Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 151(2): 384-99, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-23063127

RESUMEN

Vascular endothelial growth factor and its receptors, FLK1/KDR and FLT1, are key regulators of angiogenesis. Unlike FLK1/KDR, the role of FLT1 has remained elusive. FLT1 is produced as soluble (sFLT1) and full-length isoforms. Here, we show that pericytes from multiple tissues produce sFLT1. To define the biologic role of sFLT1, we chose the glomerular microvasculature as a model system. Deletion of Flt1 from specialized glomerular pericytes, known as podocytes, causes reorganization of their cytoskeleton with massive proteinuria and kidney failure, characteristic features of nephrotic syndrome in humans. The kinase-deficient allele of Flt1 rescues this phenotype, demonstrating dispensability of the full-length isoform. Using cell imaging, proteomics, and lipidomics, we show that sFLT1 binds to the glycosphingolipid GM3 in lipid rafts on the surface of podocytes, promoting adhesion and rapid actin reorganization. sFLT1 also regulates pericyte function in vessels outside of the kidney. Our findings demonstrate an autocrine function for sFLT1 to control pericyte behavior.


Asunto(s)
Glomérulos Renales/citología , Glomérulos Renales/metabolismo , Podocitos/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Gangliósidos/metabolismo , Humanos , Técnicas In Vitro , Metabolismo de los Lípidos , Lípidos/química , Ratones , Ratones Transgénicos , Pericitos/metabolismo , Proteinuria/metabolismo , Transducción de Señal , Sindecanos/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
2.
Int J Mol Sci ; 25(8)2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38673910

RESUMEN

Endothelial cell (EC) injury is a crucial contributor to the progression of diabetic kidney disease (DKD), but the specific EC populations and mechanisms involved remain elusive. Kidney ECs (n = 5464) were collected at three timepoints from diabetic BTBRob/ob mice and non-diabetic littermates. Their heterogeneity, transcriptional changes, and alternative splicing during DKD progression were mapped using SmartSeq2 single-cell RNA sequencing (scRNAseq) and elucidated through pathway, network, and gene ontology enrichment analyses. We identified 13 distinct transcriptional EC phenotypes corresponding to different kidney vessel subtypes, confirmed through in situ hybridization and immunofluorescence. EC subtypes along nephrons displayed extensive zonation related to their functions. Differential gene expression analyses in peritubular and glomerular ECs in DKD underlined the regulation of DKD-relevant pathways including EIF2 signaling, oxidative phosphorylation, and IGF1 signaling. Importantly, this revealed the differential alteration of these pathways between the two EC subtypes and changes during disease progression. Furthermore, glomerular and peritubular ECs also displayed aberrant and dynamic alterations in alternative splicing (AS), which is strongly associated with DNA repair. Strikingly, genes displaying differential transcription or alternative splicing participate in divergent biological processes. Our study reveals the spatiotemporal regulation of gene transcription and AS linked to DKD progression, providing insight into pathomechanisms and clues to novel therapeutic targets for DKD treatment.


Asunto(s)
Empalme Alternativo , Nefropatías Diabéticas , Células Endoteliales , Análisis de la Célula Individual , Transcriptoma , Animales , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Ratones , Análisis de la Célula Individual/métodos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Riñón/metabolismo , Riñón/patología , Regulación de la Expresión Génica , Transcripción Genética , Perfilación de la Expresión Génica/métodos , Masculino
3.
Circ Res ; 128(4): e46-e62, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33375813

RESUMEN

RATIONALE: Pericytes are capillary mural cells playing a role in stabilizing newly formed blood vessels during development and tissue repair. Loss of pericytes has been described in several brain disorders, and genetically induced pericyte deficiency in the brain leads to increased macromolecular leakage across the blood-brain barrier (BBB). However, the molecular details of the endothelial response to pericyte deficiency remain elusive. OBJECTIVE: To map the transcriptional changes in brain endothelial cells resulting from lack of pericyte contact at single-cell level and to correlate them with regional heterogeneities in BBB function and vascular phenotype. METHODS AND RESULTS: We reveal transcriptional, morphological, and functional consequences of pericyte absence for brain endothelial cells using a combination of methodologies, including single-cell RNA sequencing, tracer analyses, and immunofluorescent detection of protein expression in pericyte-deficient adult Pdgfbret/ret mice. We find that endothelial cells without pericyte contact retain a general BBB-specific gene expression profile, however, they acquire a venous-shifted molecular pattern and become transformed regarding the expression of numerous growth factors and regulatory proteins. Adult Pdgfbret/ret brains display ongoing angiogenic sprouting without concomitant cell proliferation providing unique insights into the endothelial tip cell transcriptome. We also reveal heterogeneous modes of pericyte-deficient BBB impairment, where hotspot leakage sites display arteriolar-shifted identity and pinpoint putative BBB regulators. By testing the causal involvement of some of these using reverse genetics, we uncover a reinforcing role for angiopoietin 2 at the BBB. CONCLUSIONS: By elucidating the complexity of endothelial response to pericyte deficiency at cellular resolution, our study provides insight into the importance of brain pericytes for endothelial arterio-venous zonation, angiogenic quiescence, and a limited set of BBB functions. The BBB-reinforcing role of ANGPT2 (angiopoietin 2) is paradoxical given its wider role as TIE2 (TEK receptor tyrosine kinase) receptor antagonist and may suggest a unique and context-dependent function of ANGPT2 in the brain.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Pericitos/citología , Animales , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/patología , Proliferación Celular , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Linfocinas/deficiencia , Linfocinas/genética , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Pericitos/metabolismo , Pericitos/patología , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/genética , Análisis de la Célula Individual , Transcriptoma
4.
Annu Rev Physiol ; 78: 437-61, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26863327

RESUMEN

The glomerulus is a highly specialized microvascular bed that filters blood to form primary urinary filtrate. It contains four cell types: fenestrated endothelial cells, specialized vascular support cells termed podocytes, perivascular mesangial cells, and parietal epithelial cells. Glomerular cell-cell communication is critical for the development and maintenance of the glomerular filtration barrier. VEGF, ANGPT, EGF, SEMA3A, TGF-ß, and CXCL12 signal in paracrine fashions between the podocytes, endothelium, and mesangium associated with the glomerular capillary bed to maintain filtration barrier function. In this review, we summarize the current understanding of these signaling pathways in the development and maintenance of the glomerulus and the progression of disease.


Asunto(s)
Capilares/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Enfermedades Renales/metabolismo , Glomérulos Renales/metabolismo , Animales , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Humanos , Transducción de Señal/fisiología
5.
BMC Cancer ; 17(1): 539, 2017 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-28800750

RESUMEN

BACKGROUND: Angipoietin-1 activation of the tyrosine kinase receptor Tek expressed mainly on endothelial cells leads to survival and stabilization of endothelial cells. Studies have shown that Angiopoietin-1 counteracts permeability induced by a number of stimuli. Here, we test the hypothesis that loss of Angiopoietin-1/Tek signaling in the vasculature would increase metastasis. METHODS: Angiopoietin-1 was deleted in mice just before birth using floxed Angiopoietin-1 and Tek mice crossed to doxycycline-inducible bitransgenic ROSA-rtTA/tetO-Cre mice. By crossing Angiopoietin-1 knockout mice to the MMTV-PyMT autochthonous mouse breast cancer model, we investigated primary tumor growth and metastasis to the lung. Furthermore, we utilized B16F10 melanoma cells subcutaneous and experimental lung metastasis models in Angiopoietin-1 and Tek knockout mice. RESULTS: We found that primary tumor growth in MMTV-PyMT mice was unaffected, while metastasis to the lung was significantly increased in Angiopoietin-1 knockout MMTV-PyMT mice. In addition, angiopoietin-1 deficient mice exhibited a significant increase in lung metastasis of B16F10 melanoma cells, compared to wild type mice 3 weeks after injection. Additional experiments showed that this was likely an early event due to increased attachment or extravasation of tumor cells, since seeding of tumor cells was significantly increased 4 and 24 h post tail vein injection. Finally, using inducible Tek knockout mice, we showed a significant increase in tumor cell seeding to the lung, suggesting that Angiopoietin-1/Tek signaling is important for vascular integrity to limit metastasis. CONCLUSIONS: This study show that loss of the Angiopoietin-1/Tek vascular growth factor system leads to increased metastasis without affecting primary tumor growth.


Asunto(s)
Angiopoyetina 1/genética , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/patología , Melanoma/patología , Transducción de Señal , Angiopoyetina 1/metabolismo , Animales , Femenino , Expresión Génica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Noqueados , Metástasis de la Neoplasia/genética
6.
J Am Soc Nephrol ; 25(11): 2459-70, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24904088

RESUMEN

Podocytes are terminally differentiated cells with an elaborate cytoskeleton and are critical components of the glomerular barrier. We identified a bHLH transcription factor, Tcf21, that is highly expressed in developing and mature podocytes. Because conventional Tcf21 knockout mice die in the perinatal period with major cardiopulmonary defects, we generated a conditional Tcf21 knockout mouse to explore the role of this transcription factor in podocytes in vivo. Tcf21 was deleted from podocytes and podocyte progenitors using podocin-cre (podTcf21) and wnt4-cre (wnt4creTcf21) driver strains, respectively. Loss of Tcf21 from capillary-loop stage podocytes (podTcf21) results in simplified glomeruli with a decreased number of endothelial and mesangial cells. By 5 weeks of age, 40% of podTcf21 mice develop massive proteinuria and lesions similar to FSGS. Notably, the remaining 60% of mice do not develop proteinuria even when aged to 8 months. By contrast, earlier deletion of Tcf21 from podocyte precursors (wnt4creTcf21) results in a profound developmental arrest of podocyte differentiation and renal failure in 100% of mice during the perinatal period. Taken together, our results demonstrate a critical role for Tcf21 in the differentiation and maintenance of podocytes. Identification of direct targets of this transcription factor may provide new therapeutic avenues for proteinuric renal disease, including FSGS.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diabetes Mellitus Experimental/fisiopatología , Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Podocitos/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Línea Celular , Senescencia Celular/fisiología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Glomeruloesclerosis Focal y Segmentaria/genética , Glomeruloesclerosis Focal y Segmentaria/patología , Glomérulos Renales/embriología , Glomérulos Renales/patología , Glomérulos Renales/fisiopatología , Operón Lac , Ratones Noqueados , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Podocitos/patología , Proteinuria/genética , Proteinuria/patología , Proteinuria/fisiopatología
7.
Dev Biol ; 367(1): 40-54, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22546694

RESUMEN

There is a reciprocal interaction between pancreatic islet cells and vascular endothelial cells (EC) in which EC-derived signals promote islet cell differentiation and islet development while islet cell-derived angiogenic factors promote EC recruitment and extensive islet vascularization. To examine the role of angiogenic factors in the coordinated development of islets and their associated vessels, we used a "tet-on" inducible system (mice expressing rat insulin promoter-reverse tetracycline activator transgene and a tet-operon-angiogenic factor transgene) to increase the ß cell production of vascular endothelial growth factor-A (VEGF-A), angiopoietin-1 (Ang1), or angiopoietin-2 (Ang2) during islet cell differentiation and islet development. In VEGF-A overexpressing embryos, ECs began to accumulate around epithelial tubes residing in the central region of the developing pancreas (associated with endocrine cells) as early as embryonic day 12.5 (E12.5) and increased dramatically by E16.5. While α and ß cells formed islet cell clusters in control embryos at E16.5, the increased EC population perturbed endocrine cell differentiation and islet cell clustering in VEGF-A overexpressing embryos. With continued overexpression of VEGF-A, α and ß cells became scattered, remained adjacent to ductal structures, and never coalesced into islets, resulting in a reduction in ß cell proliferation and ß cell mass at postnatal day 1. A similar impact on islet morphology was observed when VEGF-A was overexpressed in ß cells during the postnatal period. In contrast, increased expression of Ang1 or Ang2 in ß cells in developing or adult islets did not alter islet differentiation, development, or morphology, but altered islet EC ultrastructure. These data indicate that (1) increased EC number does not promote, but actually impairs ß cell proliferation and islet formation; (2) the level of VEGF-A production by islet endocrine cells is critical for islet vascularization during development and postnatally; (3) angiopoietin-Tie2 signaling in endothelial cells does not have a crucial role in the development or maintenance of islet vascularization.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Animales , Recuento de Células , Células Endoteliales/metabolismo , Islotes Pancreáticos/irrigación sanguínea , Islotes Pancreáticos/metabolismo , Ratones
8.
Am J Physiol Renal Physiol ; 304(1): F120-6, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23054256

RESUMEN

Deletion of the von Hippel-Lindau tumor suppressor (Vhl) gene from renal podocytes of mice (podVhl KO) leads to rapidly progressive glomerulonephritis (RPGN), a clinical syndrome characterized by rapid loss of renal function and crescents on renal biopsy. Genomic profiling of glomeruli isolated from podVhl knockout (KO) mice and from patients with RPGN identified a fingerprint of genes regulated by hypoxia-inducible factors (HIF), important substrates of the product of the VHL gene. Here, we show that stabilization of Hifs in podocytes is both required and sufficient for the glomerular phenotype observed in podVhl KO mice. Genetic deletion of the obligate dimerization partner Arnt/Hif1b that is essential for Hif transcriptional function rescues the phenotype. Conversely, stabilization of HIF2A alone in podocytes results in crescentic glomerular disease. Together, our results show that the Hif pathway and Hif2a in particular are key players in maintenance of the glomerular barrier.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Glomerulonefritis/patología , Glomérulos Renales/metabolismo , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/fisiología , Glomerulonefritis/genética , Humanos , Glomérulos Renales/patología , Ratones , Ratones Noqueados , Podocitos/fisiología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
9.
Biomedicines ; 10(6)2022 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-35740360

RESUMEN

Hypercoagulation and endothelial dysfunction play central roles in severe forms of COVID-19 infections, but the molecular mechanisms involved are unclear. Increased plasma levels of the inflammatory cytokine and TIE2 receptor antagonist Angiopoietin-2 were reported in severely ill COVID-19 patients. In vitro experiments suggest that Angiopoietin-2 bind and inhibits thrombomodulin. Thrombomodulin is expressed on the luminal surface of endothelial cells where it is an important member of the intrinsic anticoagulant pathway through activation of protein C. Using clinical data, mouse models, and in vitro assays, we tested if Angiopoietin-2 plays a causal role in COVID-19-associated hypercoagulation through direct inhibition of thrombin/thrombomodulin-mediated physiological anticoagulation. Angiopoietin-2 was measured in 61 patients at admission, and after 10 days in the 40 patients remaining in the ICU. We found that Angiopoietin-2 levels were increased in COVID-19 patients in correlation with disease severity, hypercoagulation, and mortality. In support of a direct effect of Angiopoietin-2 on coagulation, we found that injected Angiopoietin-2 in mice associated to thrombomodulin and resulted in a shortened tail bleeding time, decreased circulating levels of activated protein C, and increased plasma thrombin/antithrombin complexes. Conversely, bleeding time was increased in endothelial-specific Angiopoietin-2 knockout mice, while knockout of Tie2 had no effect on tail bleeding. Using in vitro assays, we found that Angiopoietin-2 inhibited thrombomodulin-mediated anticoagulation and protein C activation in human donor plasma. Our data suggest a novel in vivo mechanism for Angiopoietin-2 in COVID-19-associated hypercoagulation, implicating that Angiopoietin-2 inhibitors may be effective in the treatment of hypercoagulation in severe COVID-19 infection.

10.
Curr Opin Nephrol Hypertens ; 20(3): 258-62, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21455064

RESUMEN

PURPOSE OF REVIEW: This review discusses the recent evidence that intrinsic glomerular cells including podocytes, parietal epithelial cells and progenitor cells within Bowman's capsule contribute to cellular crescents. RECENT FINDINGS: Using a variety of newer molecular markers and lineage tracing experiments, investigators have clearly demonstrated that glomerular cells play a key role in the development and progression of cellular crescents in experimental and human disease. SUMMARY: Crescentic glomerulonephritis is associated with significant morbidity and mortality. Current therapies target the immune system. The recent finding that nonimmune cells also play a role in crescent formation highlights the need to identify alternate and complimentary therapeutic strategies.


Asunto(s)
Cápsula Glomerular/patología , Glomerulonefritis/etiología , Podocitos/fisiología , Animales , Glomerulonefritis/patología , Humanos , Proteínas de Filamentos Intermediarios/fisiología , Ratones , Proteínas del Tejido Nervioso/fisiología , Nestina , Células Madre/fisiología
11.
Nat Commun ; 12(1): 2141, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33837218

RESUMEN

Molecular characterization of the individual cell types in human kidney as well as model organisms are critical in defining organ function and understanding translational aspects of biomedical research. Previous studies have uncovered gene expression profiles of several kidney glomerular cell types, however, important cells, including mesangial (MCs) and glomerular parietal epithelial cells (PECs), are missing or incompletely described, and a systematic comparison between mouse and human kidney is lacking. To this end, we use Smart-seq2 to profile 4332 individual glomerulus-associated cells isolated from human living donor renal biopsies and mouse kidney. The analysis reveals genetic programs for all four glomerular cell types (podocytes, glomerular endothelial cells, MCs and PECs) as well as rare glomerulus-associated macula densa cells. Importantly, we detect heterogeneity in glomerulus-associated Pdgfrb-expressing cells, including bona fide intraglomerular MCs with the functionally active phagocytic molecular machinery, as well as a unique mural cell type located in the central stalk region of the glomerulus tuft. Furthermore, we observe remarkable species differences in the individual gene expression profiles of defined glomerular cell types that highlight translational challenges in the field and provide a guide to design translational studies.


Asunto(s)
Células Endoteliales/metabolismo , Mesangio Glomerular/metabolismo , Podocitos/metabolismo , Biosíntesis de Proteínas/genética , Transcriptoma/fisiología , Animales , Separación Celular , Biología Computacional , Citometría de Flujo , Heterogeneidad Genética , Mesangio Glomerular/citología , Humanos , Masculino , Ratones , RNA-Seq , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptores de Fosfolipasa A2/genética , Análisis de la Célula Individual , Especificidad de la Especie
12.
J Am Soc Nephrol ; 20(1): 114-22, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19073829

RESUMEN

The pathophysiology underlying the nephrotic syndrome is becoming clear for several inherited podocytopathies; the mechanisms of injury that lead to the acquired forms of this disease are not well understood. We explored these mechanisms using the mouse model of adriamycin-induced proteinuria.We estimated the fractional clearances for FITC-Ficolls, albumin, and neutral albumin in cooled, isolated,perfused kidneys (cIPK) in situ. Treatment with adriamycin led to a significant increase in the fractional clearance of albumin and of Ficoll with radii larger than 20 A. Neutral albumin (33.4 A) and similarly sized Ficoll behaved similarly to each other. In addition, adriamycin led to a significant loss of charge density and size selectivity of the glomerular barrier. The thickness of the glomerular endothelial surface layer(i.e., or the glycocalyx) in adriamycin-treated animals was only 20% of that in normal animals. Finally,several proteoglycans were downregulated in isolated glomeruli. In summary, adriamycin thins the glomerular glycocalyx, perhaps by downregulating proteoglycan synthesis, and alters glomerular charge- and size selectivity. These data suggest that the glomerular endothelium may play a role in the pathogenesis of proteinuric renal diseases.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Doxorrubicina/toxicidad , Endotelio/efectos de los fármacos , Glomérulos Renales/efectos de los fármacos , Proteinuria/inducido químicamente , Animales , Endotelio/metabolismo , Femenino , Glomérulos Renales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , ARN Mensajero/análisis , Versicanos/análisis , Versicanos/genética
13.
Curr Opin Nephrol Hypertens ; 18(4): 331-5, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19458528

RESUMEN

PURPOSE OF REVIEW: In 2008, more than 376 papers were published on the glomerular barrier. Most of them dealt with the podocyte and its role in kidney disease. RECENT FINDINGS: There is new information on signaling pathways that are utilized in podocytes during proteinuria. Interestingly, the glomerular endothelium, with its fenestrae and glycocalyx, seems to be important for the maintenance of an intact glomerular barrier. All new advances at the molecular level are compatible with a highly size and charge-selective glomerular membrane and refute the concept of a 'leaky' glomerular barrier with tubular retrieval of intact albumin. Still, the hypothesis has its advocates, keeping a stimulating 'charge debate' alive. SUMMARY: Glomerular diseases account for 90% of chronic kidney disease requiring dialysis and transplantation at an annual cost of $20 billion in the USA. In clinical practice, we lack specific treatment of these diseases, giving us plenty of room for improvement. Future research should be directed toward deeper understanding of the signaling pathways involved in different conditions of proteinuria, the cross-talk between cell types in the glomerulus, and the identification of novel targets for treatment of acquired kidney disease.


Asunto(s)
Glomérulos Renales/metabolismo , Animales , Células Endoteliales/fisiología , Humanos , Permeabilidad , Podocitos/fisiología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/fisiología
14.
PLoS One ; 13(1): e0189433, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29293543

RESUMEN

Presence of tubulointerstitial fibrosis is predictive of progressive decline in kidney function, independent of its underlying cause. Injury to the renal microvasculature is a major factor in the progression of fibrosis and identification of factors that regulate endothelium in fibrosis is desirable as they might be candidate targets for treatment of kidney diseases. The current study investigates how loss of Angipoietin-1 (Angpt1), a ligand for endothelial tyrosine-kinase receptor Tek (also called Tie2), affects tubulointerstitial fibrosis and renal microvasculature. Inducible Angpt1 knockout mice were subjected to unilateral ureteral obstruction (UUO) to induce fibrosis, and kidneys were collected at different time points up to 10 days after obstruction. Staining for aSMA showed that Angpt1 deficient kidneys had significantly more fibrosis compared to wildtype mice 3, 6, and 10 days after UUO. Further investigation 3 days after UUO showed a significant increase of Col1a1 and vimentin in Angpt1 deficient mice, as well as increased gene expression of Tgfb1, Col1a1, Fn1, and CD44. Kidney injury molecule 1 (Kim1/Havcr1) was significantly more increased in Angpt1 deficient mice 1 and 3 days after UUO, suggesting a more severe injury early in the fibrotic process in Angpt1 deficient mice. Staining for endomucin showed that capillary rarefaction was evident 3 days after UUO and Angpt1 deficient mice had significantly less capillaries 6 and 10 days after UUO compared to UUO kidneys in wildtype mice. RNA sequencing revealed downregulation of several markers for endothelial cells 3 days after UUO, and that Angpt1 deficient mice had a further downregulation of Emcn, Plvap, Pecam1, Erg, and Tek. Our results suggest that loss of Angpt1 is central in capillary rarefaction and fibrogenesis and propose that manipulations to maintain Angpt1 levels may slow down fibrosis progression.


Asunto(s)
Angiopoyetina 1/fisiología , Capilares/fisiopatología , Riñón/irrigación sanguínea , Nefritis Intersticial/genética , Angiopoyetina 1/genética , Animales , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
Eur J Pharm Biopharm ; 114: 186-193, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28161551

RESUMEN

BACKGROUND: Docetaxel is a cytostatic agent approved for treatment of non-small cell lung cancer as well as other cancers. Although docetaxel is an effective cytostatic agent, its effectiveness in clinical practice is associated with a variety of acute and long term side-effects. To overcome systemic side-effects, a slow release formulation based on calcium sulfate with docetaxel for intra-tumoral administration was developed. METHODS: Two formulations with the calcium sulfate NanoZolid technology were generated with a twofold difference in docetaxel drug load. The formulations were injected intra-tumorally as a paste which solidified within the tumor. The effects of the two intra-tumoral injection formulations were tested in female mice (n=60) inoculated with subcutaneous Lewis lung carcinoma cells. The two formulations were compared to systemic intraperitoneal injection of docetaxel and a placebo formulation without docetaxel. Tumor volumes were measured and systemic side-effects were evaluated using body weight and cell counts from whole blood as well as plasma concentrations. RESULTS: Both docetaxel formulations showed a significantly higher antitumor efficacy compared to placebo, which was comparable to that of systemic administration of docetaxel. Moreover, the intra-tumoral formulations with docetaxel showed reduced systemic toxicity compared to systemic treatment, including less weight loss and no decrease in blood cell counts. CONCLUSIONS: The results suggest that intra-tumoral slow release calcium sulfate based formulations with docetaxel can be an alternative strategy as an efficient local antitumoral treatment with reduced systemic toxicity.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacología , Sulfato de Calcio/química , Taxoides/administración & dosificación , Taxoides/farmacología , Animales , Antineoplásicos Fitogénicos/toxicidad , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/patología , Docetaxel , Composición de Medicamentos , Excipientes , Femenino , Humanos , Inyecciones Intralesiones , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C57BL , Taxoides/toxicidad
16.
Eur J Pharmacol ; 539(3): 184-91, 2006 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-16707122

RESUMEN

The present study investigated whether sympathetic neurotransmission is altered at an early stage of diabetes in mesenteric small arteries isolated from female non-obese diabetic (NOD) and control animals without diabetes from the same mouse strain. The NOD diabetic mice had increased plasma glucose and hypertension. Confocal microscopy showed distribution of nerve terminals was similar, but immunoreaction intensity for neuropeptide Y (NPY) and tyrosine hydroxylase was higher in small arteries from NOD diabetic compared with NOD control mice. In the presence of prazosin and activated with vasopressin, electrical field stimulation evoked contractions which were more pronounced in mesenteric arteries from NOD diabetic versus NOD control mice and inhibited by the NPY Y(1) receptor antagonist, BIBP 3226. NPY concentration-response curves were leftward shifted in arteries from NOD diabetic versus NOD control both in arteries with and without endothelium, but not in the presence of the BIBP 3226. The present findings suggest that enhanced NPY content and vasoconstriction to NPY by activation of NPY Y(1) receptors in arteries from diabetic mice may contribute to the enhanced sympathetic nerve activity and vascular resistance in female non-obese early diabetic animals.


Asunto(s)
Diabetes Mellitus Experimental/inmunología , Arterias Mesentéricas/inmunología , Neuropéptido Y/fisiología , Vasoconstricción/inmunología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Fluoroinmunoensayo , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/enzimología , Ratones , Ratones Endogámicos NOD , Neuropéptido Y/inmunología , Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/fisiología , Tirosina 3-Monooxigenasa/metabolismo , Vasoconstricción/efectos de los fármacos
17.
J Clin Invest ; 124(10): 4320-4, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25202984

RESUMEN

Glaucoma is a leading cause of blindness, afflicting more than 60 million people worldwide. Increased intraocular pressure (IOP) due to impaired aqueous humor drainage is a major risk factor for the development of glaucoma. Here, we demonstrated that genetic disruption of the angiopoietin/TIE2 (ANGPT/TIE2) signaling pathway results in high IOP, buphthalmos, and classic features of glaucoma, including retinal ganglion degeneration and vision loss. Eyes from mice with induced deletion of Angpt1 and Angpt2 (A1A2Flox(WB) mice) lacked drainage pathways in the corneal limbus, including Schlemm's canal and lymphatic capillaries, which share expression of the PROX1, VEGFR3, and FOXC family of transcription factors. VEGFR3 and FOXCs have been linked to lymphatic disorders in patients, and FOXC1 has been linked to glaucoma. In contrast to blood endothelium, in which ANGPT2 is an antagonist of ANGPT1, we have shown that both ligands cooperate to regulate TIE2 in the lymphatic network of the eye. While A1A2Flox(WB) mice developed high IOP and glaucoma, expression of ANGPT1 or ANGPT2 alone was sufficient for ocular drainage. Furthermore, we demonstrated that loss of FOXC2 from lymphatics results in TIE2 downregulation, suggesting a mechanism for ocular defects in patients with FOXC mutations. These data reveal a pathogenetic and molecular basis for glaucoma and demonstrate the importance of angiopoietin ligand cooperation in the lymphatic endothelium.


Asunto(s)
Angiopoyetina 1/genética , Glaucoma/patología , Hipertensión Ocular/patología , Receptor TIE-2/genética , Angiopoyetina 2/genética , Animales , Humor Acuoso , Separación Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Citometría de Flujo , Factores de Transcripción Forkhead/metabolismo , Proteínas de Homeodominio/metabolismo , Presión Intraocular , Ligandos , Sistema Linfático/patología , Ratones , Ratones Noqueados , Mutación , Malla Trabecular/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
Diabetes ; 61(11): 2958-66, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23093658

RESUMEN

Vascular endothelial growth factor A (VEGFA) expression is increased in glomeruli in the context of diabetes. Here, we tested the hypothesis that this upregulation of VEGFA protects the glomerular microvasculature in diabetes and that therefore inhibition of VEGFA will accelerate nephropathy. To determine the role of glomerular Vegfa in the development and progression of diabetic nephropathy, we used an inducible Cre-loxP gene-targeting system that enabled genetic deletion of Vegfa selectively from glomerular podocytes of wild-type or diabetic mice. Type 1 diabetes was induced in mice using streptozotocin (STZ). We then assessed the extent of glomerular dysfunction by measuring proteinuria, glomerular pathology, and glomerular cell apoptosis. Vegfa expression increased in podocytes in the STZ model of diabetes. After 7 weeks of diabetes, diabetic mice lacking Vegfa in podocytes exhibited significantly greater proteinuria with profound glomerular scarring and increased apoptosis compared with control mice with diabetes or Vegfa deletion without diabetes. Reduced local production of glomerular Vegfa in a mouse model of type 1 diabetes promotes endothelial injury accelerating the progression of glomerular injury. These results suggest that upregulation of VEGFA in diabetic kidneys protects the microvasculature from injury and that reduction of VEGFA in diabetes may be harmful.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/metabolismo , Microvasos/metabolismo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis , Células Cultivadas , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/complicaciones , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Nefropatías Diabéticas/orina , Progresión de la Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Humanos , Hiperglucemia/etiología , Glomérulos Renales/patología , Glomérulos Renales/fisiopatología , Ratones , Ratones Noqueados , Ratones Transgénicos , Microvasos/patología , Podocitos/metabolismo , Podocitos/patología , Proteinuria/etiología , ARN Mensajero/metabolismo , Distribución Aleatoria , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética
19.
PLoS One ; 7(11): e50996, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23226445

RESUMEN

The kidney filtration barrier is formed by the combination of endothelial cells, basement membrane and epithelial cells called podocytes. These specialized actin-rich cells form long and dynamic protrusions, the foot processes, which surround glomerular capillaries and are connected by specialized intercellular junctions, the slit diaphragms. Failure to maintain the filtration barrier leads to massive proteinuria and nephrosis. A number of proteins reside in the slit diaphragm, notably the transmembrane proteins Nephrin and Neph1, which are both able to act as tyrosine phosphorylated scaffolds that recruit cytoplasmic effectors to initiate downstream signaling. While association between tyrosine-phosphorylated Neph1 and the SH2/SH3 adaptor Grb2 was shown in vitro to be sufficient to induce actin polymerization, in vivo evidence supporting this finding is still lacking. To test this hypothesis, we generated two independent mouse lines bearing a podocyte-specific constitutive inactivation of the Grb2 locus. Surprisingly, we show that mice lacking Grb2 in podocytes display normal renal ultra-structure and function, thus demonstrating that Grb2 is not required for the establishment of the glomerular filtration barrier in vivo. Moreover, our data indicate that Grb2 is not required to restore podocyte function following kidney injury. Therefore, although in vitro experiments suggested that Grb2 is important for the regulation of actin dynamics, our data clearly shows that its function is not essential in podocytes in vivo, thus suggesting that Grb2 rather plays a secondary role in this process.


Asunto(s)
Proteína Adaptadora GRB2/metabolismo , Barrera de Filtración Glomerular/metabolismo , Animales , Cruzamientos Genéticos , Femenino , Silenciador del Gen , Genotipo , Barrera de Filtración Glomerular/patología , Barrera de Filtración Glomerular/fisiopatología , Barrera de Filtración Glomerular/ultraestructura , Integrasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Especificidad de Órganos , Podocitos/metabolismo , Podocitos/patología , Podocitos/ultraestructura
20.
J Clin Invest ; 121(6): 2278-89, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21606590

RESUMEN

Angiopoietin-1/Tek signaling is a critical regulator of blood vessel development, with conventional knockout of angiopoietin-1 or Tek in mice being embryonically lethal due to vascular defects. In addition, angiopoietin-1 is thought to be required for the stability of mature vessels. Using a Cre-Lox conditional gene targeting approach, we have studied the role of angiopoietin-1 in embryonic and adult vasculature. We report here that angiopoietin-1 is critical for regulating both the number and diameter of developing vessels but is not required for pericyte recruitment. Cardiac-specific knockout of angiopoietin-1 reproduced the phenotype of the conventional knockout, demonstrating that the early vascular abnormalities arise from flow-dependent defects. Strikingly, deletion in the entire embryo after day E13.5 produced no immediate vascular phenotype. However, when combined with injury or microvascular stress, angiopoietin-1 deficiency resulted in profound organ damage, accelerated angiogenesis, and fibrosis. These findings redefine our understanding of the biological roles of angiopoietin-1: it is dispensable in quiescent vessels but has a powerful ability to modulate the vascular response after injury.


Asunto(s)
Angiopoyetina 1/fisiología , Vasos Sanguíneos/embriología , Vasos Sanguíneos/lesiones , Neovascularización Fisiológica/fisiología , Cicatrización de Heridas/fisiología , Angiopoyetina 1/deficiencia , Angiopoyetina 1/genética , Animales , Vasos Sanguíneos/citología , Diabetes Mellitus Experimental/fisiopatología , Nefropatías Diabéticas/fisiopatología , Corazón Fetal/crecimiento & desarrollo , Corazón Fetal/patología , Regulación del Desarrollo de la Expresión Génica , Humanos , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/patología , Hígado/irrigación sanguínea , Ratones , Ratones Noqueados , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Neovascularización Patológica/embriología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Pericitos/metabolismo , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptor TIE-1/fisiología , Receptor TIE-2 , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA