Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 623
Filtrar
1.
Clin Sci (Lond) ; 138(16): 1009-1022, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39106080

RESUMEN

Diabetes mediates endothelial dysfunction and increases the risk of Alzheimer's disease and related dementias. Diabetes also dysregulates the ET system. ET-1-mediated constriction of brain microvascular pericytes (BMVPCs) has been shown to contribute to brain hypoperfusion. Cellular senescence, a process that arrests the proliferation of harmful cells and instigates phenotypical changes and proinflammatory responses in endothelial cells that impact their survival and function. Thus, we hypothesized that ET-1 mediates BMVPC senescence and phenotypical changes in diabetes-like conditions. Human BMVPCs were incubated in diabetes-like conditions with or without ET-1 (1 µmol/L) for 3 and 7 days. Hydrogen peroxide (100 µmol/L H2O2) was used as a positive control for senescence and to mimic ischemic conditions. Cells were stained for senescence-associated ß-galactosidase or processed for immunoblotting and quantitative real-time PCR analyses. In additional experiments, cells were stimulated with ET-1 in the presence or absence of ETA receptor antagonist BQ-123 (20 µmol/L) or ETB receptor antagonist BQ-788 (20 µmol/L). ET-1 stimulation increased ß-galactosidase accumulation which was prevented by BQ-123. ET-1 also increased traditional senescence marker p16 protein and pericyte-specific senescence markers, TGFB1i1, PP1CA, and IGFBP7. Furthermore, ET-1 stimulated contractile protein α-SMA and microglial marker ostepontin in high glucose suggesting a shift toward an ensheathing or microglia-like phenotype. In conclusion, ET-1 triggers senescence, alters ETA and ETB receptors, and causes phenotypical changes in BMVPCs under diabetes-like conditions. These in vitro findings need to be further studied in vivo to establish the role of ETA receptors in the progression of pericyte senescence and phenotypical changes in VCID.


Asunto(s)
Encéfalo , Senescencia Celular , Endotelina-1 , Pericitos , Receptor de Endotelina A , Humanos , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Diabetes Mellitus/metabolismo , Endotelina-1/metabolismo , Endotelina-1/farmacología , Pericitos/metabolismo , Pericitos/efectos de los fármacos , Pericitos/patología , Fenotipo , Receptor de Endotelina A/metabolismo , Receptor de Endotelina A/genética
2.
PLoS One ; 19(8): e0308132, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39106252

RESUMEN

To investigate the sex-dependent differentiation of Sox10 cells and their response to pathological conditions such as lipopolysaccharide (LPS) exposure or ischemia, we utilized Sox10 Cre-ERT2, tdTomato mice. Tamoxifen administration induced the expression of red fluorescent protein (RFP) in these cells, facilitating their subsequent tracking and analysis after LPS injection and ischemia via immunofluorescence staining. Propidium iodide (PI) was injected to label necrotic cells following LPS administration. We found that the conversion of Sox10 cells to pericytes in female mice was significantly higher than in male mice, especially in those exposed to LPS. After LPS injection, the number of PI+ necrotic cells were significantly greater in females than in males. Moreover, RFP+ cells did not co-localize with glial fibrillary acidic protein (GFAP) or cluster of differentiation 11b (CD11b). Similarly, after brain ischemia, RFP+ cells did not express cluster of differentiation 13 (CD13), neuronal nuclei (NeuN), GFAP, or ionised calcium binding adaptor molecule 1 (Iba-1). These findings indicate that the conversion of Sox10 cells to pericytes following LPS exposure is sex-dependent, with neither male nor female groups showing differentiation into other cell types after LPS exposure or under ischemic conditions. The differences in LPS-induced necrosis of pericytes between sexes may explain the variations in the conversion of Sox10 cells to pericytes in both sexes.


Asunto(s)
Lipopolisacáridos , Oligodendroglía , Pericitos , Factores de Transcripción SOXE , Animales , Femenino , Masculino , Pericitos/metabolismo , Pericitos/efectos de los fármacos , Ratones , Factores de Transcripción SOXE/metabolismo , Oligodendroglía/metabolismo , Oligodendroglía/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Caracteres Sexuales , Factores Sexuales , Ratones Transgénicos
3.
Redox Rep ; 29(1): 2382943, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39092597

RESUMEN

OBJECTIVES: Diabetes is closely linked to hearing loss, yet the exact mechanisms remain unclear. Cochlear stria vascularis and pericytes (PCs) are crucial for hearing. This study investigates whether high glucose induces apoptosis in the cochlear stria vascularis and pericytes via elevated ROS levels due to oxidative stress, impacting hearing loss. METHODS: We established a type II diabetes model in C57BL/6J mice and used auditory brainstem response (ABR), Evans blue staining, HE staining, immunohistochemistry, and immunofluorescence to observe changes in hearing, blood-labyrinth barrier (BLB) permeability, stria vascularis morphology, and apoptosis protein expression. Primary cultured stria vascularis pericytes were subjected to high glucose, and apoptosis levels were assessed using flow cytometry, Annexin V-FITC, Hoechst 33342 staining, Western blot, Mitosox, and JC-1 probes. RESULTS: Diabetic mice showed decreased hearing thresholds, reduced stria vascularis density, increased oxidative stress, cell apoptosis, and decreased antioxidant levels. High glucose exposure increased apoptosis and ROS content in pericytes, while mitochondrial membrane potential decreased, with AIF and cytochrome C (CytC) released from mitochondria to the cytoplasm. Adding oxidative scavengers reduced AIF and CytC release, decreasing pericyte apoptosis. DISCUSSION: Hyperglycemia may induce mitochondrial apoptosis of cochlear stria vascularis pericytes through oxidative stress.


Asunto(s)
Factor Inductor de la Apoptosis , Apoptosis , Citocromos c , Hiperglucemia , Ratones Endogámicos C57BL , Mitocondrias , Estrés Oxidativo , Pericitos , Proteínas Proto-Oncogénicas c-bcl-2 , Especies Reactivas de Oxígeno , Estría Vascular , Animales , Pericitos/metabolismo , Pericitos/efectos de los fármacos , Pericitos/patología , Estría Vascular/metabolismo , Estría Vascular/patología , Ratones , Especies Reactivas de Oxígeno/metabolismo , Mitocondrias/metabolismo , Citocromos c/metabolismo , Factor Inductor de la Apoptosis/metabolismo , Hiperglucemia/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Masculino , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Cóclea/metabolismo , Cóclea/patología
4.
J Transl Med ; 22(1): 712, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39085929

RESUMEN

BACKGROUND: Excessive pericyte coverage promotes tumor growth, and a downregulation may solve this dilemma. Due to the double-edged sword role of vascular pericytes in tumor microenvironment (TME), indiscriminately decreasing pericyte coverage by imatinib causes poor treatment outcomes. Here, we optimized the use of imatinib in a colorectal cancer (CRC) model in high pericyte-coverage status, and revealed the value of multiparametric magnetic resonance imaging (mpMRI) at 9.4T in monitoring treatment-related changes in pericyte coverage and the TME. METHODS: CRC xenograft models were evaluated by histological vascular characterizations and mpMRI. Mice with the highest pericyte coverage were treated with imatinib or saline; then, vascular characterizations, tumor apoptosis and HIF-1α level were analyzed histologically, and alterations in the expression of Bcl-2/bax pathway were assessed through qPCR. The effects of imatinib were monitored by dynamic contrast-enhanced (DCE)-, diffusion-weighted imaging (DWI)- and amide proton transfer chemical exchange saturation transfer (APT CEST)-MRI at 9.4T. RESULTS: The DCE- parameters provided a good histologic match the tumor vascular characterizations. In the high pericyte coverage status, imatinib exhibited significant tumor growth inhibition, necrosis increase and pericyte coverage downregulation, and these changes were accompanied by increased vessel permeability, decreased microvessel density (MVD), increased tumor apoptosis and altered gene expression of apoptosis-related Bcl-2/bax pathway. Strategically, a 4-day imatinib effectively decreased pericyte coverage and HIF-1α level, and continuous treatment led to a less marked decrease in pericyte coverage and re-elevated HIF-1α level. Correlation analysis confirmed the feasibility of using mpMRI parameters to monitor imatinib treatment, with DCE-derived Ve and Ktrans being most correlated with pericyte coverage, Ve with vessel permeability, AUC with microvessel density (MVD), DWI-derived ADC with tumor apoptosis, and APT CEST-derived MTRasym at 1 µT with HIF-1α. CONCLUSIONS: These results provided an optimized imatinib regimen to achieve decreasing pericyte coverage and HIF-1α level in the high pericyte-coverage CRC model, and offered an ultrahigh-field multiparametric MRI approach for monitoring pericyte coverage and dynamics response of the TME to treatment.


Asunto(s)
Apoptosis , Neoplasias Colorrectales , Subunidad alfa del Factor 1 Inducible por Hipoxia , Mesilato de Imatinib , Imágenes de Resonancia Magnética Multiparamétrica , Pericitos , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/uso terapéutico , Animales , Pericitos/metabolismo , Pericitos/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/diagnóstico por imagen , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Línea Celular Tumoral , Apoptosis/efectos de los fármacos , Humanos , Ratones Desnudos , Microambiente Tumoral/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Sex Med ; 21(9): 751-761, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39033084

RESUMEN

BACKGROUND: Heparin-binding epidermal growth factor-like growth factor (HB-EGF) serves as a pro-angiogenic factor; however, there is to our knowledge currently no reported research on the relationship between HB-EGF and diabetic erectile dysfunction (ED). AIM: In this study we aimed to determine whether HB-EGF can improve the erectile function of streptozotocin-induced diabetic mice and to explore the related mechanisms. METHODS: Eight-week-old male C57BL/6 mice were used for diabetes induction. Diabetes mellitus (DM) was induced by low-dose injections of streptozotocin (50 mg/kg) for 5 consecutive days. Eight weeks after streptozotocin injections, DM was determined by measuring blood glucose and body weight. Diabetic mice were treated with two intracavernous administrations of phosphate-buffered saline (20 µL) or various doses of HB-EGF (days -3 and 0; 1, 5, and 10 µg in 20 µL of phosphate-buffered saline). The angiogenesis effect of HB-EGF was confirmed by tube formation and migration assays in mouse cavernous endothelial cells and mouse cavernous pericytes under high-glucose conditions. Erectile function was measured by electrical stimulation of the cavernous nerve, as well as histological examination and Western blot analysis for mechanism assessment. OUTCOMES: In vitro angiogenesis, cell proliferation, in vivo intracavernous pressure, neurovascular regeneration, cavernous permeability, and survival signaling were the outcomes measured. RESULTS: Expression of HB-EGF was reduced under diabetic conditions. Exogenous HB-EGF induced angiogenesis in mouse cavernous endothelial cells and mouse cavernous pericytes under high-glucose conditions. Erectile function was decreased in the DM group, whereas administration of HB-EGF resulted in a significant improvement of erectile function (91% of the age-matched control group) in association with increased neurovascular content, including cavernous endothelial cells, pericytes, and neuronal cells. Histological and Western blot analyses revealed a significant increase in the permeability of the corpus cavernosum in DM mice, which was attenuated by HB-EGF treatment. The protein expression of phospho-Akt Ser473 and phosphorylated endothelial nitric oxide synthase Ser1177 increased after HB-EGF treatment. CLINICAL IMPLICATIONS: The use of HB-EGF may be an effective strategy to treat ED associated with DM or other neurovascular diseases. STRENGTHS AND LIMITATIONS: Similarly to other pro-angiogenic factors, HB-EGF has dual roles in vascular and neuronal development. Our study focused on broadly evaluating the role of HB-EGF in diabetic ED. In view of the properties of HB-EGF as an angiogenic factor, its dose concentration should be strictly controlled to avoid potential side effects. CONCLUSION: In the diabetic ED mouse model in this study erectile function was improved by HB-EGF, which may provide new treatment strategies for patients with ED who do not respond to phosphodiesterase 5 Inhibitors.


Asunto(s)
Diabetes Mellitus Experimental , Disfunción Eréctil , Factor de Crecimiento Similar a EGF de Unión a Heparina , Ratones Endogámicos C57BL , Erección Peniana , Animales , Masculino , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Ratones , Disfunción Eréctil/tratamiento farmacológico , Disfunción Eréctil/etiología , Erección Peniana/efectos de los fármacos , Pene/efectos de los fármacos , Pene/irrigación sanguínea , Pene/inervación , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Células Endoteliales/efectos de los fármacos
6.
J Cell Mol Med ; 28(13): e18505, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39001579

RESUMEN

Hypoxia-ischaemia (HI) can induce the death of cerebrovascular constituent cells through oxidative stress. Hydrogen is a powerful antioxidant which can activate the antioxidant system. A hypoxia-ischaemia brain damage (HIBD) model was established in 7-day-old SD rats. Rats were treated with different doses of hydrogen-rich water (HRW), and brain pericyte oxidative stress damage, cerebrovascular function and brain tissue damage were assessed. Meanwhile, in vitro-cultured pericytes were subjected to oxygen-glucose deprivation and treated with different concentrations of HRW. Oxidative injury was measured and the molecular mechanism of how HRW alleviated oxidative injury of pericytes was also examined. The results showed that HRW significantly attenuated HI-induced oxidative stress in the brain pericytes of neonatal rats, partly through the Nrf2-HO-1 pathway, further improving cerebrovascular function and reducing brain injury and dysfunction. Furthermore, HRW is superior to a single-cell death inhibitor for apoptosis, ferroptosis, parthanatos, necroptosis and autophagy and can better inhibit HI-induced pericyte death. The liver and kidney functions of rats were not affected by present used HRW dose. This study elucidates the role and mechanism of hydrogen in treating HIBD from the perspective of pericytes, providing new theoretical evidence and mechanistic references for the clinical application of hydrogen in neonatal HIE.


Asunto(s)
Animales Recién Nacidos , Encéfalo , Hidrógeno , Hipoxia-Isquemia Encefálica , Estrés Oxidativo , Pericitos , Ratas Sprague-Dawley , Animales , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Hidrógeno/farmacología , Hipoxia-Isquemia Encefálica/patología , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Ratas , Estrés Oxidativo/efectos de los fármacos , Encéfalo/patología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Antioxidantes/farmacología
7.
Exp Neurol ; 379: 114886, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38996862

RESUMEN

Spinal cord injury (SCI) induces the disruption of the blood-spinal cord barrier (BSCB) and the failure of axonal growth. SCI activates a complex series of responses, including cell apoptosis and endoplasmic reticulum (ER) stress. Pericytes play a critical role in maintaining BSCB integrity and facilitating tissue growth and repair. However, the roles of pericytes in SCI and the potential mechanisms underlying the improvements in functional recovery in SCI remain unclear. Recent evidence indicates that irisflorentin exerts neuroprotective effects against Parkinson's disease; however, whether it has potential protective roles in SCI or not is still unknown. In this study, we found that the administration of irisflorentin significantly inhibited pericyte apoptosis, protected BSCB integrity, promoted axonal growth, and ultimately improved locomotion recovery in a rat model of SCI. In vitro, we found that the positive effects of irisflorentin on axonal growth were likely to be mediated by regulating the crosstalk between pericytes and neurons. Furthermore, irisflorentin effectively ameliorated ER stress caused by incubation with thapsigargin (TG) in pericytes. Meanwhile, the protective effect of irisflorentin on BSCB disruption is strongly related to the reduction of pericyte apoptosis via inhibition of ER stress. Collectively, our findings demonstrate that irisflorentin is beneficial for functional recovery after SCI and that pericytes are a valid target of interest for future SCI therapies.


Asunto(s)
Fármacos Neuroprotectores , Ratas Sprague-Dawley , Recuperación de la Función , Traumatismos de la Médula Espinal , Animales , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/patología , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Ratas , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Axones/efectos de los fármacos , Pericitos/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Femenino , Médula Espinal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Células Cultivadas
8.
Phytother Res ; 38(8): 4036-4052, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38886264

RESUMEN

Atherosclerosis represents the major cause of mortality worldwide and triggers higher risk of acute cardiovascular events. Pericytes-endothelial cells (ECs) communication is orchestrated by ligand-receptor interaction generating a microenvironment which results in intraplaque neovascularization, that is closely associated with atherosclerotic plaque instability. Notoginsenoside R1 (R1) exhibits anti-atherosclerotic bioactivity, but its effect on angiogenesis in atherosclerotic plaque remains elusive. The aim of our study is to explore the therapeutic effect of R1 on vulnerable plaque and investigate its potential mechanism against intraplaque neovascularization. The impacts of R1 on plaque stability and intraplaque neovascularization were assessed in ApoE-/- mice induced by high-fat diet. Pericytes-ECs direct or non-direct contact co-cultured with VEGF-A stimulation were used as the in vitro angiogenesis models. Overexpressing Ang1 in pericytes was performed to investigate the underlying mechanism. In vivo experiments, R1 treatment reversed atherosclerotic plaque vulnerability and decreased the presence of neovessels in ApoE-/- mice. Additionally, R1 reduced the expression of Ang1 in pericytes. In vitro experiments demonstrated that R1 suppressed pro-angiogenic behavior of ECs induced by pericytes cultured with VEGF-A. Mechanistic studies revealed that the anti-angiogenic effect of R1 was dependent on the inhibition of Ang1 and Tie2 expression, as the effects were partially reversed after Ang1 overexpressing in pericytes. Our study demonstrated that R1 treatment inhibited intraplaque neovascularization by governing pericyte-EC association via suppressing Ang1-Tie2/PI3K-AKT paracrine signaling pathway. R1 represents a novel therapeutic strategy for atherosclerotic vulnerable plaques in clinical application.


Asunto(s)
Angiopoyetina 1 , Aterosclerosis , Células Endoteliales , Ginsenósidos , Neovascularización Patológica , Pericitos , Placa Aterosclerótica , Receptor TIE-2 , Animales , Ginsenósidos/farmacología , Ratones , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Aterosclerosis/tratamiento farmacológico , Placa Aterosclerótica/tratamiento farmacológico , Receptor TIE-2/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Angiopoyetina 1/metabolismo , Ratones Endogámicos C57BL , Masculino , Comunicación Celular/efectos de los fármacos , Humanos , Transducción de Señal/efectos de los fármacos , Apolipoproteínas E , Dieta Alta en Grasa , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Int J Mol Sci ; 25(12)2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38928202

RESUMEN

Blood-brain barrier (BBB) dysfunction is a key feature in neuroimmunological and neurodegenerative diseases. In this study, we developed a microfluidic human BBB-on-a-chip to model barrier dysfunction and immune cell migration using immortalized TY10 brain endothelial cells, pericytes, and astrocytes. It was found that immortalized TY10 brain endothelial cells developed a microvascular structure under flow. Pericytes were localized on the basal side surrounding the TY10 microvascular structure, showing an in vivo-like structure. Barrier integrity increased under co-culture with pericytes. In addition, both ethylenediaminetetraacetic acid (EDTA) and anti-Claudin-5 (CLDN5) neutralizing antibody caused a decrease in the transendothelial electrical resistance (TEER). EDTA caused the leakage of 20 kDa dextran, suggesting different effects on the BBB based on the mechanism of action, whereas anti-CLDN5 antibody did not cause leakage. In the tri-culture model, human T cells migrated through endothelial vessels towards basal C-X-C motif chemokine ligand 12 (CXCL12). The live-imaging analysis confirmed the extravasation of fluorescence-labelled T cells in a CXCL12-concentration- and time-dependent manner. Our BBB model had an in vivo-like structure and successfully represented barrier dysfunction and transendothelial T cell migration. In addition, our study suggests that the inhibition of CLDN5 attenuates the BBB in humans. This platform has various potential uses in relation to the BBB in both drug discovery research and in elucidating the mechanisms of central nervous system diseases.


Asunto(s)
Barrera Hematoencefálica , Movimiento Celular , Células Endoteliales , Dispositivos Laboratorio en un Chip , Humanos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/efectos de los fármacos , Descubrimiento de Drogas/métodos , Técnicas de Cocultivo , Pericitos/metabolismo , Pericitos/efectos de los fármacos , Claudina-5/metabolismo , Astrocitos/metabolismo , Astrocitos/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos
10.
ACS Nano ; 18(22): 14348-14366, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38768086

RESUMEN

Pericyte dysfunction severely undermines cerebrovascular integrity and exacerbates neurodegeneration in Alzheimer's disease (AD). However, pericyte-targeted therapy is a yet-untapped frontier for AD. Inspired by the elevation of vascular cell adhesion molecule-1 (VCAM-1) and reactive oxygen species (ROS) levels in pericyte lesions, we fabricated a multifunctional nanoprodrug by conjugating the hybrid peptide VLC, a fusion of the VCAM-1 high-affinity peptide VHS and the neuroprotective apolipoprotein mimetic peptide COG1410, to curcumin (Cur) through phenylboronic ester bond (VLC@Cur-NPs) to alleviate complex pericyte-related pathological changes. Importantly, VLC@Cur-NPs effectively homed to pericyte lesions via VLC and released their contents upon ROS stimulation to maximize their regulatory effects. Consequently, VLC@Cur-NPs markedly increased pericyte regeneration to form a positive feedback loop and thus improved neurovascular function and ultimately alleviated memory defects in APP/PS1 transgenic mice. We present a promising therapeutic strategy for AD that can precisely modulate pericytes and has the potential to treat other cerebrovascular diseases.


Asunto(s)
Enfermedad de Alzheimer , Ratones Transgénicos , Pericitos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Pericitos/patología , Ratones , Especies Reactivas de Oxígeno/metabolismo , Curcumina/farmacología , Curcumina/química , Profármacos/farmacología , Profármacos/química , Nanopartículas/química , Molécula 1 de Adhesión Celular Vascular/metabolismo , Humanos , Péptidos/química , Péptidos/farmacología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/química
11.
Phytomedicine ; 129: 155639, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38669966

RESUMEN

BACKGROUND: Cerebral microcirculation disturbance manifested by decrease of cerebral blood flow (CBF) is one of early features of Alzheimer's disease (AD). Shenqi Yizhi prescription (SQYZ) is widely used in the treatment of AD. However, the effect of SQYZ on the early feature of AD is not clarified. PURPOSE: To explore the effect and mechanism of SQYZ on AD-like behavior from the perspective of early pathological features of AD. METHODS: The fingerprint of SQYZ was established by ultra-high-performance liquid chromatograph. The improvement effect of SQYZ on Aß1-42 Oligomer (AßO)-induced AD-like behavior of mice was evaluated by behavioral test. The changes of CBF were detected by laser doppler meter and laser speckle imaging. The pathological changes of the hippocampus were observed by HE staining and transmission electron microscope. The expressions of intercellular communication molecules were detected by western blotting or immunofluorescence staining. The content of platelet-derived growth factor-BB (PDGF-BB) was detected by ELISA. Finally, the core components of SQYZ were docked with platelet-derived growth factor receptor beta (PDGFRß) using AutoDock Vina software. RESULTS: The similarity of the components in SQYZ extracted from different batches of medicinal materials was higher than 0.9. SQYZ administration could improve AßO-induced memory impairment and CBF reduction. Compared with the sham group, the number of neurons in the hippocampi of AßO group was significantly reduced, and the microvessels were shrunken and deformed. By contrary, SQYZ administration mitigated those pathological changes. Compared with the sham mice, the expressions of CD31, N-cadherin, PDGFRß, glial fibrillary acidic protein, phosphorylation of focal adhesion kinase, integrin ß1, and integrin α5 in the hippocampi of AßO mice were significantly increased. However, SQYZ administration significantly reduced AßO-induced expression of those proteins. Interestingly, the effect of PDGFRß inhibitor, sunitinib demonstrated a consistent modulating effect as SQYZ. Finally, the brain-entering components of SQYZ, including ginsenoside Rg5, coptisine, cryptotanshinone, dihydrotanshinone IIA, stigmasterol, and tanshinone IIA had high binding force with PDGFRß, implicating PDGFRß as a potential target for SQYZ. CONCLUSIONS: Our data indicate that SQYZ improves CBF in AßO-triggered AD-like mice through inhibiting brain pericyte contractility, indicating the treatment potential of SQYZ for AD at the early stage.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Medicamentos Herbarios Chinos , Hipocampo , Trastornos de la Memoria , Pericitos , Animales , Medicamentos Herbarios Chinos/farmacología , Péptidos beta-Amiloides/metabolismo , Masculino , Ratones , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/inducido químicamente , Enfermedad de Alzheimer/tratamiento farmacológico , Pericitos/efectos de los fármacos , Hipocampo/efectos de los fármacos , Fragmentos de Péptidos , Becaplermina/farmacología , Circulación Cerebrovascular/efectos de los fármacos , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Ginsenósidos/farmacología
12.
NanoImpact ; 34: 100508, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38663501

RESUMEN

The objective of this investigation was to evaluate the influence of micro- and nanoplastic particles composed of polyethylene terephthalate (PET), a significant contributor to plastic pollution, on human brain vascular pericytes. Specifically, we delved into their impact on mitochondrial functionality, oxidative stress, and the expression of genes associated with oxidative stress, ferroptosis and mitochondrial functions. Our findings demonstrate that the exposure of a monoculture of human brain vascular pericytes to PET particles in vitro at a concentration of 50 µg/ml for a duration of 3, 6 and 10 days did not elicit oxidative stress. Notably, we observed a reduction in various aspects of mitochondrial respiration, including maximal respiration, spare respiratory capacity, and ATP production in pericytes subjected to PET particles for 3 days, with a mitochondrial function recovery at 6 and 10 days. Furthermore, there were no statistically significant alterations in mitochondrial DNA copy number, or in the expression of genes linked to oxidative stress and ferroptosis, but an increase of the expression of the gene mitochondrial transcription factor A (TFAM) was noted at 3 days exposure. These outcomes suggest that, at a concentration of 50 µg/ml, PET particles do not induce oxidative stress in human brain vascular pericytes. Instead, at 3 days exposure, PET exposure impairs mitochondrial functions, but this is recovered at 6-day exposure. This seems to indicate a potential mitochondrial hormesis response (mitohormesis) is incited, involving the gene TFAM. Further investigations are warranted to explore the stages of mitohormesis and the potential consequences of plastics on the integrity of the blood-brain barrier and intercellular interactions. This research contributes to our comprehension of the potential repercussions of nanoplastic pollution on human health and underscores the imperative need for ongoing examinations into the exposure to plastic particles.


Asunto(s)
Encéfalo , Mitocondrias , Estrés Oxidativo , Pericitos , Tereftalatos Polietilenos , Humanos , Estrés Oxidativo/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Encéfalo/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Nanopartículas , Microplásticos/toxicidad , Células Cultivadas
13.
Neuron ; 112(13): 2177-2196.e6, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38653248

RESUMEN

White matter injury (WMI) causes oligodendrocyte precursor cell (OPC) differentiation arrest and functional deficits, with no effective therapies to date. Here, we report increased expression of growth hormone (GH) in the hypoxic neonatal mouse brain, a model of WMI. GH treatment during or post hypoxic exposure rescues hypoxia-induced hypomyelination and promotes functional recovery in adolescent mice. Single-cell sequencing reveals that Ghr mRNA expression is highly enriched in vascular cells. Cell-lineage labeling and tracing identify the GHR-expressing vascular cells as a subpopulation of pericytes. These cells display tip-cell-like morphology with kinetic polarized filopodia revealed by two-photon live imaging and seemingly direct blood vessel branching and bridging. Gain-of-function and loss-of-function experiments indicate that GHR signaling in pericytes is sufficient to modulate angiogenesis in neonatal brains, which enhances OPC differentiation and myelination indirectly. These findings demonstrate that targeting GHR and/or downstream effectors may represent a promising therapeutic strategy for WMI.


Asunto(s)
Vaina de Mielina , Neovascularización Fisiológica , Pericitos , Animales , Pericitos/metabolismo , Pericitos/efectos de los fármacos , Ratones , Vaina de Mielina/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Hormona del Crecimiento/metabolismo , Hormona del Crecimiento/farmacología , Animales Recién Nacidos , Hipoxia/metabolismo , Diferenciación Celular/efectos de los fármacos , Ratones Endogámicos C57BL , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Receptores de Somatotropina/metabolismo , Receptores de Somatotropina/genética , Angiogénesis
14.
J Control Release ; 369: 458-474, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38575077

RESUMEN

The blood-brain barrier (BBB)/blood-tumor barrier (BTB) impedes brain entry of most brain-targeted drugs, whether they are water-soluble or hydrophobic. Endothelial WNT signaling and neoplastic pericytes maintain BTB low permeability by regulating tight junctions. Here, we proposed nitazoxanide (NTZ) and ibrutinib (IBR) co-loaded ICAM-1-targeting nanoparticles (NI@I-NPs) to disrupt the BTB in a time-dependent, reversible, and size-selective manner by targeting specific ICAM-1, inactivating WNT signaling and depleting pericytes in tumor-associated blood vessels in breast cancer brain metastases. At the optimal NTZ/IBR mass ratio (1:2), BTB opening reached the optimum effect at 48-72 h without any sign of intracranial edema and cognitive impairment. The combination of NI@I-NPs and chemotherapeutic drugs (doxorubicin and etoposide) extended the median survival of mice with breast cancer brain metastases. Targeting BTB endothelial WNT signaling and tumor pericytes via NI@I-NPs could open the BTB to improve chemotherapeutic efficiency against brain metastases.


Asunto(s)
Barrera Hematoencefálica , Neoplasias Encefálicas , Nanopartículas , Pericitos , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/metabolismo , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Pericitos/metabolismo , Pericitos/efectos de los fármacos , Femenino , Humanos , Nanopartículas/administración & dosificación , Piperidinas/administración & dosificación , Piperidinas/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Tiazoles/administración & dosificación , Tiazoles/farmacología , Línea Celular Tumoral , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Pirazoles/administración & dosificación , Pirazoles/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Doxorrubicina/administración & dosificación , Doxorrubicina/uso terapéutico , Ratones Endogámicos BALB C , Vía de Señalización Wnt/efectos de los fármacos , Ratones , Sistemas de Liberación de Medicamentos , Adenina/análogos & derivados
15.
Drug Deliv Transl Res ; 14(10): 2629-2641, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38498080

RESUMEN

Drug delivery to central nervous pathologies is compromised by the blood-brain barrier (BBB). A clinically explored strategy to promote drug delivery across the BBB is sonopermeation, which relies on the combined use of ultrasound (US) and microbubbles (MB) to induce temporally and spatially controlled opening of the BBB. We developed an advanced in vitro BBB model to study the impact of sonopermeation on the delivery of the prototypic polymeric drug carrier pHPMA as a larger molecule and the small molecule antiviral drug ribavirin. This was done under standard and under inflammatory conditions, employing both untargeted and RGD peptide-coated MB. The BBB model is based on human cerebral capillary endothelial cells and human placental pericytes, which are co-cultivated in transwell inserts and which present with proper transendothelial electrical resistance (TEER). Sonopermeation induced a significant decrease in TEER values and facilitated the trans-BBB delivery of fluorescently labeled pHPMA (Atto488-pHPMA). To study drug delivery under inflamed endothelial conditions, which are typical for e.g. tumors, neurodegenerative diseases and CNS infections, tumor necrosis factor (TNF) was employed to induce inflammation in the BBB model. RGD-coated MB bound to and permeabilized the inflamed endothelium-pericyte co-culture model, and potently improved Atto488-pHPMA and ribavirin delivery. Taken together, our work combines in vitro BBB bioengineering with MB-mediated drug delivery enhancement, thereby providing a framework for future studies on optimization of US-mediated drug delivery to the brain.


Asunto(s)
Barrera Hematoencefálica , Técnicas de Cocultivo , Células Endoteliales , Microburbujas , Oligopéptidos , Pericitos , Barrera Hematoencefálica/metabolismo , Humanos , Oligopéptidos/química , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacocinética , Pericitos/metabolismo , Pericitos/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Ribavirina/administración & dosificación , Ribavirina/química , Ribavirina/farmacocinética , Ondas Ultrasónicas , Polímeros/química , Polímeros/administración & dosificación , Antivirales/administración & dosificación , Antivirales/química , Antivirales/farmacología , Antivirales/farmacocinética , Inflamación/tratamiento farmacológico
16.
Adv Healthc Mater ; 13(18): e2400388, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38465502

RESUMEN

Hydrogel-based 3D cell cultures can recapitulate (patho)physiological phenomena ex vivo. However, due to their complex multifactorial regulation, adapting these tissue and disease models for high-throughput screening workflows remains challenging. In this study, a new precision culture scaling (PCS-X) methodology combines statistical techniques (design of experiment and multiple linear regression) with automated, parallelized experiments and analyses to customize hydrogel-based vasculogenesis cultures using human umbilical vein endothelial cells and retinal microvascular endothelial cells. Variations of cell density, growth factor supplementation, and media composition are systematically explored to induce vasculogenesis in endothelial mono- and cocultures with mesenchymal stromal cells or retinal microvascular pericytes in 384-well plate formats. The developed cultures are shown to respond to vasculogenesis inhibitors in a compound- and dose-dependent manner, demonstrating the scope and power of PCS-X in creating parallelized tissue and disease models for drug discovery and individualized therapies.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana , Neovascularización Fisiológica , Humanos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Hidrogeles/química , Técnicas de Cocultivo/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Pericitos/citología , Pericitos/metabolismo , Pericitos/efectos de los fármacos , Técnicas de Cultivo de Célula/métodos , Técnicas de Cultivo de Célula/instrumentación , Células Endoteliales/citología , Células Endoteliales/metabolismo
17.
Nature ; 622(7983): 611-618, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37699522

RESUMEN

Clostridioides difficile infection (CDI) is a major cause of healthcare-associated gastrointestinal infections1,2. The exaggerated colonic inflammation caused by C. difficile toxins such as toxin B (TcdB) damages tissues and promotes C. difficile colonization3-6, but how TcdB causes inflammation is unclear. Here we report that TcdB induces neurogenic inflammation by targeting gut-innervating afferent neurons and pericytes through receptors, including the Frizzled receptors (FZD1, FZD2 and FZD7) in neurons and chondroitin sulfate proteoglycan 4 (CSPG4) in pericytes. TcdB stimulates the secretion of the neuropeptides substance P (SP) and calcitonin gene-related peptide (CGRP) from neurons and pro-inflammatory cytokines from pericytes. Targeted delivery of the TcdB enzymatic domain, through fusion with a detoxified diphtheria toxin, into peptidergic sensory neurons that express exogeneous diphtheria toxin receptor (an approach we term toxogenetics) is sufficient to induce neurogenic inflammation and recapitulates major colonic histopathology associated with CDI. Conversely, mice lacking SP, CGRP or the SP receptor (neurokinin 1 receptor) show reduced pathology in both models of caecal TcdB injection and CDI. Blocking SP or CGRP signalling reduces tissue damage and C. difficile burden in mice infected with a standard C. difficile strain or with hypervirulent strains expressing the TcdB2 variant. Thus, targeting neurogenic inflammation provides a host-oriented therapeutic approach for treating CDI.


Asunto(s)
Toxinas Bacterianas , Clostridioides difficile , Inflamación Neurogénica , Neuronas Aferentes , Pericitos , Animales , Ratones , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/farmacología , Péptido Relacionado con Gen de Calcitonina/antagonistas & inhibidores , Péptido Relacionado con Gen de Calcitonina/metabolismo , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/microbiología , Inflamación Neurogénica/inducido químicamente , Inflamación Neurogénica/microbiología , Inflamación Neurogénica/patología , Pericitos/efectos de los fármacos , Pericitos/microbiología , Pericitos/patología , Receptores de Neuroquinina-1/metabolismo , Sustancia P/antagonistas & inhibidores , Sustancia P/metabolismo , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/microbiología , Neuronas Aferentes/patología , Mediadores de Inflamación/metabolismo , Ciego/efectos de los fármacos , Ciego/metabolismo , Transducción de Señal/efectos de los fármacos
18.
Folia Morphol (Warsz) ; 82(3): 533-542, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35818807

RESUMEN

BACKGROUND: Cisplatin is a platinum-based antineoplastic agent used to treat cancers of solid organs. Neuropathy is one of its major side effects, necessitating dose reduction or cessation. Previous studies suggested that cisplatin causes microvascular toxicity, including pericyte detachment. This study aimed to clarify whether these alterations occurred in the blood-nerve barrier (BNB) of capillaries after cisplatin treatment. MATERIALS AND METHODS AND RESULTS: Electron microscopic analysis of rat sciatic nerves with cisplatin neuropathy showed increased frequency and severity of pericyte detachment. Moreover, the vascular basement membrane did not tightly encircle around the endothelial cells and pericytes. Cultured human umbilical vein endothelial cells and human brain vascular pericytes showed reduced viability, increased caspase-3 activity and enhanced oxidative stress following cisplatin treatment. In addition, cisplatin decreased transendothelial electrical resistance (TEER) and the expression of the tight junction proteins occludin and zonula occludens-1. Curcumin, a polyphenol found in the root of Curcuma longa, had favourable effects on cisplatin neuropathy in previous work. Therefore, curcumin was tested to determine whether it had any effect on these abnormalities. Curcumin alleviated pericyte detachment, cytotoxicity, oxidative stress, TEER reduction and tight junction protein expression. CONCLUSIONS: These data indicate that cisplatin causes BNB disruption in the nerves and might result in neuropathy. Curcumin might improve neuropathy via the restoration of BNB. Whether alterations in the BNB occur and curcumin is effective in patients with cisplatin neuropathy remain to be investigated.


Asunto(s)
Antineoplásicos , Barrera Hematonerviosa , Cisplatino , Curcumina , Células Endoteliales de la Vena Umbilical Humana , Pericitos , Neuropatía Ciática , Curcumina/farmacología , Barrera Hematonerviosa/efectos de los fármacos , Barrera Hematonerviosa/patología , Pericitos/efectos de los fármacos , Pericitos/patología , Cisplatino/toxicidad , Antineoplásicos/toxicidad , Animales , Ratas , Humanos , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/patología , Neuropatía Ciática/inducido químicamente , Neuropatía Ciática/prevención & control , Nervio Ciático/irrigación sanguínea , Nervio Ciático/efectos de los fármacos , Nervio Ciático/patología , Femenino , Ratas Wistar
19.
Biochem Biophys Res Commun ; 618: 61-66, 2022 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-35716596

RESUMEN

Lysophosphatidic acid (LPA) is a bioactive compound known to regulate various vascular functions. However, despite the fact that many vascular functions are regulated by peri-vascular cells such as pericytes, the effect of LPA on brain pericytes has not been fully evaluated. Thus, we designed this study to evaluate the effects of LPA on brain pericytes. These experiments revealed that while LPA receptors (LPARs) are expressed in cultured pericytes from mouse brains, LPA treatment does not influence the proliferation of these cells but does have a profound impact on their migration, which is regulated via the expression of LPAR1. LPAR1 expression was also detected in human pericyte culture and LPA treatment of these cells also induced migration. Taken together these findings imply that LPA-LPAR1 signaling is one of the key mechanisms modulating pericyte migration, which may help to control vascular function during development and repair processes.


Asunto(s)
Lisofosfolípidos , Pericitos , Receptores del Ácido Lisofosfatídico , Animales , Movimiento Celular , Lisofosfolípidos/farmacología , Ratones , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA