Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Odontology ; 111(1): 57-67, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35739380

RESUMEN

NG2+ pericytes, as the possible precursor cells of mesenchymal stem cells (MSCs), have drawn attention due to their ability to differentiate into odontoblasts. Cav1.2 is involved in the differentiation process of stem cells, but its role in the differentiation of NG2+ pericytes is not clear. The aim of the present study was to examine the role of Cav1.2 in the differentiation of NG2+ pericytes into odontoblasts. NG2+ pericytes were obtained from human dental pulp cells by magnetic-activated cell sorting. During the odontogenic differentiation of NG2+ pericytes, the effects of the Cav1.2 inhibitors, nimodipine and Cav1.2 knockdown shRNA, were analyzed by real-time polymerase chain reaction and alizarin red staining. NG2CreERT2/Rosa26-GFP lineage-tracing mice were established to further investigate the roles of NG2+ pericytes and Cav1.2 in incisor self-repair after injury in vivo. At 10 min, 1 day, and 3 days after pulp injuries in transgenic mice, NG2-GFP+ and Cav1.2 immunofluorescence co-staining was performed on the incisors. Nimodipine treatment and Cav1.2 knockdown showed similar inhibition of calcium nodule formation and mRNA levels of osteogenic markers (DSPP, DMP1, and Runx2, p < 0.05). NG2+ pericytes migrated from their inherent perivascular location to the odontoblast layers after pulp injury. Cav1.2 showed a similar response pattern as NG2+ pericytes and gradually returned to normal levels. In addition, many co-stained areas of Cav1.2 and NG2+ pericytes, both near the perivascular and odontoblast layers, were observed. These results indicate that Cav1.2 played a vital role in the odontogenic differentiation of NG2+ pericytes, and that it might be closely linked to the NG2+ pericytes-mediated repair of dental pulp injury in vivo.


Asunto(s)
Proteínas de la Matriz Extracelular , Pericitos , Ratones , Humanos , Animales , Pericitos/química , Nimodipina , Pulpa Dental , Diferenciación Celular , Odontoblastos , Células Cultivadas
2.
Chin J Traumatol ; 23(2): 89-95, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32192909

RESUMEN

Pericyte, a kind of pluripotent cell, may regulate the irrigation flow and permeability of microcirculation. Pericytes are similar to the smooth muscle cells, which express several kinds of contractile proteins and have contractility. The dysfunction of pericytes is related to many microvascular diseases, including hypoxia, hypertension, diabetic retinopathy, fibrosis, inflammation, Alzheimer's disease, multiple sclerosis, and tumor formation. For a long time, their existence and function have been neglected. The distribution, structure, biomarker, related signaling pathways as well as the roles of pericytes on vascular diseases will be introduced in this review.


Asunto(s)
Pericitos , Investigación , Proteínas Contráctiles/metabolismo , Humanos , Microcirculación , Pericitos/química , Pericitos/patología , Pericitos/fisiología , Enfermedades Vasculares/etiología
3.
Neurobiol Dis ; 113: 70-81, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29432809

RESUMEN

BACKGROUND: Cerebrovascular dysfunction and inflammation occur in epilepsy. Here we asked whether pericytes, a pivotal cellular component of brain capillaries, undergo pathological modifications during experimental epileptogenesis and in human epilepsy. We evaluated whether pro-inflammatory cytokines, present in the brain during seizures, contribute to pericyte morphological modifications. METHODS: In vivo, unilateral intra-hippocampal kainic acid (KA) injections were performed in NG2DsRed/C57BL6 mice to induce status epilepticus (SE), epileptogenesis, and spontaneous recurrent seizures (SRS). NG2DsRed mice were used to visualize pericytes during seizure progression. The effect triggered by recombinant IL-1ß, TNFα, or IL-6 on pericytes was evaluated in NG2DsRed hippocampal slices and in human-derived cell culture. Human brain specimens obtained from temporal lobe epilepsy (TLE) with or without sclerosis (HS) and focal cortical dysplasia (FCD-IIb) were evaluated for pericyte-microglial cerebrovascular assembly. RESULTS: A disarray of NG2DsRed+ pericyte soma and ramifications was found 72 h post-SE and 1 week post-SE (epileptogenesis) in the hippocampus. Pericyte modifications topographically overlapped with IBA1+ microglia clustering around the capillaries with cases of pericytes lodged within the microglial cells. Microglial clustering around the NG2DsRed pericytes lingered at SRS. Pericyte proliferation (Ki67+) occurred 72 h post-SE and during epileptogenesis and returned towards control levels at SRS. Human epileptic brain tissues showed pericyte-microglia assemblies with IBA1/HLA microglial cells outlining the capillary wall in TLE-HS and FCD-IIb specimens. Inflammatory mediators contributed to pericyte modifications, in particular IL-1ß elicited pericyte morphological changes and pericyte-microglia clustering in NG2DsRed hippocampal slices. Modifications also occurred when pro-inflammatory cytokines were added to an in vitro culture of pericytes. CONCLUSIONS: These results indicate the occurrence of pericytosis during seizures and introduce a pericyte-microglial mediated mechanism of blood-brain barrier dysfunction in epilepsy.


Asunto(s)
Circulación Cerebrovascular/fisiología , Progresión de la Enfermedad , Mediadores de Inflamación/metabolismo , Microglía/metabolismo , Pericitos/metabolismo , Convulsiones/metabolismo , Adolescente , Adulto , Animales , Barrera Hematoencefálica/química , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Células Cultivadas , Niño , Preescolar , Femenino , Hipocampo/irrigación sanguínea , Hipocampo/química , Hipocampo/metabolismo , Hipocampo/fisiopatología , Humanos , Lactante , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/química , Pericitos/química , Convulsiones/fisiopatología
4.
Microsc Microanal ; 21(4): 837-48, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26040442

RESUMEN

The aim of the present research was to trace CD34+ stromal fibroblastic cells (CD34+ SFCs) in the palatal connective tissue harvested for muco-gingival surgical procedures and in granulation tissues from periodontal pockets using immunohistochemical and transmission electron microscopy. Immunohistochemical analysis targeted the presence of three antigens: CD31, α-smooth muscle actin (α-SMA), and CD34. In the palate, CD31 staining revealed a colored inner ring of the vessels representing the endothelium, α-SMA+ was located in the medial layer of the vasculature, and CD34 was intensely expressed by endothelial cells and artery adventitial cells (considered to be CD34+ SFCs). Granulation tissue showed the same pattern for CD31+ and α-SMA, but a different staining pattern for CD34. Ultrastructural examination of the palatal tissue highlighted perivascular cells with fibroblast-like characteristics and pericytes in close spatial relationship to endothelial cells. The ultrastructural evaluation of granulation tissue sections confirmed the presence of neovasculature and the inflammatory nature of this tissue. The present study traced the presence of CD34+ SFCs and of pericytes in the palatal connective tissue thus highlighting once more its intrinsic regenerative capabilities. The clinical and systemic factors triggering mobilization and influencing the fate of local CD34+SCFs and other progenitors are issues to be further investigated.


Asunto(s)
Antígenos CD34/análisis , Fibroblastos/fisiología , Encía/fisiología , Tejido de Granulación/crecimiento & desarrollo , Mucosa Bucal/fisiología , Hueso Paladar/fisiología , Regeneración , Fibroblastos/química , Encía/citología , Humanos , Inmunohistoquímica , Microscopía Electrónica de Transmisión , Mucosa Bucal/citología , Hueso Paladar/citología , Pericitos/química , Pericitos/fisiología
5.
Nat Cell Biol ; 9(3): 255-67, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17293855

RESUMEN

Cells derived from blood vessels of human skeletal muscle can regenerate skeletal muscle, similarly to embryonic mesoangioblasts. However, adult cells do not express endothelial markers, but instead express markers of pericytes, such as NG2 proteoglycan and alkaline phosphatase (ALP), and can be prospectively isolated from freshly dissociated ALP(+) cells. Unlike canonical myogenic precursors (satellite cells), pericyte-derived cells express myogenic markers only in differentiated myotubes, which they form spontaneously with high efficiency. When transplanted into severe combined immune deficient-X-linked, mouse muscular dystrophy (scid-mdx) mice, pericyte-derived cells colonize host muscle and generate numerous fibres expressing human dystrophin. Similar cells isolated from Duchenne patients, and engineered to express human mini-dystrophin, also give rise to many dystrophin-positive fibres in vivo. These data show that myogenic precursors, distinct from satellite cells, are associated with microvascular walls in the human skeletal muscle, may represent a correlate of embryonic 'mesoangioblasts' present after birth and may be a promising candidate for future cell-therapy protocols in patients.


Asunto(s)
Células Madre Adultas/citología , Músculo Esquelético/citología , Pericitos/citología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/citología , Adolescente , Adulto , Células Madre Adultas/metabolismo , Células Madre Adultas/trasplante , Anciano , Animales , Antígenos CD/análisis , Técnicas de Cultivo de Célula/métodos , Niño , Preescolar , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos mdx , Ratones Desnudos , Ratones SCID , Persona de Mediana Edad , Proteínas Musculares/análisis , Proteínas Musculares/genética , Músculo Esquelético/química , Músculo Esquelético/fisiología , Distrofia Muscular de Duchenne/fisiopatología , Distrofia Muscular de Duchenne/cirugía , Pericitos/química , Pericitos/trasplante , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/trasplante , Trasplante de Células Madre/métodos , Resultado del Tratamiento
6.
Cancer Cell ; 6(6): 553-63, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15607960

RESUMEN

The recent landmark Phase III clinical trial with a VEGF-specific antibody suggests that antiangiogenic therapy must be combined with cytotoxic therapy for the treatment of solid tumors. However, there are no guidelines for optimal scheduling of these therapies. Here we show that VEGFR2 blockade creates a "normalization window"--a period during which combined radiation therapy gives the best outcome. This window is characterized by an increase in tumor oxygenation, which is known to enhance radiation response. During the normalization window, but not before or after it, VEGFR2 blockade increases pericyte coverage of brain tumor vessels via upregulation of Ang1 and degrades their pathologically thick basement membrane via MMP activation.


Asunto(s)
Vasos Sanguíneos/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/uso terapéutico , Angiopoyetina 1/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antígenos/análisis , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Membrana Basal/efectos de los fármacos , Membrana Basal/metabolismo , Membrana Basal/patología , Vasos Sanguíneos/química , Vasos Sanguíneos/efectos de la radiación , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Colágeno Tipo IV/análisis , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Terapia Combinada/métodos , Dipéptidos/farmacología , Efrina-B2/genética , Angiografía con Fluoresceína , Rayos gamma/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/metabolismo , Glioma/radioterapia , Humanos , Inmunohistoquímica , Masculino , Inhibidores de la Metaloproteinasa de la Matriz , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Desnudos , Modelos Biológicos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/radioterapia , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxígeno/metabolismo , Pericitos/química , Pericitos/citología , Pericitos/fisiología , Proteoglicanos/análisis , Receptor TIE-2/antagonistas & inhibidores , Receptor TIE-2/inmunología , Factores de Tiempo , Transfección , Regulación hacia Arriba/genética
7.
Minerva Stomatol ; 60(1-2): 25-33, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21252847

RESUMEN

AIM: Tuberous sclerosis is a neurocutaneous syndrome characterized by affect multiple organs such as brain, kidneys, heart, eyes, lungs and skin. The aim of this study was to analyze the pattern of immunolocalization of markers MMP-1, MMP-10, TIMP-1, α-SMA and TGF-ß1 in oral and facial angiofibromas in individuals affected by tuberous sclerosis. METHODS: Microscopical analyses on hematoxilin-eosin and immunohistochemistry reactions were performed to analyze the previously cited biological markers pattern in orofacial angiofibromas. RESULTS: Reactivity was observed for MMP-1, MMP-10 and TGF-ß1, in addition to negative for TIMP-1 and α-SMA, except perivascular and epithelial staining for this. Concerning the intensity, a strong marking for MMP-1 in the basal layer of the epithelium, and a slight positivity in the suprabasal layers predominated. MMP-10 was slightly expressed in all epithelial layers. The connective tissue showed slight to moderate reactivity for MMP-1 and MMP-10. TIMP-1 demonstrated slight to moderate marking in the various layers of a single lesion and to TGF-ß1 expression showed varied in intensity staining both between lesions and between tissue layers. CONCLUSION: MMP-1, MMP-10 and TGF-ß1 exhibited reactivity in oral and cutaneous angiofibromas with heterogeneous distribution patterns among both tissue elements analyzed in the intensity of marking the same among the specimens. TIMP-1 showed reactivity predominantly negative in the specimens analyzed and α-SMA presented restricted to epithelial and perivascular regions of these lesions.


Asunto(s)
Actinas/análisis , Angiofibroma/química , Biomarcadores de Tumor/análisis , Neoplasias Faciales/química , Metaloproteinasa 10 de la Matriz/análisis , Metaloproteinasa 1 de la Matriz/análisis , Neoplasias de la Boca/química , Proteínas de Neoplasias/análisis , Neoplasias Primarias Múltiples/química , Inhibidor Tisular de Metaloproteinasa-1/análisis , Factor de Crecimiento Transformador beta1/análisis , Esclerosis Tuberosa/metabolismo , Adolescente , Adulto , Anciano , Angiofibroma/genética , Niño , Células Epiteliales/química , Células Epiteliales/ultraestructura , Neoplasias Faciales/genética , Femenino , Fibroblastos/química , Fibroblastos/ultraestructura , Neoplasias Gingivales/química , Neoplasias Gingivales/genética , Humanos , Técnicas para Inmunoenzimas , Neoplasias de los Labios/química , Neoplasias de los Labios/genética , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/genética , Neoplasias Primarias Múltiples/genética , Pericitos/química , Pericitos/ultraestructura , Neoplasias Cutáneas/química , Neoplasias Cutáneas/genética
8.
Fluids Barriers CNS ; 18(1): 43, 2021 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-34544422

RESUMEN

BACKGROUND: The pathways that control protein transport across the blood-brain barrier (BBB) remain poorly characterized. Despite great advances in recapitulating the human BBB in vitro, current models are not suitable for systematic analysis of the molecular mechanisms of antibody transport. The gaps in our mechanistic understanding of antibody transcytosis hinder new therapeutic delivery strategy development. METHODS: We applied a novel bioengineering approach to generate human BBB organoids by the self-assembly of astrocytes, pericytes and brain endothelial cells with unprecedented throughput and reproducibility using micro patterned hydrogels. We designed a semi-automated and scalable imaging assay to measure receptor-mediated transcytosis of antibodies. Finally, we developed a workflow to use CRISPR/Cas9 gene editing in BBB organoid arrays to knock out regulators of endocytosis specifically in brain endothelial cells in order to dissect the molecular mechanisms of receptor-mediated transcytosis. RESULTS: BBB organoid arrays allowed the simultaneous growth of more than 3000 homogenous organoids per individual experiment in a highly reproducible manner. BBB organoid arrays showed low permeability to macromolecules and prevented transport of human non-targeting antibodies. In contrast, a monovalent antibody targeting the human transferrin receptor underwent dose- and time-dependent transcytosis in organoids. Using CRISPR/Cas9 gene editing in BBB organoid arrays, we showed that clathrin, but not caveolin, is required for transferrin receptor-dependent transcytosis. CONCLUSIONS: Human BBB organoid arrays are a robust high-throughput platform that can be used to discover new mechanisms of receptor-mediated antibody transcytosis. The implementation of this platform during early stages of drug discovery can accelerate the development of new brain delivery technologies.


Asunto(s)
Anticuerpos/metabolismo , Bioingeniería/métodos , Barrera Hematoencefálica/metabolismo , Organoides/metabolismo , Receptores de Transferrina/metabolismo , Transcitosis/fisiología , Animales , Anticuerpos/análisis , Astrocitos/química , Astrocitos/metabolismo , Barrera Hematoencefálica/química , Barrera Hematoencefálica/citología , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/química , Células Endoteliales/metabolismo , Humanos , Organoides/química , Organoides/citología , Pericitos/química , Pericitos/metabolismo , Receptores de Transferrina/análisis
9.
J Comp Neurol ; 529(6): 1121-1134, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32812219

RESUMEN

Pericytes are a unique class of mural cells essential for angiogenesis, maintenance of the vasculature and are key players in microvascular pathology. However, their diversity and specific roles are poorly understood, limiting our insight into vascular physiology and the ability to develop effective therapies. Here, in the mouse retina, a tractable model of the CNS, we evaluated distinct classes of mural cells along the vascular tree for both structural characterization and physiological manipulation of blood flow. To accomplish this, we first tested three inducible mural cell-specific mouse lines using a sensitive Ai14 reporter and tamoxifen application either by a systemic injection, or by local administration in the form of eye drops. The specificity and pattern of cre activation varied significantly across the three lines, under either the PDGFRß or NG2 promoter (Pdgfrß-CreRha, Pdgfrß-CreCsln, and Cspg4-Cre). In particular, a mouse line with Cre under the NG2 promoter resulted in sparse TdTomato labeling of mural cells, allowing for an unambiguous characterization of anatomical features of individual sphincter cells and capillary pericytes. Furthermore, in one PDGFRß line, we found that focal eye drop application of tamoxifen led to an exclusive Cre-activation in pericytes, without affecting arterial mural cells. We then used this approach to boost capillary blood flow by selective expression of Halorhodopsin, a highly precise hyperpolarizing optogenetic actuator. The ability to exclusively target capillary pericytes may prove a precise and potentially powerful tool to treat microcirculation deficits, a common pathology in numerous diseases.


Asunto(s)
Velocidad del Flujo Sanguíneo/fisiología , Capilares/fisiología , Pericitos/fisiología , Flujo Sanguíneo Regional/fisiología , Retina/fisiología , Administración Oftálmica , Animales , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Capilares/química , Capilares/efectos de los fármacos , Ratones , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Pericitos/química , Pericitos/efectos de los fármacos , Flujo Sanguíneo Regional/efectos de los fármacos , Retina/química , Retina/citología , Retina/efectos de los fármacos , Tamoxifeno/administración & dosificación
10.
Virchows Arch ; 478(5): 841-849, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33244708

RESUMEN

Two cases of myopericytosis combined with pericytoma originating within the lung are reported. These are rare pulmonary tumors. The differential diagnosis for hemangiopericytoma and pericytic tumors with glomus elements is discussed. Both myopericytic lesions mimic other lesions, which are more commonly seen in the lung. Based on the expression of vascular growth factor receptors 2 and 3, an antiangiogenic therapy was suggested for the patient with the myopericytoma. A treatment with an angiogenesis inhibitor resulted in a regression of the tumor, but not the precursor lesion. Probably a more specific therapy using tyrosine kinase inhibitors for VEGFR2/3 might better control these myopericytic proliferations.


Asunto(s)
Neoplasias Pulmonares/patología , Pulmón/patología , Myopericytoma/patología , Pericitos/patología , Lesiones Precancerosas/patología , Adulto , Anciano de 80 o más Años , Inhibidores de la Angiogénesis/uso terapéutico , Biomarcadores de Tumor/análisis , Femenino , Humanos , Pulmón/química , Pulmón/diagnóstico por imagen , Pulmón/efectos de los fármacos , Neoplasias Pulmonares/química , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/tratamiento farmacológico , Myopericytoma/química , Myopericytoma/diagnóstico por imagen , Myopericytoma/tratamiento farmacológico , Pericitos/química , Pericitos/efectos de los fármacos , Lesiones Precancerosas/diagnóstico por imagen , Lesiones Precancerosas/tratamiento farmacológico , Lesiones Precancerosas/metabolismo , Resultado del Tratamiento , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis , Receptor 3 de Factores de Crecimiento Endotelial Vascular/análisis
11.
BMC Cancer ; 10: 659, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21118571

RESUMEN

BACKGROUND: Angiogenesis is important in the growth and metastasis of various kinds of solid tumors, including gastric cancers. The angiogenic process is triggered by several key growth factors, including vascular endothelial growth factor (VEGF)-A and platelet-derived growth factor (PDGF)-B, that are secreted by tumors. Our aim was to define: i) the expression pattern of VEGF-A and PDGF-B in tumor cells and the activation of PDGF receptor (PDGFR)-ß tyrosine kinase in stromal cells of human gastric adenocarcinomas; and ii) the relationship between VEGF-A and PDGF-B expression and microvessel density (MVD), to determine if there is a rationale for a new therapeutic strategy. METHODS: A series of 109 gastric adenocarcinoma cases that had undergone surgical resection was examined immunohistochemically using antibodies against VEGF-A, PDGF-B, and CD34, followed by further examination of PDGFR-ß phosphorylation by immunoblotting analysis. RESULTS: MVD was higher in diffuse-type than intestinal-type cancers (p < 0.001). VEGF-A overexpression correlated to PDGF-B overexpression in both the intestinal-type (p < 0.005) and diffuse-type (p < 0.0001) groups, indicating that VEGF-A and PDGF-B are secreted simultaneously in the same tumor, and may thus play important roles together in angiogenesis. However, several differences between intestinal-type and diffuse-type cancers were observed. In the diffuse-type cancer group, higher MVD was related to the PDGF-B proportion (p < 0.05) and VEGF-A overexpression (p < 0.05), but not to PDGF-B overexpression or the VEGF-A proportion. On the other hand, in the intestinal-type cancer group, higher MVD was correlated to overexpression (p < 0.005), intensity (p < 0.05), and proportion (p < 0.05) of PDGF-B, but not of VEGF-A. In addition, phosphorylation of PDGFR-ß was correlated with depth of cancer invasion at statistically significant level. CONCLUSIONS: Our results indicate that PDGF-B, which is involved in the maintenance of microvessels, plays a more important role in angiogenesis in intestinal-type gastric carcinomas than VEGF-A, which plays a key role mainly in the initiation of new blood vessel formation. In contrast, VEGF-A has a critical role for angiogenesis more in diffuse-type cancers, but less in those of intestinal type. Thus, a therapy targeting the PDGF-B signaling pathway could be effective for intestinal-type gastric carcinoma, whereas targeting VEGF-A or both VEGF-A and PDGF-B signaling pathways could be effective for diffuse-type gastric carcinomas.


Asunto(s)
Adenocarcinoma/química , Neovascularización Patológica/metabolismo , Proteínas Proto-Oncogénicas c-sis/análisis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/análisis , Neoplasias Gástricas/química , Factor A de Crecimiento Endotelial Vascular/análisis , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Adenocarcinoma/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD34/análisis , Distribución de Chi-Cuadrado , Femenino , Gastrectomía , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Inmunohistoquímica , Japón , Estimación de Kaplan-Meier , Masculino , Microvasos/química , Microvasos/patología , Persona de Mediana Edad , Invasividad Neoplásica , Neovascularización Patológica/patología , Pericitos/química , Pericitos/patología , Fosforilación , Neoplasias Gástricas/irrigación sanguínea , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Neoplasias Gástricas/cirugía , Resultado del Tratamiento
12.
Epigenomics ; 11(12): 1359-1370, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31357869

RESUMEN

Aim: Pericytes maintain homeostatic functions in the blood-brain barrier. N6-methyladenosine (m6A) is critical for various biological processes, but the role of mRNA m6A methylation in hypertension has not been fully elucidated. Methods: The m6A methylation levels of Wistar Kyoto rat pericytes and spontaneously hypertensive rat pericytes were detected via m6A high-throughput sequencing. Results: The m6A methylations were more enriched in the coding sequence region, 3'UTR and 5'UTR of mRNAs, with the m6A motifs being relatively conserved across the different conditions investigated. The average m6A abundance of spontaneously hypertensive rat pericytes exhibited global reductions in the pericytes. Conclusion: This study revealed the m6A landscapes and identified an epitranscriptomic mechanism during the development of mammalian hypertension.


Asunto(s)
Adenosina/análogos & derivados , Epigenómica/métodos , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Hipertensión/genética , Pericitos/química , Regiones no Traducidas 3' , Regiones no Traducidas 5' , Adenosina/química , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Metilación , Especificidad de Órganos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
13.
Acta Neuropathol Commun ; 7(1): 194, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31796114

RESUMEN

An impairment of amyloid ß-peptide (Aß) clearance is suggested to play a key role in the pathogenesis of sporadic Alzheimer's disease (AD). Amyloid degradation is mediated by various mechanisms including fragmentation by enzymes like neprilysin, matrix metalloproteinases (MMPs) and a recently identified amyloidolytic activity of ß-site amyloid precursor protein cleaving enzyme 1 (BACE1). BACE1 cleavage of Aß40 and Aß42 results in the formation of a common Aß34 intermediate which was found elevated in cerebrospinal fluid levels of patients at the earliest disease stages. To further investigate the role of Aß34 as a marker for amyloid clearance in AD, we performed a systematic and comprehensive analysis of Aß34 immunoreactivity in hippocampal and cortical post-mortem brain tissue from AD patients and non-demented elderly individuals. In early Braak stages, Aß34 was predominantly detectable in a subset of brain capillaries associated with pericytes, while in later disease stages, in clinically diagnosed AD, this pericyte-associated Aß34 immunoreactivity was largely lost. Aß34 was also detected in isolated human cortical microvessels associated with brain pericytes and its levels correlated with Aß40, but not with Aß42 levels. Moreover, a significantly decreased Aß34/Aß40 ratio was observed in microvessels from AD patients in comparison to non-demented controls suggesting a reduced proteolytic degradation of Aß40 to Aß34 in AD. In line with the hypothesis that pericytes at the neurovascular unit are major producers of Aß34, biochemical studies in cultured human primary pericytes revealed a time and dose dependent increase of Aß34 levels upon treatment with recombinant Aß40 peptides while Aß34 production was impaired when Aß40 uptake was reduced or BACE1 activity was inhibited. Collectively, our findings indicate that Aß34 is generated by a novel BACE1-mediated Aß clearance pathway in pericytes of brain capillaries. As amyloid clearance is significantly reduced in AD, impairment of this pathway might be a major driver of the pathogenesis in sporadic AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Capilares/metabolismo , Fragmentos de Péptidos/metabolismo , Pericitos/metabolismo , Proteolisis , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/análisis , Encéfalo/patología , Capilares/química , Capilares/patología , Células Cultivadas , Femenino , Humanos , Masculino , Fragmentos de Péptidos/análisis , Pericitos/química , Pericitos/patología
14.
Nat Protoc ; 14(11): 3059-3081, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31586162

RESUMEN

The study of cerebral microvessels is becoming increasingly important in a wide variety of conditions, such as stroke, sepsis, traumatic brain injury and neurodegenerative diseases. However, the molecular mechanisms underlying cerebral microvascular dysfunction in these conditions are largely unknown. The molecular characterization of cerebral microvessels in experimental disease models has been hindered by the lack of a standardized method to reproducibly isolate intact cerebral microvessels with consistent cellular compositions and without the use of enzymatic digestion, which causes undesirable molecular and metabolic changes. Herein, we describe an optimized protocol for microvessel isolation from mouse brain cortex that yields microvessel fragments with consistent populations of discrete blood-brain barrier (BBB) components (endothelial cells, pericytes and astrocyte end feet) while retaining high RNA integrity and protein post-translational modifications (e.g., phosphorylation). We demonstrate that this protocol allows the quantification of changes in gene expression in a disease model (stroke) and the activation of signaling pathways in mice subjected to drug administration in vivo. We also describe the isolation of genomic DNA (gDNA) and bisulfite treatment for the assessment of DNA methylation, as well as the optimization of chromatin extraction and shearing from cortical microvessels. This optimized protocol and the described applications should improve the understanding of the molecular mechanisms governing cerebral microvascular dysfunction, which may help in the development of novel therapies for stroke and other neurologic conditions.


Asunto(s)
Encéfalo/irrigación sanguínea , Microvasos/citología , Animales , Astrocitos/química , Astrocitos/citología , Barrera Hematoencefálica/química , Barrera Hematoencefálica/citología , Química Encefálica , ADN/análisis , Células Endoteliales/química , Células Endoteliales/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microvasos/química , Pericitos/química , Pericitos/citología , ARN/análisis
15.
Eur Rev Med Pharmacol Sci ; 23(14): 6018-6025, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31364104

RESUMEN

OBJECTIVE: To investigate the role of microRNA-15b in diabetic retinopathy and its underlying mechanism. MATERIALS AND METHODS: Diabetes rat model was established by streptozotocin injection. The mRNA expression of microRNA-15b in retinal capillary endothelial cells and pericytes of diabetic rats was detected by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). The mRNA and protein expressions of vascular endothelial growth factor A (VEGFA) were detected by qRT-PCR and Western blot, respectively. MicroRNA-15b mimics or inhibitor were transfected into retinal capillary endothelial cells and pericytes of diabetic rats, respectively. The mRNA expressions of microRNA-15b and VEGFA were detected by qRT-PCR. Cell counting kit-8 (CCK-8) assay was used to detect the proliferation of capillary endothelial cells and pericytes. Dual-Luciferase reporter gene assay was conducted to verify the binding condition of microRNA-15b and VEGFA. RNA immunoprecipitation (RIP) assay was performed to determine whether microRNA-15b could bind to AGO2. Rescue experiments were finally carried out by detecting the proliferation of retinal capillary endothelial cells and pericytes after downregulation or overexpression of microRNA-15b and VEGFA. RESULTS: The expression of microRNA-15b decreased, whereas VEGFA expression increased in retinal capillary endothelial cells and pericytes of diabetic rats. High expression of microRNA-15b in retinal capillary endothelial cells and pericytes resulted in VEGFA down-regulation and decreased proliferation. RIP assay results indicated that microRNA-15b could interact with AGO2. Additionally, Dual-Luciferase reporter gene assay showed that VEGFA is a direct target gene of microRNA-15b. VEGFA overexpression could reverse the inhibited proliferation of retinal capillary endothelial cells and pericytes induced by microRNA-15b overexpression. Similarly, VEGFA knockdown could reverse the effect of the low expression of microRNA-15b on the proliferation of retinal capillary endothelial cells and pericytes. CONCLUSIONS: Low expression of microRNA-15b in retinal capillary endothelial cells and pericytes of diabetic rats promotes the development of diabetic retinopathy by up-regulating VEGFA.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Retinopatía Diabética/genética , MicroARNs/genética , Pericitos/citología , Vasos Retinianos/citología , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Proliferación Celular , Células Cultivadas , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células Endoteliales/química , Células Endoteliales/citología , Pericitos/química , Ratas , Vasos Retinianos/química , Estreptozocina , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
J Control Release ; 302: 63-78, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-30930215

RESUMEN

The delivery of anticancer drugs is hampered by tumor vessels with abnormal structure and function, which requires that vessel normalization be mediated by pharmaceutics. The current strategies for vessel normalization focus on direct modulation of endothelial cells (ECs), which frequently affect vessels in normal tissues. Modulating EC-supporting cells, such as pericytes (PCs), is a new direction. Here, we produced a fusion protein, Z-TNFα, by fusing the platelet-derived growth factor receptor ß (PDGFRß)- antagonistic affibody ZPDGFRß to tumor necrosis factor α (TNFα). Owing to the affinity of fused ZPDGFRß for PDGFRß, Z-TNFα binds PDGFRß+ PCs but not PDGFRß- ECs. Low-dose (1 µg/mouse) Z-TNFα treatment remodeled the tumor vessels, thus reducing vessel permeability and increasing vessel perfusion. As a result, the Z-TNFα treatment improved the delivery of doxorubicin (DOX) and enhanced its antitumor effect, indicating that Z-TNFα induced normalization of tumor vessels. Mechanically, the tumor vessel normalization mediated by Z-TNFα might be attributed to the reduction of vascular endothelial growth factor (VEGF) secretion by PCs and the elevated expression of intercellular cell adhesion molecule-1 (ICAM-1) in PCs, which might suppress the proliferation and migration of ECs and simultaneously trigger interaction between perivascular macrophages and PCs. These results demonstrated that tumor-associated PCs could be considered novel target cells for vessel normalization, and Z-TNFα might be developed as a potential tool for antitumor combination therapy.


Asunto(s)
Antineoplásicos/química , Doxorrubicina/química , Portadores de Fármacos/química , Pericitos/química , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/química , Proteínas Recombinantes de Fusión/química , Factor de Necrosis Tumoral alfa/química , Animales , Antineoplásicos/farmacología , Transporte Biológico , Línea Celular Tumoral , Técnicas Químicas Combinatorias/métodos , Doxorrubicina/farmacología , Liberación de Fármacos , Células Endoteliales/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Endogámicos ICR , Neoplasias Experimentales , Inhibidores de Proteínas Quinasas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular
17.
Neuron ; 99(2): 362-375.e4, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-29937277

RESUMEN

Functional hyperemia, a regional increase of blood flow triggered by local neural activation, is used to map brain activity in health and disease. However, the spatial-temporal dynamics of functional hyperemia remain unclear. Two-photon imaging of the entire vascular arbor in NG2-creERT2;GCaMP6f mice shows that local synaptic activation, measured via oligodendrocyte precursor cell (OPC) Ca2+ signaling, generates a synchronous Ca2+ drop in pericytes and smooth muscle cells (SMCs) enwrapping all upstream vessels feeding the activated synapses. Surprisingly, the onset timing, direction, and amplitude of vessel diameter and blood velocity changes vary dramatically from juxta-synaptic capillaries back to the pial arteriole. These results establish a precise spatial-temporal sequence of vascular changes triggered by neural activity and essential for the interpretation of blood-flow-based imaging techniques such as BOLD-fMRI.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/fisiología , Hiperemia/fisiopatología , Piamadre/irrigación sanguínea , Piamadre/fisiología , Sinapsis/fisiología , Animales , Química Encefálica/fisiología , Hiperemia/diagnóstico , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal/métodos , Músculo Liso Vascular/química , Músculo Liso Vascular/fisiología , Pericitos/química , Pericitos/fisiología , Piamadre/química , Sinapsis/química
18.
Ann Anat ; 218: 124-128, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29678517

RESUMEN

Telocytes (TCs) are stromal cells with telopodes, which represent long, thin, moniliform cell processes; however, this morphological feature alone is insufficient to define a cell type. Specific markers of lymphatic endothelial cells (LECs), such as Prox-1, podoplanin (D2-40) or LYVE-1, are not usually tested in TCs. We thus aimed at performing a study in light microscopy to evaluate whether or not LECs could be mistaken for TCs. Therefore we used CD34, α-smooth muscle actin and D2-40 for an immunohistochemical study on archived paraffin-embedded samples of uterine leiomyoma. Lymphatic vessels were identified by the expression of D2-40, but on the microscopic slides, false spindle-shaped TCs appearances either corresponded to collapsed lymphatic lumina or were determined by grazing longitudinal cuts of lymphatics. It is therefore mandatory to check the expression of lymphatic markers in telocyte-like cells and, moreover, to carefully examine the bidimensional cuts in order to avoid false results.


Asunto(s)
Glicoproteínas de Membrana/química , Telocitos/patología , Telopodos/patología , Actinas/análisis , Antígenos CD34/análisis , Biomarcadores , Femenino , Humanos , Inmunohistoquímica , Leiomioma/química , Leiomioma/patología , Sistema Linfático/química , Sistema Linfático/patología , Persona de Mediana Edad , Adhesión en Parafina , Pericitos/química , Telocitos/química , Telopodos/química , Neoplasias Uterinas/química , Neoplasias Uterinas/patología
19.
J Mol Histol ; 49(4): 399-409, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29916090

RESUMEN

Follistatin like-1 (Fstl1) is a secreted glycoprotein and can be up-regulated by TGF-ß1. To better study the function of Fstl1 in lung development, we examined Fstl1 expression in the developing lung, in a cell type specific manner, using a tamoxifen inducible Fstl1-reporter mouse strain. Our results show that Fstl1 is ubiquitously expressed at saccular stage in the developing lung. At E18.5, Fstl1 expression is robust in most type of mesenchymal cells, including airway smooth muscle cells surrounding airways, vascular smooth muscle cells, endothelial cells, and vascular pericytes from blood vessel, but not PDGFRα+ fibroblasts in the distal alveolar sacs. Meanwhile, relative weak and sporadic signals of Fstl1 expression are observed in epithelium, including a subgroup of club cells in proximal airways and a few type II alveolar epithelial cells in distal airways. Our data help to understand the critical role of Fstl1 in lung development and lung disease pathogenesis.


Asunto(s)
Células/metabolismo , Proteínas Relacionadas con la Folistatina/metabolismo , Pulmón/embriología , Animales , Embrión de Mamíferos , Células Endoteliales/química , Células Endoteliales/metabolismo , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/metabolismo , Ratones , Miocitos del Músculo Liso/química , Miocitos del Músculo Liso/metabolismo , Pericitos/química , Pericitos/metabolismo , Sistema Respiratorio/citología , Distribución Tisular
20.
Haematologica ; 92(5): 597-604, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17488682

RESUMEN

BACKGROUND AND OBJECTIVES: Myelofibrotic bone marrow displays abnormal angiogenesis but the pathogenic mechanisms of this are poorly understood. Since pericyte abnormalities are described on solid tumor vessels we studied whether vessel morphology and pericyte coverage in bone marrow samples from patients with myelofibrosis differed from that in samples from controls. DESIGN AND METHODS: We assessed the microvascular density (MVD), vessel morphology and pericyte coverage in bone marrows from 19 myelofibrosis patients and nine controls. We also studied the same parameters in two mouse models of myelofibrosis, with genetic alterations affecting megakaryocyte differentiation (i.e. one model with low GATA-1 expression and the other with over-expression of thrombopoietin). RESULTS: In myelofibrotic marrows, MVD was 3.8-fold greater than in controls (p<0.001) and vessels displayed 5.9-fold larger mean perimeters (p<0.001). MVD was 1.8-fold greater in JAK2 V617F-positive than in negative patients (p=0.026). Moreover, 92+/-11 % of vessels in patients with myelofibrosis were pericyte-coated but only 51+/-20 % of vessels in controls (p<0.001). In the two mouse models of myelofibrosis caused by targeting megakaryocytopoesis, wide, pericyte-coated and morphologically aberrant vessels were detected. MVD was significantly greater in bone marrow and spleen samples from animals with myelofibrosis than in wild-type mice. INTERPRETATION AND CONCLUSIONS: We conclude that angiogenesis is similarly abnormal in human and murine myelofibrosis with intense pericyte coating, presumably related to abnormal megakaryocytopoiesis.


Asunto(s)
Médula Ósea/patología , Megacariocitos/patología , Neovascularización Patológica/patología , Pericitos/patología , Mielofibrosis Primaria/patología , Células del Estroma/patología , Actinas/análisis , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores , Médula Ósea/irrigación sanguínea , Capilares/patología , Modelos Animales de Enfermedad , Femenino , Fibrosis , Factor de Transcripción GATA1/deficiencia , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Neovascularización Patológica/etiología , Pericitos/química , Mielofibrosis Primaria/genética , Quimera por Radiación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Bazo/irrigación sanguínea , Bazo/patología , Células del Estroma/inmunología , Trombopoyetina/biosíntesis , Trombopoyetina/genética , Factor de Crecimiento Transformador beta1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA