Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 859
Filtrar
1.
PLoS Biol ; 22(4): e3002590, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38683849

RESUMEN

Brain pericytes are one of the critical cell types that regulate endothelial barrier function and activity, thus ensuring adequate blood flow to the brain. The genetic pathways guiding undifferentiated cells into mature pericytes are not well understood. We show here that pericyte precursor populations from both neural crest and head mesoderm of zebrafish express the transcription factor nkx3.1 develop into brain pericytes. We identify the gene signature of these precursors and show that an nkx3.1-, foxf2a-, and cxcl12b-expressing pericyte precursor population is present around the basilar artery prior to artery formation and pericyte recruitment. The precursors later spread throughout the brain and differentiate to express canonical pericyte markers. Cxcl12b-Cxcr4 signaling is required for pericyte attachment and differentiation. Further, both nkx3.1 and cxcl12b are necessary and sufficient in regulating pericyte number as loss inhibits and gain increases pericyte number. Through genetic experiments, we have defined a precursor population for brain pericytes and identified genes critical for their differentiation.


Asunto(s)
Encéfalo , Diferenciación Celular , Pericitos , Factores de Transcripción , Proteínas de Pez Cebra , Pez Cebra , Pericitos/metabolismo , Pericitos/citología , Animales , Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Encéfalo/metabolismo , Encéfalo/embriología , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Diferenciación Celular/genética , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/metabolismo , Cresta Neural/citología , Mesodermo/metabolismo , Mesodermo/citología , Transducción de Señal , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/genética
2.
Dev Dyn ; 253(5): 519-541, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38112237

RESUMEN

BACKGROUND: Mural cells are an essential perivascular cell population that associate with blood vessels and contribute to vascular stabilization and tone. In the embryonic zebrafish vasculature, pdgfrb and tagln are commonly used as markers for identifying pericytes and vascular smooth muscle cells. However, the overlapping and distinct expression patterns of these markers in tandem have not been fully described. RESULTS: Here, we used the Tg(pdgfrb:Gal4FF; UAS:RFP) and Tg(tagln:NLS-EGFP) transgenic lines to identify single- and double-positive perivascular cell populations on the cranial, axial, and intersegmental vessels between 1 and 5 days postfertilization. From this comparative analysis, we discovered two novel regions of tagln-positive cell populations that have the potential to function as mural cell precursors. Specifically, we found that the hypochord-a reportedly transient structure-contributes to tagln-positive cells along the dorsal aorta. We also identified a unique mural cell progenitor population that resides along the midline between the neural tube and notochord and contributes to intersegmental vessel mural cell coverage. CONCLUSION: Together, our findings highlight the variability and versatility of tracking both pdgfrb and tagln expression in mural cells of the developing zebrafish embryo and reveal unexpected embryonic cell populations that express pdgfrb and tagln.


Asunto(s)
Animales Modificados Genéticamente , Pericitos , Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/embriología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Pericitos/citología , Pericitos/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Vasos Sanguíneos/embriología , Vasos Sanguíneos/citología , Vasos Sanguíneos/metabolismo , Desarrollo Embrionario/fisiología
3.
Cells ; 12(8)2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-37190075

RESUMEN

Cardiac diseases are the foremost cause of morbidity and mortality worldwide. The heart has limited regenerative potential; therefore, lost cardiac tissue cannot be replenished after cardiac injury. Conventional therapies are unable to restore functional cardiac tissue. In recent decades, much attention has been paid to regenerative medicine to overcome this issue. Direct reprogramming is a promising therapeutic approach in regenerative cardiac medicine that has the potential to provide in situ cardiac regeneration. It consists of direct cell fate conversion of one cell type into another, avoiding transition through an intermediary pluripotent state. In injured cardiac tissue, this strategy directs transdifferentiation of resident non-myocyte cells (NMCs) into mature functional cardiac cells that help to restore the native tissue. Over the years, developments in reprogramming methods have suggested that regulation of several intrinsic factors in NMCs can help to achieve in situ direct cardiac reprogramming. Among NMCs, endogenous cardiac fibroblasts have been studied for their potential to be directly reprogrammed into both induced cardiomyocytes and induced cardiac progenitor cells, while pericytes can transdifferentiate towards endothelial cells and smooth muscle cells. This strategy has been indicated to improve heart function and reduce fibrosis after cardiac injury in preclinical models. This review summarizes the recent updates and progress in direct cardiac reprogramming of resident NMCs for in situ cardiac regeneration.


Asunto(s)
Transdiferenciación Celular , Técnicas de Reprogramación Celular , Reprogramación Celular , Fibroblastos , Cardiopatías , Corazón , Pericitos , Regeneración , Corazón/fisiología , Cardiopatías/terapia , Fibroblastos/citología , Fibroblastos/fisiología , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Pericitos/citología , Pericitos/fisiología , Células Endoteliales/citología , Células Endoteliales/fisiología , Humanos , Animales
4.
Cell Biochem Funct ; 40(5): 439-450, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35707856

RESUMEN

Regular soft tissue healing relies on the well-organized interaction of different stromal cell types with endothelial cells. However, spatiotemporal conditions might provoke high densities of one special stromal cell type, potentially leading to impaired healing. Detailed knowledge of the functions of rivaling stromal cell types aiming for tissue contraction and stabilization as well as vascular support is mandatory. By the application of an in vitro approach comprising the evaluation of cell proliferation, cell morphology, myofibroblastoid differentiation, and cytokine release, we verified a density-dependent modulation of these functions among juvenile and adult fibroblasts, pericytes, and adipose-derived stem cells during their interaction with microvascular endothelial cells in cocultures. Results indicate that juvenile fibroblasts rather support angiogenesis via paracrine regulation at the early stage of healing, a role potentially compromised in adult fibroblasts. In contrast, pericytes showed a more versatile character aiming at angiogenesis, vessel stabilization, and tissue contraction. Such a universal character was even more pronounced among adipose-derived stem cells. The explicit knowledge of the characteristic functions of stromal cell types is a prerequisite for the development of new analytical and therapeutic approaches for impaired soft tissue healing. The present study delivers new considerations concerning the roles of rivaling stromal cell types within a granulation tissue, pointing to extraordinary properties of pericytes and adipose-derived stem cells.


Asunto(s)
Células Endoteliales , Células del Estroma , Cicatrización de Heridas , Tejido Adiposo/citología , Recuento de Células , Células Endoteliales/citología , Fibroblastos/citología , Humanos , Neovascularización Patológica , Pericitos/citología , Células Madre/citología , Células del Estroma/citología
5.
Science ; 375(6584): eabi7377, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35084939

RESUMEN

Cerebrovascular diseases are a leading cause of death and neurologic disability. Further understanding of disease mechanisms and therapeutic strategies requires a deeper knowledge of cerebrovascular cells in humans. We profiled transcriptomes of 181,388 cells to define a cell atlas of the adult human cerebrovasculature, including endothelial cell molecular signatures with arteriovenous segmentation and expanded perivascular cell diversity. By leveraging this reference, we investigated cellular and molecular perturbations in brain arteriovenous malformations, which are a leading cause of stroke in young people, and identified pathologic endothelial transformations with abnormal vascular patterning and the ontology of vascularly derived inflammation. We illustrate the interplay between vascular and immune cells that contributes to brain hemorrhage and catalog opportunities for targeting angiogenic and inflammatory programs in vascular malformations.


Asunto(s)
Vasos Sanguíneos/citología , Encéfalo/irrigación sanguínea , Malformaciones Arteriovenosas Intracraneales/patología , Transcriptoma , Adulto , Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiología , Vasos Sanguíneos/fisiopatología , Células Cultivadas , Corteza Cerebral/irrigación sanguínea , Hemorragia Cerebral/patología , Hemorragia Cerebral/fisiopatología , Circulación Cerebrovascular , Células Endoteliales/citología , Células Endoteliales/patología , Células Endoteliales/fisiología , Fibroblastos/citología , Fibroblastos/fisiología , Humanos , Inflamación , Malformaciones Arteriovenosas Intracraneales/metabolismo , Monocitos/citología , Monocitos/fisiología , Músculo Liso Vascular/citología , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiología , Pericitos/citología , Pericitos/fisiología , RNA-Seq , Análisis de la Célula Individual
6.
Nat Protoc ; 17(1): 95-128, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34997242

RESUMEN

The blood-brain barrier (BBB) greatly restricts the entry of biological and engineered therapeutic molecules into the brain. Due to challenges in translating results from animal models to the clinic, relevant in vitro human BBB models are needed to assess pathophysiological molecular transport mechanisms and enable the design of targeted therapies for neurological disorders. This protocol describes an in vitro model of the human BBB self-assembled within microfluidic devices from stem-cell-derived or primary brain endothelial cells, and primary brain pericytes and astrocytes. This protocol requires 1.5 d for device fabrication, 7 d for device culture and up to 5 d for downstream imaging, protein and gene expression analyses. Methodologies to measure the permeability of any molecule in the BBB model, which take 30 min per device, are also included. Compared with standard 2D assays, the BBB model features relevant cellular organization and morphological characteristics, as well as values of molecular permeability within the range expected in vivo. These properties, coupled with a functional brain endothelial expression profile and the capability to easily test several repeats with low reagent consumption, make this BBB model highly suitable for widespread use in academic and industrial laboratories.


Asunto(s)
Barrera Hematoencefálica , Permeabilidad Capilar/fisiología , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas/instrumentación , Modelos Cardiovasculares , Astrocitos/citología , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/fisiología , Encéfalo/citología , Células Cultivadas , Células Endoteliales/citología , Humanos , Pericitos/citología
7.
J Neurosci ; 42(3): 362-376, 2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-34819341

RESUMEN

Multifaceted microglial functions in the developing brain, such as promoting the differentiation of neural progenitors and contributing to the positioning and survival of neurons, have been progressively revealed. Although previous studies have noted the relationship between vascular endothelial cells and microglia in the developing brain, little attention has been given to the importance of pericytes, the mural cells surrounding endothelial cells. In this study, we attempted to dissect the role of pericytes in microglial distribution and function in developing mouse brains. Our immunohistochemical analysis showed that approximately half of the microglia attached to capillaries in the cerebral walls. Notably, a magnified observation of the position of microglia, vascular endothelial cells and pericytes demonstrated that microglia were preferentially associated with pericytes that covered 79.8% of the total capillary surface area. Through in vivo pericyte depletion induced by the intraventricular administration of a neutralizing antibody against platelet-derived growth factor receptor (PDGFR)ß (clone APB5), we found that microglial density was markedly decreased compared with that in control antibody-treated brains because of their low proliferative capacity. Moreover, in vitro coculture of isolated CD11b+ microglia and NG2+PDGFRα- cells, which are mostly composed of pericytes, from parenchymal cells indicated that pericytes promote microglial proliferation via the production of soluble factors. Furthermore, pericyte depletion by APB5 treatment resulted in a failure of microglia to promote the differentiation of neural stem cells into intermediate progenitors. Taken together, our findings suggest that pericytes facilitate microglial homeostasis in the developing brains, thereby indirectly supporting microglial effects on neural progenitors.SIGNIFICANCE STATEMENT This study highlights the novel effect of pericytes on microglia in the developing mouse brain. Through multiple analyses using an in vivo pericyte depletion mouse model and an in vitro coculture study of isolated pericytes and microglia from parenchymal cells, we demonstrated that pericytes contribute to microglial proliferation and support microglia in efficiently promoting the differentiation of neural stem cells into intermediate progenitors. Our present data provide evidence that pericytes function not only in the maintenance of cerebral microcirculation and blood brain barrier (BBB) integrity but also in microglial homeostasis in the developing cerebral walls. These findings will expand our knowledge and help elucidate the mechanism of brain development both in healthy and disease conditions.


Asunto(s)
Corteza Cerebral/citología , Homeostasis/fisiología , Microglía/citología , Células-Madre Neurales/citología , Pericitos/citología , Animales , Anticuerpos Neutralizantes , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/embriología , Permeabilidad Capilar/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/embriología , Ácido Clodrónico/farmacología , Homeostasis/efectos de los fármacos , Liposomas , Ratones , Microglía/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Pericitos/efectos de los fármacos , Receptor beta de Factor de Crecimiento Derivado de Plaquetas
8.
Arterioscler Thromb Vasc Biol ; 42(2): 205-222, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34879709

RESUMEN

OBJECTIVE: We sought to determine how endothelial cell (EC) expression of the activating k-Ras (kirsten rat sarcoma 2 viral oncogene homolog) mutation, k-RasV12, affects their ability to form lumens and tubes and interact with pericytes during capillary assembly Approach and Results: Using defined bioassays where human ECs undergo observable tubulogenesis, sprouting behavior, pericyte recruitment to EC-lined tubes, and pericyte-induced EC basement membrane deposition, we assessed the impact of EC k-RasV12 expression on these critical processes that are necessary for proper capillary network formation. This mutation, which is frequently seen in human ECs within brain arteriovenous malformations, was found to markedly accentuate EC lumen formation mechanisms, with strongly accelerated intracellular vacuole formation, vacuole fusion, and lumen expansion and with reduced sprouting behavior, leading to excessively widened tube networks compared with control ECs. These abnormal tubes demonstrate strong reductions in pericyte recruitment and pericyte-induced EC basement membranes compared with controls, with deficiencies in fibronectin, collagen type IV, and perlecan deposition. Analyses of signaling during tube formation from these k-RasV12 ECs reveals strong enhancement of Src (Src proto-oncogene, non-receptor tyrosine kinase), Pak2 (P21 [RAC1 (Rac family small GTPase 1)] activated kinase 2), b-Raf (v-raf murine sarcoma viral oncogene homolog B1), Erk (extracellular signal-related kinase), and Akt (AK strain transforming) activation and increased expression of PKCε (protein kinase C epsilon), MT1-MMP (membrane-type 1 matrix metalloproteinase), acetylated tubulin and CDCP1 (CUB domain-containing protein 1; most are known EC lumen regulators). Pharmacological blockade of MT1-MMP, Src, Pak, Raf, Mek (mitogen-activated protein kinase) kinases, Cdc42 (cell division cycle 42)/Rac1, and Notch markedly interferes with lumen and tube formation from these ECs. CONCLUSIONS: Overall, this novel work demonstrates that EC expression of k-RasV12 disrupts capillary assembly due to markedly excessive lumen formation coupled with strongly reduced pericyte recruitment and basement membrane deposition, which are critical pathogenic features predisposing the vasculature to develop arteriovenous malformations.


Asunto(s)
Membrana Basal/citología , Capilares/fisiología , Células Endoteliales/citología , Neovascularización Fisiológica , Pericitos/citología , Proteínas Proto-Oncogénicas p21(ras)/genética , Membrana Basal/metabolismo , Línea Celular , Células Endoteliales/metabolismo , Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mutación , Pericitos/metabolismo
9.
Exp Cell Res ; 411(1): 112983, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34921827

RESUMEN

After the severe initial insults of acute kidney injury, progressive kidney tubulointerstitial fibrosis may occur, the peritubular capillary (PTC) rarefaction plays a key role in the disease progression. However, the mechanisms of PTC damage were not fully understood and potential therapeutic interventions were not explored. Previous studies of our research team and others in this field suggested that bone marrow-derived mesenchymal stem cells (BMSCs) transplanted into the AKI rat model may preserve the kidney function and pathological changes. In the current study, with the ischemia/reperfusion AKI rat model, we revealed that BMSCs transplantation attenuated the renal function decrease in the AKI model through preserving the peritubular capillaries (PTCs) function. The density of PTCs is maintained by BMSCs transplantation in the AKI model, detachment and relocation of pericytes in the PTCs diminished. Then we established that BMSCs transplantation may attenuate the renal fibrosis and preserve the kidney function after AKI by repairing the PTCs. Improving the vitality of pericytes, suppressing the detachment and trans-differentiation of pericytes, directly differentiation of BMSCs into pericytes by BMSCs transplantation all participate in the PTC repair. Through these processes, BMSCs rescued the microvascular damage and improved the density of PTCs. As a result, a preliminary conclusion can be reached that BMSCs transplantation can be an effective therapy for delaying renal fibrosis after AKI.


Asunto(s)
Lesión Renal Aguda/complicaciones , Endotelio Vascular/citología , Fibrosis/terapia , Enfermedades Renales/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Pericitos/citología , Animales , Fibrosis/etiología , Fibrosis/metabolismo , Fibrosis/patología , Enfermedades Renales/etiología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Masculino , Ratas , Ratas Sprague-Dawley
10.
Int J Mol Sci ; 22(24)2021 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-34948214

RESUMEN

The term "stromal cells" refers to a highly heterogeneous class of connective tissue cells that build the infrastructure of any organ and fulfill a variety of fundamental roles in health and disease [...].


Asunto(s)
Células del Estroma/citología , Células del Tejido Conectivo/citología , Fibroblastos/citología , Humanos , Células Madre Mesenquimatosas/citología , Pericitos/citología , Telocitos/citología
11.
Neurobiol Dis ; 161: 105561, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34780863

RESUMEN

Coronavirus disease 19 (COVID-19) is a respiratory illness caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). COVID-19 pathogenesis causes vascular-mediated neurological disorders via elusive mechanisms. SARS-CoV-2 infects host cells via the binding of viral Spike (S) protein to transmembrane receptor, angiotensin-converting enzyme 2 (ACE2). Although brain pericytes were recently shown to abundantly express ACE2 at the neurovascular interface, their response to SARS-CoV-2 S protein is still to be elucidated. Using cell-based assays, we found that ACE2 expression in human brain vascular pericytes was increased upon S protein exposure. Pericytes exposed to S protein underwent profound phenotypic changes associated with an elongated and contracted morphology accompanied with an enhanced expression of contractile and myofibrogenic proteins, such as α-smooth muscle actin (α-SMA), fibronectin, collagen I, and neurogenic locus notch homolog protein-3 (NOTCH3). On the functional level, S protein exposure promoted the acquisition of calcium (Ca2+) signature of contractile ensheathing pericytes characterized by highly regular oscillatory Ca2+ fluctuations. Furthermore, S protein induced lipid peroxidation, oxidative and nitrosative stress in pericytes as well as triggered an immune reaction translated by activation of nuclear factor-kappa-B (NF-κB) signaling pathway, which was potentiated by hypoxia, a condition associated with vascular comorbidities that exacerbate COVID-19 pathogenesis. S protein exposure combined to hypoxia enhanced the production of pro-inflammatory cytokines involved in immune cell activation and trafficking, namely macrophage migration inhibitory factor (MIF). Using transgenic mice expressing the human ACE2 that recognizes S protein, we observed that the intranasal infection with SARS-CoV-2 rapidly induced hypoxic/ischemic-like pericyte reactivity in the brain of transgenic mice, accompanied with an increased vascular expression of ACE2. Moreover, we found that SARS-CoV-2 S protein accumulated in the intranasal cavity reached the brain of mice in which the nasal mucosa is deregulated. Collectively, these findings suggest that SARS-CoV-2 S protein impairs the vascular and immune regulatory functions of brain pericytes, which may account for vascular-mediated brain damage. Our study provides a better understanding for the mechanisms underlying cerebrovascular disorders in COVID-19, paving the way to develop new therapeutic interventions.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Encéfalo/metabolismo , COVID-19/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia/metabolismo , Inflamación/metabolismo , Pericitos/metabolismo , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Actinas/metabolismo , Enzima Convertidora de Angiotensina 2/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/genética , Animales , Encéfalo/irrigación sanguínea , COVID-19/fisiopatología , Señalización del Calcio , Colágeno Tipo I/metabolismo , Fibronectinas/metabolismo , Humanos , Hipoxia-Isquemia Encefálica/fisiopatología , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/genética , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Miofibroblastos , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Mucosa Nasal , Estrés Nitrosativo , Estrés Oxidativo , Pericitos/citología , Pericitos/efectos de los fármacos , Fenotipo , Receptor Notch3/metabolismo , Receptores de Coronavirus/efectos de los fármacos , Receptores de Coronavirus/genética , Receptores de Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/farmacología
12.
Dis Markers ; 2021: 7124835, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34630739

RESUMEN

Inner blood-retina barrier (iBRB) is primarily formed of retinal microvascular endothelial cells (ECs) with tight junctions, which are surrounded and supported by retinal microvascular pericytes (RMPs) and basement membrane. Pericytes are believed to be critically involved in the physiology and pathology of iBRB. However, the underlying mechanism remains to be fully elucidated. We developed a novel in vitro iBRB model which was composed of primary cultures of rat retinal ECs and RMPs based on Transwell system. We tested the involvement of pericytes in the migration and invasion of ECs, examined the expression and activity of matrix metalloproteinase- (MMP-) 2/MMP-9 in the culture, evaluated the TEER and permeability of iBRB, and assessed the expression of ZO-1, occludin, claudin-5, and VE-cadherin of endothelial junctions. We found that RMPs with indirect contact of ECs can increase the expression of MMP-2 and upgrade the activity of MMP-2/9 in the coculture, which subsequently decreased TJ protein abundance of ZO-1 and occludin in ECs, promoted the migration of ECs, and finally reduced the integrity of iBRB. Taken together, our data show that RMP relative location with ECs is involved in the integrity of iBRB via MMP-2/9 and has important implications for treating diabetic retinopathy and other retinal disorders involving iBRB dysfunction.


Asunto(s)
Barrera Hematorretinal/citología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Pericitos/citología , Retina/citología , Animales , Barrera Hematorretinal/metabolismo , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Masculino , Modelos Biológicos , Pericitos/metabolismo , Cultivo Primario de Células , Ratas , Retina/metabolismo , Uniones Estrechas/metabolismo
13.
Exp Eye Res ; 212: 108771, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34624336

RESUMEN

The choriocapillaris (CC), the capillary bed in the choroid, essentially nourishes the photoreceptor cells. Its damage in aging and age-related diseases significantly influences the survival of the photoreceptor cells. Earlier reports implicated endothelial loss in aged and diseased CC; however, age-related pericyte changes and their contribution in CC death remain unknown. We examined human donor eyes (age: 56-94 years; N = 24), and found that CC pericyte damage preceded endothelial changes. With aging (>70 years), the sub-macular choroid accumulated debris in Bruch's membrane (BM). Of the debris content, the long-spaced collagens had a tendency to settle over the capillary basal lamina (BL), and this often resulted in endothelial projection into capillary lumen. Between 75 and 83 years, pericytes contained dark mitochondria, and their processes facing the BM debris showed partial loss of BL and intermediate filaments (IFs), when the endothelium remained unaltered. The endothelial changes appeared beyond 83 years, the abundance of IFs and autophagy reinforced their survival until late aging. TUNEL+ pericytes, and immunoreactivity to carboxymethyl lysine and 4-hydroxy 2-nonenal, but no nitro-tyrosine, was detected in aged CC walls. Iba-1+ dystrophic microglia were present in the vicinity of the CC. Our data indicate that (1) BM debris exerts pressure on the CC, leading to the damage of the capillary BL and pericyte processes (2) loss of IFs results in early pericyte destabilization (3) capillary wall undergoes lipid peroxidative and glycative damage, and (4) pericyte damage leads to late endothelial changes and ultimately CC loss. Future research should explore the normal ways of pericyte maintenance in the aging nervous system.


Asunto(s)
Envejecimiento/fisiología , Coroides/citología , Endotelio Vascular/citología , Estrés Oxidativo/fisiología , Pericitos/citología , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Etiquetado Corte-Fin in Situ , Masculino , Persona de Mediana Edad
14.
Cells ; 10(9)2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34571892

RESUMEN

An important objective of vascularized tissue regeneration is to develop agents for osteonecrosis. We aimed to identify the pro-angiogenic and osteogenic efficacy of adipose tissue-derived (AD) pericytes combined with Nel-like protein-1 (NELL-1) to investigate the therapeutic effects on osteonecrosis. Tube formation and cell migration were assessed to determine the pro-angiogenic efficacy. Vessel formation was evaluated in vivo using the chorioallantoic membrane assay. A mouse model with a 2.5 mm necrotic bone fragment in the femoral shaft was used as a substitute for osteonecrosis in humans. Bone formation was assessed radiographically (plain radiographs, three-dimensional images, and quantitative analyses), and histomorphometric analyses were performed. To identify factors related to the effects of NELL-1, analysis using microarrays, qRT-PCR, and Western blotting was performed. The results for pro-angiogenic efficacy evaluation identified synergistic effects of pericytes and NELL-1 on tube formation, cell migration, and vessel formation. For osteogenic efficacy analysis, the mouse model for osteonecrosis was treated in combination with pericytes and NELL-1, and the results showed maximum bone formation using radiographic images and quantitative analyses, compared with other treatment groups and showed robust bone and vessel formation using histomorphometric analysis. We identified an association between FGF2 and the effects of NELL-1 using array-based analysis. Thus, combinatorial therapy using AD pericytes and NELL-1 may have potential as a novel treatment for osteonecrosis.


Asunto(s)
Tejido Adiposo/citología , Proteínas de Unión al Calcio/metabolismo , Neovascularización Fisiológica , Osteogénesis , Osteonecrosis/terapia , Pericitos/citología , Tejido Adiposo/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Apoptosis , Proteínas de Unión al Calcio/genética , Movimiento Celular , Proliferación Celular , Células Cultivadas , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Osteonecrosis/etiología , Osteonecrosis/metabolismo , Osteonecrosis/patología , Pericitos/metabolismo
15.
Cells ; 10(9)2021 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-34571963

RESUMEN

Stroke is the third leading cause of mortality in women and it kills twice as many women as breast cancer. A key role in the pathophysiology of stroke plays the disruption of the blood-brain barrier (BBB) within the neurovascular unit. While estrogen induces vascular protective actions, its influence on stroke remains unclear. Moreover, experiments assessing its impact on endothelial cells to induce barrier integrity are non-conclusive. Since pericytes play an active role in regulating BBB integrity and function, we hypothesize that estradiol may influence BBB by regulating their activity. In this study using human brain vascular pericytes (HBVPs) we investigated the impact of estradiol on key pericyte functions known to influence BBB integrity. HBVPs expressed estrogen receptors (ER-α, ER-ß and GPER) and treatment with estradiol (10 nM) inhibited basal cell migration but not proliferation. Since pericyte migration is a hallmark for BBB disruption following injury, infection and inflammation, we investigated the effects of estradiol on TNFα-induced PC migration. Importantly, estradiol prevented TNFα-induced pericyte migration and this effect was mimicked by PPT (ER-α agonist) and DPN (ER-ß agonist), but not by G1 (GPR30 agonist). The modulatory effects of estradiol were abrogated by MPP and PHTPP, selective ER-α and ER-ß antagonists, respectively, confirming the role of ER-α and ER-ß in mediating the anti-migratory actions of estrogen. To delineate the intracellular mechanisms mediating the inhibitory actions of estradiol on PC migration, we investigated the role of AKT and MAPK activation. While estradiol consistently reduced the TNFα-induced MAPK and Akt phosphorylation, only the inhibition of MAPK, but not Akt, significantly abrogated the migratory actions of TNFα. In transendothelial electrical resistance measurements, estradiol induced barrier function (TEER) in human brain microvascular endothelial cells co-cultured with pericytes, but not in HBMECs cultured alone. Importantly, transcriptomics analysis of genes modulated by estradiol in pericytes showed downregulation of genes known to increase cell migration and upregulation of genes known to inhibit cell migration. Taken together, our findings provide the first evidence that estradiol modulates pericyte activity and thereby improves endothelial integrity.


Asunto(s)
Encéfalo/irrigación sanguínea , Movimiento Celular/efectos de los fármacos , Estradiol/farmacología , Perfilación de la Expresión Génica , Pericitos/citología , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Estrógenos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
16.
J Cell Mol Med ; 25(21): 10088-10100, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34592781

RESUMEN

It was shown that endothelial progenitor cells (EPCs) have bidirectional differentiation potential and thus perform different biological functions. The purpose of this study was to investigate the effects of slight up-regulation of the Kir2.1 channel on EPC transdifferentiation and the potential mechanism on cell function and transformed cell type. First, we found that the slight up-regulation of Kir2.1 expression promoted the expression of the stem cell stemness factors ZFX and NS and inhibited the expression of senescence-associated ß-galactosidase. Further studies showed the slightly increased expression of Kir2.1 could also improve the expression of pericyte molecular markers NG2, PDGFRß and Desmin. Moreover, adenovirus-mediated Kir2.1 overexpression had an enhanced contractile response to norepinephrine of EPCs. These results suggest that the up-regulated expression of the Kir2.1 channel promotes EPC transdifferentiation into a pericyte phenotype. Furthermore, the mechanism of EPC transdifferentiation to mesenchymal cells (pericytes) was found to be closely related to the channel functional activity of Kir2.1 and revealed that this channel could promote EPC EndoMT by activating the Akt/mTOR/Snail signalling pathway. Overall, this study suggested that in the early stage of inflammatory response, regulating the Kir2.1 channel expression affects the biological function of EPCs, thereby determining the maturation and stability of neovascularization.


Asunto(s)
Diferenciación Celular , Células Progenitoras Endoteliales/metabolismo , Pericitos/metabolismo , Canales de Potasio de Rectificación Interna/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Biomarcadores , Autorrenovación de las Células , Células Cultivadas , Senescencia Celular , Desmina/metabolismo , Células Progenitoras Endoteliales/citología , Modelos Biológicos , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Pericitos/citología , Canales de Potasio de Rectificación Interna/metabolismo , Ratas , Transducción de Señal
17.
Cells ; 10(7)2021 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-34359953

RESUMEN

Many pathological conditions of the brain are associated with structural abnormalities within the neurovascular system and linked to pericyte (PC) loss and/or dysfunction. Since crosstalk between endothelial cells (ECs) and PCs greatly impacts the function of the blood-brain barrier (BBB), effects of PCs on endothelial integrity and function have been investigated extensively. However, the impact of ECs on the function and activity of PCs remains largely unknown. Hence, using co-cultures of human brain vascular PCs with human cerebral microvascular ECs on opposite sides of porous Transwell inserts which facilitates direct EC-PC contact and improves EC barrier function, we analyzed EC-driven transcriptomic changes in PCs using microarrays and changes in cytokines/chemokines using proteome arrays. Gene expression analysis (GEA) in PCs co-cultured with ECs versus PCs cultured alone showed significant upregulation of 1'334 genes and downregulation of 964 genes. GEA in co-cultured PCs revealed increased expression of five prominent PC markers as well as soluble factors, such as transforming growth factor beta, fibroblast growth factor, angiopoietin 1, brain-derived neurotrophic factor, all of which are involved in EC-PC crosstalk and BBB induction. Pathway enrichment analysis of modulated genes showed a strong impact on many inflammatory and extracellular matrix (ECM) pathways including interferon and interleukin signaling, TGF-ß and interleukin-1 regulation of ECM, as well as on the mRNA processing pathway. Interestingly, while co-culture induced the mRNA expression of many chemokines and cytokines, including several CCL- and CXC-motif ligands and interleukins, we observed a decreased expression of the same inflammatory mediators on the protein level. Importantly, in PCs, ECs significantly induced interferon associated proteins (IFIT1, IFI44L, IF127, IFIT3, IFI6, IFI44) with anti-viral actions; downregulated prostaglandin E receptor 2 (prevent COX-2 mediated BBB damage); upregulated fibulin-3 and connective tissue growth factor essential for BBB integrity; and multiple ECMs (collagens and integrins) that inhibit cell migration. Our findings suggest that via direct contact, ECs prime PCs to induce molecules to promote BBB integrity and cell survival during infection and inflammatory insult. Taken together, we provide first evidence that interaction with ECs though porous membranes induces major changes in the transcriptomic and proteomic profile of PCs. ECs influence genes involved in diverse aspects of PC function including PC maturation, cell survival, anti-viral defense, blood flow regulation, immuno-modulation and ECM deposition.


Asunto(s)
Encéfalo/metabolismo , Movimiento Celular/fisiología , Células Endoteliales/metabolismo , Pericitos/citología , Transporte Biológico/fisiología , Endotelio Vascular/metabolismo , Humanos
18.
Nat Biomed Eng ; 5(8): 847-863, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34385693

RESUMEN

The therapeutic efficacy of stem cells transplanted into an ischaemic brain depends primarily on the responses of the neurovascular unit. Here, we report the development and applicability of a functional neurovascular unit on a microfluidic chip as a microphysiological model of ischaemic stroke that recapitulates the function of the blood-brain barrier as well as interactions between therapeutic stem cells and host cells (human brain microvascular endothelial cells, pericytes, astrocytes, microglia and neurons). We used the model to track the infiltration of a number of candidate stem cells and to characterize the expression levels of genes associated with post-stroke pathologies. We observed that each type of stem cell showed unique neurorestorative effects, primarily by supporting endogenous recovery rather than through direct cell replacement, and that the recovery of synaptic activities is correlated with the recovery of the structural and functional integrity of the neurovascular unit rather than with the regeneration of neurons.


Asunto(s)
Accidente Cerebrovascular Isquémico/terapia , Dispositivos Laboratorio en un Chip , Trasplante de Células Madre , Astrocitos/citología , Astrocitos/metabolismo , Barrera Hematoencefálica/química , Barrera Hematoencefálica/metabolismo , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Microglía/citología , Microglía/metabolismo , Microvasos/citología , Modelos Biológicos , Neuronas/citología , Neuronas/metabolismo , Pericitos/citología , Pericitos/metabolismo , Células Madre/citología , Células Madre/metabolismo
19.
BMC Urol ; 21(1): 103, 2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-34362357

RESUMEN

BACKGROUND: Peyronie's disease (PD) is a severe fibrotic disease of the tunica albuginea that causes penis curvature and leads to penile pain, deformity, and erectile dysfunction. The role of pericytes in the pathogenesis of fibrosis has recently been determined. Extracellular vesicle (EV)-mimetic nanovesicles (NVs) have attracted attention regarding intercellular communication between cells in the field of fibrosis. However, the global gene expression of pericyte-derived EV-mimetic NVs (PC-NVs) in regulating fibrosis remains unknown. Here, we used RNA-sequencing technology to investigate the potential target genes regulated by PC-NVs in primary fibroblasts derived from human PD plaque. METHODS: Human primary fibroblasts derived from normal and PD patients was cultured and treated with cavernosum pericytes isolated extracellular vesicle (EV)-mimetic nanovesicles (NVs). A global gene expression RNA-sequencing assay was performed on normal fibroblasts, PD fibroblasts, and PD fibroblasts treated with PC-NVs. Reverse transcription polymerase chain reaction (RT-PCR) was used for sequencing data validation. RESULTS: A total of 4135 genes showed significantly differential expression in the normal fibroblasts, PD fibroblasts, and PD fibroblasts treated with PC-NVs. However, only 91 contra-regulated genes were detected among the three libraries. Furthermore, 20 contra-regulated genes were selected and 11 showed consistent changes in the RNA-sequencing assay, which were validated by RT-PCR. CONCLUSION: The gene expression profiling results suggested that these validated genes may be good targets for understanding potential mechanisms and conducting molecular studies into PD.


Asunto(s)
Vesículas Extracelulares/genética , Fibroblastos/citología , Perfilación de la Expresión Génica , Induración Peniana/genética , ARN/análisis , Análisis de Secuencia de ARN , Células Cultivadas , Vesículas Extracelulares/metabolismo , Biblioteca de Genes , Humanos , Masculino , Induración Peniana/patología , Pene/citología , Pericitos/citología , ARN/metabolismo
20.
Int J Mol Sci ; 22(16)2021 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-34445338

RESUMEN

Recently, another new cell type was found in the perivascular space called a novel desmin-immunopositive perivascular (DIP) cell. However, the differences between this novel cell type and other nonhormone-producing cells have not been clarified. Therefore, we introduced several microscopic techniques to gain insight into the morphological characteristics of this novel DIP cell. We succeeded in identifying novel DIP cells under light microscopy using desmin immunocryosection, combining resin embedding blocks and immunoelectron microscopy. In conventional transmission electron microscopy, folliculostellate cells, capsular fibroblasts, macrophages, and pericytes presented a flat cisternae of rough endoplasmic reticulum, whereas those of novel DIP cells had a dilated pattern. The number of novel DIP cells was greatest in the intact rats, though nearly disappeared under prolactinoma conditions. Additionally, focused ion beam scanning electron microscopy showed that these novel DIP cells had multidirectional processes and some processes reached the capillary, but these processes did not tightly wrap the vessel, as is the case with pericytes. Interestingly, we found that the rough endoplasmic reticulum was globular and dispersed throughout the cytoplasmic processes after three-dimensional reconstruction. This study clearly confirms that novel DIP cells are a new cell type in the rat anterior pituitary gland, with unique characteristics.


Asunto(s)
Desmina/metabolismo , Pericitos , Adenohipófisis/diagnóstico por imagen , Animales , Desmina/análisis , Inmunohistoquímica , Masculino , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Pericitos/citología , Pericitos/metabolismo , Adenohipófisis/citología , Adenohipófisis/metabolismo , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...