Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Artif Organs ; 24(3): 402-406, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33459910

RESUMEN

Lymphoma-associated Hemophagocytic lymphohistiocytosis (HLH) represents a severe complication of disease progression, mediated through cytokine release from the lymphoma cells. Cytokine adsorption may contribute as a supportive treatment to stabilize organ function by reduction of cytokine levels. So far, no experiences of cytokine adsorption and simultaneous stem cell transplantation were published. We report the case of a patient with aggressive lymphoma secondary to chronic lymphocytic leukemia with rapidly progressive HLH (Richter's transformation) upon conditioning chemotherapy prior to allogeneic stem cell transplantation (ASCT). Continuous hemodiafiltration was initiated in the treatment of shock with acute renal failure, lactacidosis and need for high-dose catecholamine therapy, integrating an additional cytokine-adsorbing filter (CytoSorb®) to reduce cytokine levels. This was followed by scheduled allogenic stem cell transplantation. We observed a marked decrease in interleukin-6 plasma levels, associated with a reduced need for vasopressor therapy and organ function stabilization. Hematopoietic engraftment was present at day 14 post-ASCT, leading to disease-free discharge at day 100 post-transplantation. Cytokine adsorption may serve as a safe adjunct to HLH/sepsis treatment during allogeneic stem cell transplantation. Clinical studies are required to make future treatment recommendations.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Linfohistiocitosis Hemofagocítica , Linfoma , Adsorción , Citocinas , Humanos , Linfohistiocitosis Hemofagocítica/diagnóstico , Linfohistiocitosis Hemofagocítica/terapia , Trasplante de Células Madre , Acondicionamiento Pretrasplante
2.
Int J Mol Sci ; 22(11)2021 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-34070742

RESUMEN

Nephrogenesis is driven by complex signaling pathways that control cell growth and differentiation. The endoplasmic reticulum chaperone calreticulin (Calr) is well known for its function in calcium storage and in the folding of glycoproteins. Its role in kidney development is still not understood. We provide evidence for a pivotal role of Calr in nephrogenesis in this investigation. We show that Calr deficiency results in the disrupted formation of an intact nephrogenic zone and in retardation of nephrogenesis, as evidenced by the disturbance in the formation of comma-shaped and s-shaped bodies. Using proteomics and transcriptomics approaches, we demonstrated that in addition to an alteration in Wnt-signaling key proteins, embryonic kidneys from Calr-/- showed an overall impairment in expression of ribosomal proteins which reveals disturbances in protein synthesis and nephrogenesis. CRISPR/cas9 mediated knockout confirmed that Calr deficiency is associated with a deficiency of several ribosomal proteins and key proteins in ribosome biogenesis. Our data highlights a direct link between Calr expression and the ribosome biogenesis.


Asunto(s)
Calcio/metabolismo , Calreticulina/genética , Riñón/metabolismo , Biogénesis de Organelos , Proteínas Ribosómicas/genética , Ribosomas/genética , Animales , Señalización del Calcio , Calreticulina/deficiencia , Embrión de Mamíferos , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Femenino , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas/clasificación , Glicoproteínas/genética , Glicoproteínas/metabolismo , Riñón/crecimiento & desarrollo , Riñón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organogénesis/genética , Pliegue de Proteína , Proteómica/métodos , Proteínas Ribosómicas/deficiencia , Ribosomas/metabolismo , Ribosomas/patología , Vía de Señalización Wnt
3.
Int J Mol Sci ; 21(9)2020 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-32344896

RESUMEN

The field of genome editing started with the discovery of meganucleases (e.g., the LAGLIDADG family of homing endonucleases) in yeast. After the discovery of transcription activator-like effector nucleases and zinc finger nucleases, the recently discovered clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated proteins (Cas) system has opened a new window of applications in the field of gene editing. Here, we review different Cas proteins and their corresponding features including advantages and disadvantages, and we provide an overview of the different endonuclease-deficient Cas protein (dCas) derivatives. These dCas derivatives consist of an endonuclease-deficient Cas9 which can be fused to different effector domains to perform distinct in vitro applications such as tracking, transcriptional activation and repression, as well as base editing. Finally, we review the in vivo applications of these dCas derivatives and discuss their potential to perform gene activation and repression in vivo, as well as their potential future use in human therapy.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas CRISPR-Cas , Endodesoxirribonucleasas/metabolismo , Epigenómica/métodos , Edición Génica/métodos , Proteína 9 Asociada a CRISPR/metabolismo , Cromatina/ultraestructura , ADN/metabolismo , Endonucleasas/metabolismo , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Imagen Óptica , ARN Guía de Kinetoplastida/genética , Proteínas Recombinantes de Fusión/análisis , Especificidad por Sustrato , Telómero/ultraestructura , Nucleasas de los Efectores Tipo Activadores de la Transcripción/metabolismo , Efectores Tipo Activadores de la Transcripción/metabolismo , Transcripción Genética , Dedos de Zinc
4.
Kidney Int ; 91(1): 157-176, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27692563

RESUMEN

Acute kidney injury (AKI) and progressive chronic kidney disease (CKD) are intrinsically tied syndromes. In this regard, the acutely injured kidney often does not achieve its full regenerative capacity and AKI directly transitions into progressive CKD associated with tubulointerstitial fibrosis. Underlying mechanisms of such AKI-to-CKD progression are still incompletely understood and specific therapeutic interventions are still elusive. Because epigenetic modifications play a role in maintaining tissue fibrosis, we used a murine model of ischemia-reperfusion injury to determine whether aberrant promoter methylation of RASAL1 contributes causally to the switch between physiological regeneration and tubulointerstitial fibrogenesis, a hallmark of AKI-to-CKD progression. It is known that the antihypertensive drug hydralazine has demethylating activity, and that its optimum demethylating activity occurs at concentrations below blood pressure-lowering doses. Administration of low-dose hydralazine effectively induced expression of hydroxylase TET3, which catalyzed RASAL1 hydroxymethylation and subsequent RASAL1 promoter demethylation. Hydralazine-induced CpG promoter demethylation subsequently attenuated renal fibrosis and preserved excretory renal function independent of its blood pressure-lowering effects. In comparison, RASAL1 demethylation and inhibition of tubulointerstitial fibrosis was not detected upon administration of the angiotensin-converting enzyme inhibitor Ramipril in this model. Thus, RASAL1 promoter methylation and subsequent transcriptional RASAL1 suppression plays a causal role in AKI-to-CKD progression.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Proteínas de Unión al ADN/metabolismo , Proteínas Activadoras de GTPasa/genética , Hidralazina/uso terapéutico , Riñón/patología , Proteínas Proto-Oncogénicas/metabolismo , Insuficiencia Renal Crónica/prevención & control , Vasodilatadores/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Islas de CpG , Metilación de ADN , Dioxigenasas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Epigénesis Genética , Fibroblastos/metabolismo , Fibrosis , Humanos , Hidralazina/administración & dosificación , Riñón/citología , Riñón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Cultivo Primario de Células , Regiones Promotoras Genéticas , Ramipril/farmacología , Eliminación Renal/efectos de los fármacos , Daño por Reperfusión/complicaciones , Vasodilatadores/administración & dosificación
5.
Circ Res ; 116(5): 857-66, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25587097

RESUMEN

RATIONALE: Endocardial fibroelastosis (EFE) is a unique form of fibrosis, which forms a de novo subendocardial tissue layer encapsulating the myocardium and stunting its growth, and which is typically associated with congenital heart diseases of heterogeneous origin, such as hypoplastic left heart syndrome. Relevance of EFE was only recently highlighted through the establishment of staged biventricular repair surgery in infant patients with hypoplastic left heart syndrome, where surgical removal of EFE tissue has resulted in improvement in the restrictive physiology leading to the growth of the left ventricle in parallel with somatic growth. However, pathomechanisms underlying EFE formation are still scarce, and specific therapeutic targets are not yet known. OBJECTIVE: Here, we aimed to investigate the cellular origins of EFE tissue and to gain insights into the underlying molecular mechanisms to ultimately develop novel therapeutic strategies. METHODS AND RESULTS: By utilizing a novel EFE model of heterotopic transplantation of hearts from newborn reporter mice and by analyzing human EFE tissue, we demonstrate for the first time that fibrogenic cells within EFE tissue originate from endocardial endothelial cells via aberrant endothelial to mesenchymal transition. We further demonstrate that such aberrant endothelial to mesenchymal transition involving endocardial endothelial cells is caused by dysregulated transforming growth factor beta/bone morphogenetic proteins signaling and that this imbalance is at least in part caused by aberrant promoter methylation and subsequent transcriptional suppression of bone morphogenetic proteins 5 and 7. Finally, we provide evidence that supplementation of exogenous recombinant bone morphogenetic proteins 7 effectively ameliorates endothelial to mesenchymal transition and experimental EFE in rats. CONCLUSIONS: In summary, our data point to aberrant endothelial to mesenchymal transition as a common denominator of infant EFE development in heterogeneous, congenital heart diseases, and to bone morphogenetic proteins 7 as an effective treatment for EFE and its restriction of heart growth.


Asunto(s)
Transdiferenciación Celular/fisiología , Fibroelastosis Endocárdica/patología , Endocardio/patología , Epitelio/patología , Mesodermo/patología , Animales , Animales Recién Nacidos , Antígenos CD/genética , Biomarcadores , Proteína Morfogenética Ósea 7/genética , Proteína Morfogenética Ósea 7/fisiología , Proteína Morfogenética Ósea 7/uso terapéutico , Cadherinas/genética , Transdiferenciación Celular/genética , Células Cultivadas , Metilación de ADN , Fibroelastosis Endocárdica/tratamiento farmacológico , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Trasplante de Corazón , Humanos , Síndrome del Corazón Izquierdo Hipoplásico/patología , Síndrome del Corazón Izquierdo Hipoplásico/cirugía , Lactante , Recién Nacido , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Ratas , Ratas Endogámicas Lew , Proteínas Recombinantes/uso terapéutico , Transducción de Señal/fisiología , Proteínas Smad/genética , Proteínas Smad/fisiología , Factor de Crecimiento Transformador beta/fisiología , Trasplante Heterotópico
6.
Differentiation ; 92(4): 225-236, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27266810

RESUMEN

Endothelial cells derived from human induced pluripotent stem cells (hiPSC- EC) are of significant value for research on human vascular development, in vitro disease models and drug screening. Here we report an alternative, highly efficient and cost-effective simple three step method (mesoderm induction, endothelial cell differentiation and endothelial cell expansion) to differentiate hiPSC directly into endothelial cells. We demonstrate that efficiency of described method to derive CD31+ and VE-Cadherin+ double positive cells is higher than 80% in 12 days. Most notably we established that hiPSC-EC differentiation efficacy depends on optimization of both mesoderm differentiation and endothelial cell differentiation steps.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/genética , Células Endoteliales/citología , Células Madre Pluripotentes Inducidas/citología , Antígenos CD/metabolismo , Cadherinas/metabolismo , Linaje de la Célula/genética , Proliferación Celular , Humanos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Células Madre Pluripotentes
7.
J Biol Chem ; 290(27): 16653-64, 2015 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-25971970

RESUMEN

Endothelial to mesenchymal transition (EndMT) was originally described in heart development where the endocardial endothelial cells that line the atrioventricular canal undergo an EndMT to form the endocardial mesenchymal cushion that later gives rise to the septum and mitral and tricuspid valves. In the postnatal heart specifically, endothelial cells that originate from the endocardium maintain increased susceptibility to undergo EndMT as remnants from their embryonic origin. Such EndMT involving adult coronary endothelial cells contributes to microvascular rarefaction and subsequent chronification of hypoxia in the injured heart, ultimately leading to cardiac fibrosis. Although in most endothelial beds hypoxia induces tip cell formation and sprouting angiogenesis, here we demonstrate that hypoxia is a stimulus for human coronary endothelial cells to undergo phenotypic changes reminiscent of EndMT via a mechanism involving hypoxia-inducible factor 1α-induced activation of the EndMT master regulatory transcription factor SNAIL. Our study adds further evidence for the unique susceptibility of endocardium-derived endothelial cells to undergo EndMT and provides novel insights into how hypoxia contributes to progression of cardiac fibrosis. Additional studies may be required to discriminate between distinct sprouting angiogenesis and EndMT responses of different endothelial cells populations.


Asunto(s)
Endocardio/metabolismo , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Endocardio/citología , Células Endoteliales/citología , Humanos , Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
8.
Biochem Biophys Res Commun ; 472(3): 459-64, 2016 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-26774342

RESUMEN

Phosphate is an essential constituent of critical cellular functions including energy metabolism, nucleic acid synthesis and phosphorylation-dependent cell signaling. Increased plasma phosphate levels are an independent risk factor for lowered life-expectancy as well as for heart and kidney failure. Nevertheless, direct cellular effects of elevated phosphate concentrations within the microenvironment are poorly understood and have been largely neglected in favor of phosphor-regulatory hormones. Because interstitial fibrosis is the common determinant of chronic progressive kidney disease, and because fibroblasts are major mediators of fibrogenesis, we here explored the effect of high extracellular phosphate levels on renal fibroblasts. We demonstrate that high inorganic phosphate directly induces fibrotic fibroblast activation associated with increased proliferative activity, increased expression of α-smooth muscle actin and increased synthesis of type I collagen. We further demonstrate that such fibroblast activation is dependent on phosphate influx, aberrant phosphorylation of DNA methyltransferase DNMT1 and aberrant CpG island promoter methylation. In summary, our studies demonstrate that elevated phosphate concentrations induce pro-fibrotic fibroblast activation independent of phospho-regulatory hormones.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Fosfatos/toxicidad , Línea Celular , Proliferación Celular/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasa 1 , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibrosis , Humanos , Fosfatos/administración & dosificación , Fosforilación/efectos de los fármacos
9.
Kidney Int ; 88(3): 429-31, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26323066

RESUMEN

Among gliptins, linagliptin is unique, because decreased glomerular filtration rate does not require dose reduction. Linagliptin was originally developed to lower blood glucose by inhibiting dipeptidyl peptidase-4 (DPP-4). However, DPP-4 has numerous additional substrates, and thus gliptins possess a vast range of additional off-target effects. Shi et al. report that linagliptin directly targets interaction of DPP-4 with integrin ß1, preventing endothelial-mesenchymal transition and ultimately renal fibrosis, providing additional rationale for use of linagliptin in diabetic nephropathy.


Asunto(s)
Diabetes Mellitus Experimental/enzimología , Nefropatías Diabéticas/enzimología , Dipeptidil Peptidasa 4/metabolismo , Células Endoteliales/enzimología , Transición Epitelial-Mesenquimal , Integrina beta1/metabolismo , Riñón/enzimología , Animales , Masculino
10.
Am J Pathol ; 184(10): 2687-98, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25111226

RESUMEN

Thrombospondin-1 (TSP1) is a multifunctional matricellular protein known to promote progression of chronic kidney disease. To gain insight into the underlying mechanisms through which TSP1 accelerates chronic kidney disease, we compared disease progression in Col4a3 knockout (KO) mice, which develop spontaneous kidney failure, with that of Col4a3;Tsp1 double-knockout (DKO) mice. Decline of excretory renal function was significantly delayed in the absence of TSP1. Although Col4a3;Tsp1 DKO mice did progress toward end-stage renal failure, their kidneys exhibited distinct histopathological lesions, compared with creatinine level-matched Col4a3 KO mice. Although kidneys of both Col4a3 KO and Col4a3;Tsp1 DKO mice exhibited a widened tubulointerstitium, predominant lesions in Col4a3 KO kidneys were collagen deposition and fibroblast accumulation, whereas in Col4a3;Tsp1 DKO kidney inflammation was predominant, with less collagen deposition. Altered disease progression correlated with impaired activation of transforming growth factor-ß1 (TGF-ß1) in vivo and in vitro in the absence of TSP1. In summary, our findings suggest that TSP1 contributes to progression of chronic kidney disease by catalyzing activation of latent TGF-ß1, resulting in promotion of a fibroproliferative response over an inflammatory response. Furthermore, the findings suggest that fibroproliferative and inflammatory lesions are independent entities, both of which contribute to decline of renal function.


Asunto(s)
Fibrosis/patología , Fallo Renal Crónico/patología , Insuficiencia Renal/patología , Trombospondina 1/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Autoantígenos/metabolismo , Colágeno Tipo IV/metabolismo , Creatinina/sangre , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis/metabolismo , Riñón/patología , Fallo Renal Crónico/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nefritis , Insuficiencia Renal/metabolismo
11.
Nat Rev Cancer ; 6(5): 392-401, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16572188

RESUMEN

Tumours are known as wounds that do not heal - this implies that cells that are involved in angiogenesis and the response to injury, such as endothelial cells and fibroblasts, have a prominent role in the progression, growth and spread of cancers. Fibroblasts are associated with cancer cells at all stages of cancer progression, and their structural and functional contributions to this process are beginning to emerge. Their production of growth factors, chemokines and extracellular matrix facilitates the angiogenic recruitment of endothelial cells and pericytes. Fibroblasts are therefore a key determinant in the malignant progression of cancer and represent an important target for cancer therapies.


Asunto(s)
Fibroblastos/patología , Neoplasias/patología , Neovascularización Patológica , Animales , Progresión de la Enfermedad , Fibroblastos/metabolismo , Humanos , Metástasis de la Neoplasia , Neoplasias/metabolismo
12.
J Am Soc Nephrol ; 25(5): 905-12, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24480825

RESUMEN

Methylation of CpG island promoters is an epigenetic event that can effectively silence transcription over multiple cell generations. Hypermethylation of the Rasal1 promoter contributes to activation of fibroblasts and progression of kidney fibrosis. Here, we explored whether such causative hypermethylation could be reversed through endogenous mechanisms and whether such reversal of hypermethylation is a constituent of the antifibrotic activity of bone morphogenic protein 7 (BMP7). We show that successful inhibition of experimental kidney fibrosis through administration of BMP7 associates with normalization of Rasal1 promoter hypermethylation. Furthermore, this reversal of pathologic hypermethylation was achieved specifically through Tet3-mediated hydroxymethylation. Collectively, our findings reveal a new mechanism that may be exploited to facilitate therapeutic DNA demethylation to reverse kidney fibrosis.


Asunto(s)
Proteína Morfogenética Ósea 7/uso terapéutico , Metilación de ADN/efectos de los fármacos , Proteínas de Unión al ADN/fisiología , Proteínas Activadoras de GTPasa/genética , Silenciador del Gen , Nefroesclerosis/etiología , Nefroesclerosis/prevención & control , Proteínas Proto-Oncogénicas/fisiología , Animales , Biomarcadores/metabolismo , Proteína Morfogenética Ósea 7/metabolismo , Proteína Morfogenética Ósea 7/farmacología , Células Cultivadas , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Dioxigenasas , Epigénesis Genética , Ratones , Nefroesclerosis/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Obstrucción Ureteral/etiología , Obstrucción Ureteral/genética , Obstrucción Ureteral/prevención & control
13.
Nephrol Dial Transplant ; 29 Suppl 1: i1-i8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24046191

RESUMEN

Epigenetics are omnipresent in eukaryotic cells and influence cell differentiation and maintenance of cell metabolism in health and disease. Here, we discuss how the 'second genetic code' impacts the fate of the injured kidney. We provide a glimpse of how recent insights into epigenetic mechanisms of chronic kidney disease might lead to novel diagnostic and therapeutic tools.


Asunto(s)
Epigénesis Genética/genética , Insuficiencia Renal Crónica/genética , Animales , Progresión de la Enfermedad , Humanos
14.
Nephrol Dial Transplant ; 29 Suppl 4: iv72-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23975750

RESUMEN

Chronic kidney disease (CKD) which can lead to end-stage renal failure remains a principal challenge in Nephrology. While mechanistic studies provided extensive insights into the common pathways of fibrogenesis which underlie the progression of CKD, these pre-clinical studies fail to fully explain the vastly different progression slopes of individual patients. Recent studies provide evidence that genetic polymorphisms and epigenetic variations determine the individual susceptibility of patients to develop chronic progressive kidney disease. Here, we review recent insights that were provided by genome-wide association studies (GWASs), gene-linkage studies and epigenome analysis. The progression of CKD towards end-stage renal failure remains a principal unsolved problem in Nephrology as effective therapies and predictive tests are still not available [ 1, 2]. Chronic progressive kidney disease is caused by a wide range of diseases, with diabetes mellitus, hypertension and primary glomerulopathies being the most common causes in the Western world [ 3]. Infections, physical obstruction, interstitial nephritides and genetic cystic kidney diseases are also common causes of end-stage renal disease (ESRD) [ 3]. Regardless of the primary underlying disease, chronically injured kidneys are histomorphologically characterized by tubulointerstitial fibrosis [ 1]. In fact, the extent of tubulointerstitial fibrosis is the best predictor for kidney survival, irrespective of the underlying disease. For this reason, fibrosis is considered the common pathway of chronic progressive kidney disease [ 1]. Fibrogenesis is a pathological scarring process which involves accumulation of activated fibroblasts, excessive deposition of extracellular matrix, failed regeneration of tubular epithelium, microvascular rarefaction and (mostly sterile) inflammation [ 4]. Fibrogenesis depends on a complex interaction of the involved cell types which is orchestrated by an extensive network of growth factors and signalling pathways (which are reviewed extensively elsewhere) [ 1]. In view of the detailed mechanistic knowledge of the pathways that orchestrate renal fibrogenesis, it is puzzling why progression rates of CKD differ dramatically among patients with identical underlying diseases [ 1, 2]. The fibrotic pathways are known, but the switches that control their intensities and which determine the speed at which fibrosis moves along the progression slope are not yet understood [ 1, 2]. The concept that genetic polymorphisms are the basis for individual progression rates of CKD is an obvious and attractive one. Distinct susceptibilities of different mouse and rat strains to experimental CKD are a strong testament of the impact of genetic variations on renal fibrogenesis. Identification of the underlying genetic polymorphisms and mechanistic proof of their involvement in the progression of CKD, however, is an ongoing challenge. There are two basic approaches: one strategy is to perform unbiased screening to identify genes which are associated with chronic progressive kidney disease and to then prove their mechanistic relevance in experimental studies ('genotype to phenotype approach'). The second strategy is to selectively analyse polymorphisms of genes which have been identified in mechanistic studies as drivers of renal fibrogenesis with regard to their association with CKD (phenotype to genotype approach). The puzzling observation, however, is that genetic analysis and mechanistic studies so far rarely complement each other. The current state of affairs is reviewed in more detail below.


Asunto(s)
Epigénesis Genética , Genómica , Riñón/patología , Animales , Progresión de la Enfermedad , Fibrosis/diagnóstico , Fibrosis/genética , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Ratas , Transducción de Señal
15.
J Pathol ; 229(2): 264-73, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23097091

RESUMEN

The aberrant methylation of CpG island promoters of selected genes is the prominent epigenetic mechanism by which gene transcription can be effectively silenced. Aberrant hypermethylation of a few selected genes plays an important role in facilitating fibrotic fibroblast activation and in driving fibrogenesis. Here we review mechanisms of DNA methylation and demethylation and their implications for fibroblast activation and tissue fibrosis.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Fibroblastos/metabolismo , Enfermedades Renales/genética , Riñón/metabolismo , Regiones Promotoras Genéticas , Animales , Islas de CpG , Fibroblastos/patología , Fibrosis , Regulación de la Expresión Génica , Silenciador del Gen , Humanos , Riñón/patología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Proteínas Activadoras de ras GTPasa/genética
16.
Nat Med ; 13(8): 952-61, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17660828

RESUMEN

Cardiac fibrosis, associated with a decreased extent of microvasculature and with disruption of normal myocardial structures, results from excessive deposition of extracellular matrix, which is mediated by the recruitment of fibroblasts. The source of these fibroblasts is unclear and specific anti-fibrotic therapies are not currently available. Here we show that cardiac fibrosis is associated with the emergence of fibroblasts originating from endothelial cells, suggesting an endothelial-mesenchymal transition (EndMT) similar to events that occur during formation of the atrioventricular cushion in the embryonic heart. Transforming growth factor-beta1 (TGF-beta1) induced endothelial cells to undergo EndMT, whereas bone morphogenic protein 7 (BMP-7) preserved the endothelial phenotype. The systemic administration of recombinant human BMP-7 (rhBMP-7) significantly inhibited EndMT and the progression of cardiac fibrosis in mouse models of pressure overload and chronic allograft rejection. Our findings show that EndMT contributes to the progression of cardiac fibrosis and that rhBMP-7 can be used to inhibit EndMT and to intervene in the progression of chronic heart disease associated with fibrosis.


Asunto(s)
Diferenciación Celular , Fibrosis Endomiocárdica/patología , Células Endoteliales/patología , Mesodermo/patología , Animales , Células de la Médula Ósea/patología , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/farmacología , Proteínas Morfogenéticas Óseas/uso terapéutico , Línea Celular , Células Cultivadas , Enfermedad Crónica , Fibrosis Endomiocárdica/tratamiento farmacológico , Células Endoteliales/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/patología , Mesodermo/efectos de los fármacos , Ratones , Ratones Transgénicos , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta/uso terapéutico , Factor de Crecimiento Transformador beta1/farmacología
17.
Proc Natl Acad Sci U S A ; 108(38): 16002-7, 2011 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-21911392

RESUMEN

Increased numbers of S100A4(+) cells are associated with poor prognosis in patients who have cancer. Although the metastatic capabilities of S100A4(+) cancer cells have been examined, the functional role of S100A4(+) stromal cells in metastasis is largely unknown. To study the contribution of S100A4(+) stromal cells in metastasis, we used transgenic mice that express viral thymidine kinase under control of the S100A4 promoter to specifically ablate S100A4(+) stromal cells. Depletion of S100A4(+) stromal cells significantly reduced metastatic colonization without affecting primary tumor growth. Multiple bone marrow transplantation studies demonstrated that these effects of S100A4(+) stromal cells are attributable to local non-bone marrow-derived S100A4(+) cells, which are likely fibroblasts in this setting. Reduction in metastasis due to the loss of S100A4(+) fibroblasts correlated with a concomitant decrease in the expression of several ECM molecules and growth factors, particularly Tenascin-C and VEGF-A. The functional importance of stromal Tenascin-C and S100A4(+) fibroblast-derived VEGF-A in metastasis was established by examining Tenascin-C null mice and transgenic mice expressing Cre recombinase under control of the S100A4 promoter crossed with mice carrying VEGF-A alleles flanked by loxP sites, which exhibited a significant decrease in metastatic colonization without effects on primary tumor growth. In particular, S100A4(+) fibroblast-derived VEGF-A plays an important role in the establishment of an angiogenic microenvironment at the metastatic site to facilitate colonization, whereas stromal Tenascin-C may provide protection from apoptosis. Our study demonstrates a crucial role for local S100A4(+) fibroblasts in providing the permissive "soil" for metastatic colonization, a challenging step in the metastatic cascade.


Asunto(s)
Proteínas S100/metabolismo , Células del Estroma/metabolismo , Tenascina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Ganciclovir/farmacología , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína de Unión al Calcio S100A4 , Proteínas S100/genética , Células del Estroma/efectos de los fármacos , Tenascina/genética , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Microambiente Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética
18.
Am J Physiol Cell Physiol ; 304(3): C216-25, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23255577

RESUMEN

Fibrosis is a pathological scarring process that leads to destruction of organ architecture and impairment of organ function. Chronic loss of organ function in most organs, including bone marrow, heart, intestine, kidney, liver, lung, and skin, is associated with fibrosis, contributing to an estimated one third of natural deaths worldwide. Effective therapies to prevent or to even reverse existing fibrotic lesions are not yet available in any organ. There is hope that an understanding of common fibrosis pathways will lead to development of antifibrotic therapies that are effective in all of these tissues in the future. Here we review common and organ-specific pathways of tissue fibrosis.


Asunto(s)
Cicatriz/patología , Fibrosis/patología , Cicatrización de Heridas/fisiología , Animales , Cicatriz/tratamiento farmacológico , Fibrosis/tratamiento farmacológico , Humanos , Cicatrización de Heridas/efectos de los fármacos
19.
Clin Epigenetics ; 15(1): 149, 2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37697352

RESUMEN

BACKGROUND: Aortic stenosis (AS) is one of the most common cardiac diseases and major cause of morbidity and mortality in the elderly. Transcatheter aortic valve implantation (TAVI) is performed in such patients with symptomatic severe AS and reduces mortality for the majority of these patients. However, a significant percentage dies within the first two years after TAVI, such that there is an interest to identify parameters, which predict outcome and could guide pre-TAVI patient selection. High levels of cardiac fibrosis have been identified as such independent predictor of cardiovascular mortality after TAVI. Promoter hypermethylation commonly leads to gene downregulation, and the Iroquois homeobox 3 (IRX3) gene was identified in a genome-wide transcriptome and methylome to be hypermethylated and downregulated in AS patients. In a well-described cohort of 100 TAVI patients in which cardiac fibrosis levels were quantified histologically in cardiac biopsies, and which had a follow-up of up to two years, we investigated if circulating methylated DNA of IRX3 in the peripheral blood is associated with cardiac fibrosis and/or mortality in AS patients undergoing TAVI and thus could serve as a biomarker to add information on outcome after TAVI. RESULTS: Patients with high levels of methylation in circulating IRX3 show a significantly increased survival as compared to patients with low levels of IRX3 methylation indicating that high peripheral IRX3 methylation is associated with an improved outcome. In the multivariable setting, peripheral IRX3 methylation acts as an independent predictor of all-cause mortality. While there is no significant correlation of levels of IRX3 methylation with cardiac death, there is a significant but very weak inverse correlation between circulating IRX3 promoter methylation level and the amount of cardiac fibrosis. Higher levels of peripheral IRX3 methylation further correlated with decreased cardiac IRX3 expression and vice versa. CONCLUSIONS: High levels of IRX3 methylation in the blood of AS patients at the time of TAVI are associated with better overall survival after TAVI and at least partially reflect myocardial IRX3 expression. Circulating methylated IRX3 might aid as a potential biomarker to help guide both pre-TAVI patient selection and post-TAVI monitoring.


Asunto(s)
Estenosis de la Válvula Aórtica , Ácidos Nucleicos Libres de Células , Reemplazo de la Válvula Aórtica Transcatéter , Humanos , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/cirugía , Biopsia , Metilación de ADN , Proteínas de Homeodominio/genética , Reemplazo de la Válvula Aórtica Transcatéter/efectos adversos , Factores de Transcripción/genética
20.
Dev Cell ; 58(17): 1562-1577.e8, 2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37625403

RESUMEN

Oncogenic KRASG12D (KRAS∗) is critical for the initiation and maintenance of pancreatic ductal adenocarcinoma (PDAC) and is a known repressor of tumor immunity. Conditional elimination of KRAS∗ in genetic mouse models of PDAC leads to the reactivation of FAS, CD8+ T cell-mediated apoptosis, and complete eradication of tumors. KRAS∗ elimination recruits activated CD4+ and CD8+ T cells and promotes the activation of antigen-presenting cells. Mechanistically, KRAS∗-mediated immune evasion involves the epigenetic regulation of Fas death receptor in cancer cells, via methylation of its promoter region. Furthermore, analysis of human RNA sequencing identifies that high KRAS expression in PDAC tumors shows a lower proportion of CD8+ T cells and demonstrates shorter survival compared with tumors with low KRAS expression. This study highlights the role of CD8+ T cells in the eradication of PDAC following KRAS∗ elimination and provides a rationale for the combination of KRAS∗ targeting with immunotherapy to control PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Proteínas Proto-Oncogénicas p21(ras) , Animales , Humanos , Ratones , Apoptosis , Carcinoma Ductal Pancreático/genética , Linfocitos T CD8-positivos , Epigénesis Genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA