors. The goal was to investigate this regulatory system in endothelial differentiation and vascular growth. METHODS AND RESULTS: Oscillations in the expression of MGP and CV2 were detected in endothelial cells in vitro, using quantitative real-time polymerase chain reaction and immunoblotting. These organized temporally downstream BMP-related activities, including expression of stalk-cell markers and cell proliferation, consistent with an integral role of BMP9 in vessel maturation. In vivo, the inhibitors were located in distinct zones in relation to the front of the expanding retinal network, as determined by immunofluorescence. Time-dependent changes of the CV2 location in the retina and the existence of an endothelial population with signs of oscillatory MGP expression in developing vasculature supported the in vitro findings. Loss of MGP or its BMP4-binding capacity disrupted the retinal vasculature, resulting in poorly formed networks, especially in the venous drainage areas, and arteriovenous malformations as determined by increased cell coverage and functional testing. CONCLUSIONS: Our results suggest a previously unknown mechanism of temporal orchestration of BMP4 and BMP9 activities that utilize the tandem actions of the extracellular antagonists MGP and CV2. Disruption of this mechanism may contribute to vascular malformations and disease.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Modelos Cardiovasculares , Neovascularización Fisiológica , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Proteínas Morfogenéticas Óseas/genética , Humanos , Proteína Gla de la Matriz
4.
Mol Metab ; 25: 50-63, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31027994

RESUMEN

OBJECTIVE: Obesity has increased to pandemic levels and enhanced understanding of adipose regulation is required for new treatment strategies. Although bone morphogenetic proteins (BMPs) influence adipogenesis, the effect of BMP antagonists such as Noggin is largely unknown. The aim of the study was to define the role of Noggin, an extracellular BMP inhibitor, in adipogenesis. METHODS: We generated adipose-derived progenitor cells and a mouse model with adipocyte-specific Noggin deletion using the AdiponectinCre transgenic mouse, and determined the adipose phenotype of Noggin-deficiency. RESULTS: Our studies showed that Noggin is expressed in progenitor cells but declines in adipocytes, possibly allowing for lipid accumulation. Correspondingly, adipocyte-specific Noggin deletion in vivo promoted age-related obesity in both genders with no change in food intake. Although the loss of Noggin caused white adipose tissue hypertrophy, and whitening and impaired function in brown adipose tissue in both genders, there were clear gender differences with the females being most affected. The females had suppressed expression of brown adipose markers and thermogenic genes including peroxisome proliferator activated receptor gamma coactivator 1 alpha (PGC1alpha) and uncoupling protein 1 (UCP1) as well as genes associated with adipogenesis and lipid metabolism. The males, on the other hand, had early changes in a few BAT markers and thermogenic genes, but the main changes were in the genes associated with adipogenesis and lipid metabolism. Further characterization revealed that both genders had reductions in VO2, VCO2, and RER, whereas females also had reduced heat production. Noggin was also reduced in diet-induced obesity in inbred mice consistent with the obesity phenotype of the Noggin-deficient mice. CONCLUSIONS: BMP signaling regulates female and male adipogenesis through different metabolic pathways. Modulation of adipose tissue metabolism by select BMP antagonists may be a strategy for long-term regulation of age-related weight gain and obesity.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Portadoras/metabolismo , Obesidad/metabolismo , Adipocitos/patología , Adipogénesis/genética , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Proteínas Morfogenéticas Óseas/efectos de los fármacos , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Ingestión de Alimentos , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Estudios de Asociación Genética , Producto de la Acumulación de Lípidos , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Obesos , Ratones Transgénicos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Transducción de Señal/genética , Termogénesis/genética , Termogénesis/fisiología , Transcriptoma , Proteína Desacopladora 1/genética
5.
Obes Rev ; 20(5): 648-658, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30609449

RESUMEN

The bone morphogenetic proteins (BMPs) belong to the same superfamily as related to transforming growth factor ß (TGFß), growth and differentiation factors (GDFs), and activins. They were initially described as inducers of bone formation but are now known to be involved in morphogenetic activities and cell differentiation throughout the body, including the development of adipose tissue and adipogenic differentiation. BMP4 and BMP7 are the most studied BMPs in adipose tissue, with major roles in white adipogenesis and brown adipogenesis, respectively, but other BMPs such as BMP2, BMP6, and BMP8b as well as some inhibitors and modulators have been shown to also affect adipogenesis. It has become ever more important to understand adipose regulation, including the BMP pathways, in light of the strong links between obesity and metabolic and cardiovascular disease. In this review, we summarize the available information on BMP signaling in adipose tissue using preferentially articles that have appeared in the last decade, which together demonstrate the importance of BMP signaling in adipose biology.


Asunto(s)
Adipogénesis/fisiología , Tejido Adiposo/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Transducción de Señal/fisiología , Animales , Humanos
6.
Artículo en Inglés | MEDLINE | ID: mdl-33954280

RESUMEN

Multipotent cells derived from white mature adipocytes, referred to as dedifferentiated fat (DFAT) cells have the capacity differentiate into endothelial cells. The objective of this study was to modify the isolation method for DFAT cells in order to optimize the endothelial lineage potential. The adipocytes were preincubated for 24 hours, washed, and then incubated for 5 days to allow the generated DFAT cells to remain in proximity to the adipocytes while the cells aggregated into cell clusters. The DFAT cells rapidly differentiated into adipocytes after which endothelial-like cells (ECs) emerged and formed tube-like structure closely associated with the newly differentiated adipocytes. The lipid-filled cells then gradually disappeared whereas the network of tube structure expanded over the course of 3 weeks. ECs accounted for 35-45% of the cells derived from the DFAT cells, as assessed by qPCR, immunofluorescence and fluorescence-activated cell sorting. The DFAT cell-derived ECs could also be further enriched by magnetic sorting, thereby serving as a mouse cell line for further research.

7.
J Cell Physiol ; 233(3): 1812-1822, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28464239

RESUMEN

Bone morphogenetic protein (BMP) 10, a cardiac-restricted BMP family member, is essential in cardiomyogenesis, especially during trabeculation. Crossveinless-2 (CV2, also known as BMP endothelial cell precursor derived regulator [BMPER]) is a BMP-binding protein that modulates the activity of several BMPs. The objective of this study was to examine the combined effects of BMP10 and CV2 on cardiomyocyte differentiation using mouse dedifferentiated fat (mDFAT) cells, which spontaneously differentiate into cardiomyocyte-like cells, as a model. Our results revealed that CV2 binds directly to BMP10, as determined by co-immunoprecipitation, and inhibits BMP10 from initiating SMAD signaling, as determined by luciferase reporter gene assays. BMP10 treatment induced mDFAT cell proliferation, whereas CV2 modulated the BMP10-induced proliferation. Differentiation of cardiomyocyte-like cells proceeded in a reproducible fashion in mDFAT cells, starting with small round Nkx2.5-positive progenitor cells that progressively formed myotubes of increasing length that assembled into beating colonies and stained strongly for Troponin I and sarcomeric alpha-actinin. BMP10 enhanced proliferation of the small progenitor cells, thereby securing sufficient numbers to support formation of myotubes. CV2, on the other hand, enhanced formation and maturation of large myotubes and myotube-colonies and was expressed by endothelial-like cells in the mDFAT cultures. Thus BMP10 and CV2 have important roles in coordinating cardiomyogenesis in progenitor cells.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Portadoras/metabolismo , Diferenciación Celular/fisiología , Miocitos Cardíacos/citología , Células Madre/citología , Actinina/metabolismo , Adipocitos/citología , Animales , Proliferación Celular , Células Cultivadas , Proteína Homeótica Nkx-2.5/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología , Proteínas Smad/metabolismo , Troponina I/metabolismo
8.
J Cell Biol ; 216(10): 3369-3385, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28838957

RESUMEN

The vascular endothelium is critical for induction of appropriate lineage differentiation in organogenesis. In this study, we report that dysfunctional pulmonary endothelium, resulting from the loss of matrix Gla protein (MGP), causes ectopic hepatic differentiation in the pulmonary epithelium. We demonstrate uncontrolled induction of the hepatic growth factor (HGF) caused by dysregulated cross talk between pulmonary endothelium and epithelium in Mgp-null lungs. Elevated HGF induced hepatocyte nuclear factor 4 α (Hnf4a), which competed with NK2 homeobox 1 (Nkx2.1) for binding to forkhead box A2 (Foxa2) to drive hepatic differentiation in Mgp-null airway progenitor cells. Limiting endothelial HGF reduced Hnf4a, abolished interference of Hnf4a with Foxa2, and reduced hepatic differentiation in Mgp-null lungs. Together, our results suggest that endothelial-epithelial interactions, maintained by MGP, are essential in pulmonary cell differentiation.


Asunto(s)
Comunicación Celular , Diferenciación Celular , Endotelio Vascular/metabolismo , Células Epiteliales/metabolismo , Mucosa Respiratoria/metabolismo , Animales , Endotelio Vascular/citología , Células Epiteliales/citología , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Ratones , Ratones Noqueados , Proteínas/genética , Proteínas/metabolismo , Mucosa Respiratoria/citología
9.
Stem Cells Int ; 2016: 3924858, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27822228

RESUMEN

Mesenchymal stem cells (MSCs) have attracted the attention of researchers and clinicians for their ability to differentiate into a number of cell types, participate in tissue regeneration, and repair the damaged tissues by producing various growth factors and cytokines, as well as their unique immunoprivilege in alloreactive hosts. The immunomodulatory functions of exogenous MSCs have been widely investigated in immune-mediated inflammatory diseases and transplantation research. However, a harsh environment at the site of tissue injury/inflammation with insufficient oxygen supply, abundance of reactive oxygen species, and presence of other harmful molecules that damage the adoptively transferred cells collectively lead to low survival and engraftment of the transferred cells. Preconditioning of MSCs ex vivo by hypoxia, inflammatory stimulus, or other factors/conditions prior to their use in therapy is an adaptive strategy that prepares MSCs to survive in the harsh environment and to enhance their regulatory function of the local immune responses. This review focuses on a number of approaches in preconditioning human MSCs with the goal of augmenting their capacity to regulate both innate and adaptive immune responses.

10.
World J Stem Cells ; 7(10): 1202-14, 2015 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-26640620

RESUMEN

The identification of an ideal cell source for tissue regeneration remains a challenge in the stem cell field. The ability of progeny cells to differentiate into other cell types is important for the processes of tissue reconstruction and tissue engineering and has clinical, biochemical or molecular implications. The adaptation of stem cells from adipose tissue for use in regenerative medicine has created a new role for adipocytes. Mature adipocytes can easily be isolated from adipose cell suspensions and allowed to dedifferentiate into lipid-free multipotent cells, referred to as dedifferentiated fat (DFAT) cells. Compared to other adult stem cells, the DFAT cells have unique advantages in their abundance, ease of isolation and homogeneity. Under proper condition in vitro and in vivo, the DFAT cells have exhibited adipogenic, osteogenic, chondrogenic, cardiomyogenc, angiogenic, myogenic, and neurogenic potentials. In this review, we first discuss the phenomena of dedifferentiation and transdifferentiation of cells, and then dedifferentiation of adipocytes in particular. Understanding the dedifferentiation process itself may contribute to our knowledge of normal growth processes, as well as mechanisms of disease. Second, we highlight new developments in DFAT cell culture and summarize the current understanding of DFAT cell properties. The unique features of DFAT cells are promising for clinical applications such as tissue regeneration.

11.
Circ Res ; 117(9): 758-69, 2015 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-26265629

RESUMEN

RATIONALE: Endothelial cells have the ability to undergo endothelial-mesenchymal transitions (EndMTs), by which they acquire a mesenchymal phenotype and stem cell-like characteristics. We previously found that EndMTs occurred in the endothelium deficient in matrix γ-carboxyglutamic acid protein enabling endothelial cells to contribute cells to vascular calcification. However, the mechanism responsible for initiating EndMTs is not fully understood. OBJECTIVE: To determine the role of specific serine proteases and sex determining region Y-box 2 (Sox2) in the initiation of EndMTs. METHODS AND RESULTS: In this study, we used in vivo and in vitro models of vascular calcification to demonstrate that serine proteases and Sox2 are essential for the initiation of EndMTs in matrix γ-carboxyglutamic acid protein-deficient endothelium. We showed that expression of a group of specific serine proteases was highly induced in endothelial cells at sites of vascular calcification in Mgp null aortas. Treatment with serine protease inhibitors decreased both stem cell marker expression and vascular calcification. In human aortic endothelial cells, this group of serine proteases also induced EndMTs, and the activation of proteases was mediated by Sox2. Knockdown of the serine proteases or Sox2 diminished EndMTs and calcification. Endothelial-specific deletion of Sox2 decreased expression of stem cell markers and aortic calcification in matrix γ-carboxyglutamic acid protein-deficient mice. CONCLUSIONS: Our results suggest that Sox2-mediated activation of specific serine proteases is essential for initiating EndMTs, and thus, may provide new therapeutic targets for treating vascular calcification.


Asunto(s)
Calcinosis , Endotelio Vascular/metabolismo , Mesodermo/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Células Cultivadas , Endotelio Vascular/patología , Endotelio Vascular/ultraestructura , Activación Enzimática , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Immunoblotting , Calicreínas/genética , Calicreínas/metabolismo , Mesodermo/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Elastasa Pancreática/genética , Elastasa Pancreática/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Serina Endopeptidasas/genética , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo , Proteína Gla de la Matriz
12.
J Cell Physiol ; 230(11): 2821-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25854185

RESUMEN

Diabetes mellitus affects the adipose tissue and mesenchymal stem cells derived from the adipose stroma and other tissues. Previous reports suggest that bone morphogenetic protein 4 (BMP4) is involved in diabetic complications, at the same time playing an important role in the maintenance of stem cells. In this study, we used rats transgenic for human islet amyloid polypeptide (HIP rats), a model of type 2 diabetes, to study the effect of diabetes on adipocyte-derived stem cells, referred to as dedifferentiated fat (DFAT) cells. Our results show that BMP4 expression in inguinal adipose tissue is significantly increased in HIP rats compared to controls, whereas matrix Gla protein (MGP), an inhibitor of BMP4 is decreased as determined by quantitative PCR, and immunofluorescence. In addition, adipose vascularity and expression of multiple endothelial cell markers was increased in the diabetic tissue, visualized by immunofluorescence for endothelial markers. The endothelial markers co-localized with the enhanced BMP4 expression, suggesting that vascular cells play a role BMP4 induction. The DFAT cells are multipotent stem cells derived from white mature adipocytes that undergo endothelial and adipogenic differentiation. DFAT cells prepared from the inguinal adipose tissue in HIP rats exhibited enhanced proliferative capacity compared to wild type. In addition, their ability to undergo both endothelial cell and adipogenic lineage differentiation was enhanced, as well as their response to BMP4, as assessed by lineage marker expression. We conclude that the DFAT cells are affected by diabetic changes and may contribute to the adipose dysfunction in diabetes.


Asunto(s)
Proteína Morfogenética Ósea 4/biosíntesis , Diferenciación Celular/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , Tejido Adiposo , Animales , Animales Modificados Genéticamente , Proteína Morfogenética Ósea 4/genética , Proteínas de Unión al Calcio/biosíntesis , Proteínas de Unión al Calcio/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/patología , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas de la Matriz Extracelular/genética , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/biosíntesis , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratas , Proteína Gla de la Matriz
13.
FEBS Open Bio ; 5: 257-63, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25893161

RESUMEN

Mutations in ABCC6 (ATP-binding cassette, subfamily C, member 6), an orphan transporter expressed in the liver, are the cause of pseudoxanthoma elasticum. Since ABCC6 was reported to affect matrix Gla protein (MGP), an inhibitor of bone morphogenetic proteins (BMPs), we studied BMP signaling and expression in various tissues of mice with and without functional ABCC. Enhanced BMP signaling was found in all examined tissues in the absence of ABCC6. Despite this, the expression of particular BMP proteins varied widely between tissues. Interestingly, the expression of most BMP proteins in the liver moved in the opposite direction to the same BMP proteins in kidneys in response to ABCC6 alterations. Thus, ABCC6 deficiency stimulates BMP signaling by acting on the expression of multiple BMPs.

14.
Stem Cells Transl Med ; 3(2): 161-71, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24396033

RESUMEN

White mature adipocytes give rise to so-called dedifferentiated fat (DFAT) cells that spontaneously undergo multilineage differentiation. In this study, we defined stem cell characteristics of DFAT cells as they are generated from adipocytes and the relationship between these characteristics and lineage differentiation. Both mouse and human DFAT cells, prepared from adipose tissue and lipoaspirate, respectively, showed evidence of pluripotency, with a maximum 5-7 days after adipocyte isolation. The DFAT cells spontaneously formed clusters in culture, which transiently expressed multiple stem cell markers, including stage-specific embryonic antigens, and Sca-1 (mouse) and CD105 (human), as determined by real-time polymerase chain reaction, fluorescence-activated cell sorting, and immunostaining. As the stem cell markers decreased, markers characteristic of the three germ layers and specific lineage differentiation, such as α-fetoprotein (endoderm, hepatic), Neurofilament-66 (ectoderm, neurogenic), and Troponin I (mesoderm, cardiomyogenic), increased. However, no teratoma formation was detected after injection in immunodeficient mice. A novel modification of the adipocyte isolation aimed at ensuring the initial purity of the adipocytes and avoiding ceiling culture allowed isolation of DFAT cells with pluripotent characteristics. Thus, the adipocyte-derived DFAT cells represent a plastic stem cell population that is highly responsive to changes in culture conditions and may benefit cell-based therapies.


Asunto(s)
Adipocitos Blancos/citología , Desdiferenciación Celular/fisiología , Infarto del Miocardio/patología , Miocardio/citología , Células Madre Pluripotentes/citología , Teratoma/patología , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Teratoma/etiología
15.
Proc Natl Acad Sci U S A ; 110(47): 19071-6, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24191040

RESUMEN

Cerebral arteriovenous malformations (AVMs) are common vascular malformations, which may result in hemorrhagic strokes and neurological deficits. Bone morphogenetic protein (BMP) and Notch signaling are both involved in the development of cerebral AVMs, but the cross-talk between the two signaling pathways is poorly understood. Here, we show that deficiency of matrix Gla protein (MGP), a BMP inhibitor, causes induction of Notch ligands, dysregulation of endothelial differentiation, and the development of cerebral AVMs in MGP null (Mgp(-/-)) mice. Increased BMP activity due to the lack of MGP induces expression of the activin receptor-like kinase 1, a BMP type I receptor, in cerebrovascular endothelium. Subsequent activation of activin receptor-like kinase 1 enhances expression of Notch ligands Jagged 1 and 2, which increases Notch activity and alters the expression of Ephrin B2 and Ephrin receptor B4, arterial and venous endothelial markers, respectively. Reducing the expression of Jagged 1 and 2 in the Mgp(-/-) mice by crossing them with Jagged 1 or 2 deficient mice reduces Notch activity, normalizes endothelial differentiation, and prevents cerebral AVMs, but not pulmonary or renal AVMs. Our results suggest that Notch signaling mediates and can modulate changes in BMP signaling that lead to cerebral AVMs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/metabolismo , Proteínas de la Matriz Extracelular/deficiencia , Guanilato-Quinasas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Malformaciones Arteriovenosas Intracraneales/etiología , Malformaciones Arteriovenosas Intracraneales/prevención & control , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Análisis de Varianza , Animales , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proteínas de Unión al Calcio/genética , Diferenciación Celular/fisiología , Células Endoteliales/fisiología , Efrina-B2/metabolismo , Proteínas de la Matriz Extracelular/genética , Eliminación de Gen , Immunoblotting , Proteína Jagged-1 , Proteína Jagged-2 , Ratones , Ratones Noqueados , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de la Familia Eph/metabolismo , Proteínas Serrate-Jagged , Microtomografía por Rayos X , Proteína Gla de la Matriz
16.
Circ Res ; 113(5): 495-504, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23852538

RESUMEN

RATIONALE: Vascular calcification is a regulated process that involves osteoprogenitor cells and frequently complicates common vascular disease, such as atherosclerosis and diabetic vasculopathy. However, it is not clear whether the vascular endothelium has a role in contributing osteoprogenitor cells to the calcific lesions. OBJECTIVE: To determine whether the vascular endothelium contributes osteoprogenitor cells to vascular calcification. METHODS AND RESULTS: In this study, we use 2 mouse models of vascular calcification, mice with gene deletion of matrix Gla protein, a bone morphogenetic protein (BMP)-inhibitor, and Ins2Akita/+ mice, a diabetes model. We show that enhanced BMP signaling in both types of mice stimulates the vascular endothelium to contribute osteoprogenitor cells to the vascular calcification. The enhanced BMP signaling results in endothelial-mesenchymal transitions and the emergence of multipotent cells, followed by osteoinduction. Endothelial markers colocalize with multipotent and osteogenic markers in calcified arteries by immunostaining and fluorescence-activated cell sorting. Lineage tracing using Tie2-Gfp transgenic mice supports an endothelial origin of the osteogenic cells. Enhancement of matrix Gla protein expression in Ins2Akita/+ mice, as mediated by an Mgp transgene, limits the generation of multipotent cells. Moreover, matrix Gla protein-depleted human aortic endothelial cells in vitro acquire multipotency rendering the cells susceptible to osteoinduction by BMP and high glucose. CONCLUSIONS: Our data suggest that the endothelium is a source of osteoprogenitor cells in vascular calcification that occurs in disorders with high BMP activation, such as deficiency of BMP-inhibitors and diabetes mellitus.


Asunto(s)
Calcinosis/fisiopatología , Proteínas de Unión al Calcio/fisiología , Transdiferenciación Celular/fisiología , Angiopatías Diabéticas/fisiopatología , Células Endoteliales/patología , Endotelio Vascular/fisiopatología , Proteínas de la Matriz Extracelular/fisiología , Insulina/fisiología , Células Madre Multipotentes/patología , Enfermedades Vasculares/fisiopatología , Animales , Aorta/citología , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Linaje de la Célula , Células Cultivadas/efectos de los fármacos , Diabetes Mellitus Tipo 2/genética , Angiopatías Diabéticas/genética , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Proteínas de la Matriz Extracelular/deficiencia , Proteínas de la Matriz Extracelular/genética , Glucosa/farmacología , Heterocigoto , Humanos , Insulina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de Microfilamentos/fisiología , Proteínas Musculares/fisiología , ARN Interferente Pequeño/farmacología , Receptor TIE-2/genética , Proteínas Recombinantes de Fusión/fisiología , Transducción de Señal , Proteína Gla de la Matriz
17.
J Mol Cell Cardiol ; 53(6): 790-800, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22999861

RESUMEN

White mature adipocytes give rise to multipotent cells, so-called de-differentiated fat (DFAT) cells, when losing their fat in culture. The objective of this study was to examine the ability of DFAT cells to give rise to endothelial cells (ECs) in vitro and vivo. We demonstrate that mouse and human DFAT cells, derived from adipose tissue and lipospirate, respectively, initially lack expression of CD34, CD31, CD146, CD45 and pericyte markers, distinguishing them from progenitor cells previously identified in adipose stroma. The DFAT cells spontaneously differentiate into vascular ECs in vitro, as determined by real-time PCR, fluorescence activated cell sorting, immunostaining, and formation of tube structures. Treatment with bone morphogenetic protein (BMP)4 and BMP9, important in regulating angiogenesis, significantly enhances the EC differentiation. Furthermore, adipocyte-derived cells from Green Fluorescent Protein-transgenic mice were detected in the vasculature of infarcted myocardium up to 6 weeks after ligation of the left anterior descending artery in mice. We conclude that adipocyte-derived multipotent cells are able to spontaneously give rise to ECs, a process that is promoted by BMPs and may be important in cardiovascular regeneration and in physiological and pathological changes in fat and other tissues.


Asunto(s)
Adipocitos Blancos/citología , Desdiferenciación Celular/fisiología , Diferenciación Celular/fisiología , Células Madre Multipotentes/citología , Adipocitos Blancos/efectos de los fármacos , Animales , Proteína Morfogenética Ósea 4/farmacología , Desdiferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Expresión Génica , Factores de Diferenciación de Crecimiento/farmacología , Humanos , Masculino , Ratones , Ratones Transgénicos , Células Madre Multipotentes/efectos de los fármacos , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Trasplante de Células Madre
18.
Blood ; 119(21): 5037-47, 2012 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-22474252

RESUMEN

The importance of morphogenetic proteins (BMPs) and their antagonists in vascular development is increasingly being recognized. BMP-4 is essential for angiogenesis and is antagonized by matrix Gla protein (MGP) and crossveinless 2 (CV2), both induced by the activin receptor like-kinase 1 (ALK1) when stimulated by BMP-9. In this study, however, we show that CV2 preferentially binds and inhibits BMP-9 thereby providing strong feedback inhibition for BMP-9/ALK1 signaling rather than for BMP-4/ALK2 signaling. CV2 disrupts complex formation involving ALK2, ALK1, BMP-4, and BMP-9 required for the induction of both BMP antagonists. It also limits VEGF expression, proliferation, and tube formation in ALK1-expressing endothelial cells. In vivo, CV2 deficiency translates into a dysregulation of vascular BMP signaling, resulting in an abnormal endothelium with increased endothelial cellularity and expression of lineage markers for mature endothelial cells. Thus, mutual regulation by BMP-9 and CV2 is essential in regulating the development of the vascular endothelium.


Asunto(s)
Proteínas Portadoras/fisiología , Endotelio Vascular/metabolismo , Factor 2 de Diferenciación de Crecimiento/antagonistas & inhibidores , Factor 2 de Diferenciación de Crecimiento/metabolismo , Receptores de Activinas Tipo I/antagonistas & inhibidores , Receptores de Activinas Tipo I/metabolismo , Receptores de Activinas Tipo II/antagonistas & inhibidores , Receptores de Activinas Tipo II/metabolismo , Animales , Proteína Morfogenética Ósea 4/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Bovinos , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Factor 2 de Diferenciación de Crecimiento/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Unión Proteica/efectos de los fármacos , Especificidad por Sustrato
19.
Stem Cells ; 30(3): 386-91, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22232064

RESUMEN

The vasculature, an organ that penetrates every other organ, is ideally poised to be the site where pools of stem cells are placed, to be deployed and committed in response to feedback regulation, and to respond to demands for new vascular structures. These pools of multipotent cells are often under the regulation of various members of the transforming growth factor-ß superfamily, including the bone morphogenetic proteins and their antagonists. Regulation of stem cell populations affects their recruitment, differentiation, spatial organization, and their coordination with host tissue. Loss and dysregulation of feedback control cause a variety of diseases that involve ectopic tissue formation, including atherosclerotic lesion formation and calcification, diabetic vasculopathies, and arteriovenous malformations.


Asunto(s)
Vasos Sanguíneos/citología , Neovascularización Fisiológica , Células Madre/fisiología , Animales , Vasos Sanguíneos/fisiología , Diferenciación Celular , Proliferación Celular , Retroalimentación Fisiológica , Humanos , Regeneración , Células Madre/metabolismo
20.
J Clin Invest ; 121(8): 2993-3004, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21765215

RESUMEN

Arteriovenous malformations (AVMs) in organs, such as the lungs, intestine, and brain, are characteristic of hereditary hemorrhagic telangiectasia (HHT), a disease caused by mutations in activin-like kinase receptor 1 (ALK1), which is an essential receptor in angiogenesis, or endoglin. Matrix Gla protein (MGP) is an antagonist of BMPs that is highly expressed in lungs and kidneys and is regulated by ALK1. The objective of this study was to determine the role of MGP in the vasculature of the lungs and kidneys. We found that Mgp gene deletion in mice caused striking AVMs in lungs and kidneys, where overall small organ size contrasted with greatly increased vascularization. Mechanistically, MGP deficiency increased BMP activity in lungs. In cultured lung epithelial cells, BMP-4 induced VEGF expression through induction of ALK1, ALK2, and ALK5. The VEGF secretion induced by BMP-4 in Mgp-/- epithelial cells stimulated proliferation of ECs. However, BMP-4 inhibited proliferation of lung epithelial cells, consistent with the increase in pulmonary vasculature at the expense of lung tissue in the Mgp-null mice. Similarly, BMP signaling and VEGF expression were increased in Mgp-/- mouse kidneys. We therefore conclude that Mgp gene deletion is what we believe to be a previously unidentified cause of AVMs. Because lack of MGP also causes arterial calcification, our findings demonstrate that the same gene defect has drastically different effects on distinct vascular beds.


Asunto(s)
Malformaciones Arteriovenosas/genética , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Proteínas de la Matriz Extracelular/deficiencia , Proteínas de la Matriz Extracelular/genética , Receptores de Activinas Tipo I/metabolismo , Receptores de Activinas Tipo II , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Modelos Animales de Enfermedad , Endoglina , Células Endoteliales/citología , Eliminación de Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Riñón/metabolismo , Pulmón/citología , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica , Transducción de Señal , Tomografía Computarizada por Rayos X/métodos , Proteína Gla de la Matriz
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...