Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Am J Pathol ; 194(7): 1171-1184, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38548268

RESUMEN

Interactions between endothelial cells (ECs) and mural pericytes (PCs) are critical in maintaining the stability and function of the microvascular wall. Abnormal interactions between these two cell types are a hallmark of progressive fibrotic diseases such as systemic sclerosis (also known as scleroderma). However, the role of PCs in signaling microvascular dysfunction remains underexplored. We hypothesized that integrin-matrix interactions contribute to PC migration from the vascular wall and conversion into interstitial myofibroblasts. Herein, pro-inflammatory tumor necrosis factor α (TNFα) or a fibrotic growth factor [transforming growth factor ß1 (TGF-ß1)] were used to evaluate human PC inflammatory and fibrotic phenotypes by assessing their migration, matrix deposition, integrin expression, and subsequent effects on endothelial dysfunction. Both TNFα and TGF-ß1 treatment altered integrin expression and matrix protein deposition, but only fibrotic TGF-ß1 drove PC migration in an integrin-dependent manner. In addition, integrin-dependent PC migration was correlated to changes in EC angiopoietin-2 levels, a marker of vascular instability. Finally, there was evidence of changes in vascular stability corresponding to disease state in human systemic sclerosis skin. This work shows that TNFα and TGF-ß1 induce changes in PC integrin expression and matrix deposition that facilitate migration and reduce vascular stability, providing evidence that microvascular destabilization can be an early indicator of tissue fibrosis.


Asunto(s)
Movimiento Celular , Fibrosis , Integrinas , Pericitos , Esclerodermia Sistémica , Factor de Crecimiento Transformador beta1 , Pericitos/metabolismo , Pericitos/patología , Humanos , Factor de Crecimiento Transformador beta1/metabolismo , Esclerodermia Sistémica/patología , Esclerodermia Sistémica/metabolismo , Integrinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Microvasos/patología , Microvasos/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Piel/patología , Piel/metabolismo , Piel/irrigación sanguínea
2.
FASEB J ; 35(2): e21311, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33417253

RESUMEN

Neuroblasts have a clustered phenotype critical for their unidirectional migration, which in part is dependent on signaling from microvascular endothelial cells (EC) and pericytes (PC). Diffusible signals secreted by vascular cells have been demonstrated to increase survival, proliferation, and differentiation of subventricular zone resident neural stem cells (NSC); however, the signals that promote the necessary initiating step of NSC clustering are undefined. To investigate the role of vascular cells in promoting NSC clustering and directing migration, we created a 3-D hydrogel that mimics the biomechanics, biochemistry, and architectural complexity of brain tissue. We demonstrate that EC, and not PC, have a crucial role in NSC clustering and migration, further verified through microfluidic chamber systems and traction force microscopy. Ablation of the extended NSC aggregate arm halts aggregate movement, suggesting that clustering is a prerequisite for migration. When cultured with EC, NSC clustering occurs and NSC coincidentally increase their expression of N-cadherin, as compared to NSC cultured alone. NSC-presented N-cadherin expression was increased following exposure to EC secreted metalloproteinase-2 (MMP2). We demonstrate that inhibition of MMP2 prevented NSC N-cadherin surface expression and subsequent NSC clustering, even when NSC were in direct contact with EC. Furthermore, with exogenous activation of EGFR, which serves as a downstream activator of N-cadherin cleavage, NSC form clusters. Our results suggest that EC secretion of MMP2 promotes NSC clustering through N-cadherin expression. The insight gained about the mechanisms by which EC promote NSC migration may enhance NSC therapeutic response to sites of injury.


Asunto(s)
Cadherinas/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Células-Madre Neurales/metabolismo , Animales , Cadherinas/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Movimiento Celular/genética , Movimiento Celular/fisiología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Hidrogeles/química , Metaloproteinasa 2 de la Matriz/genética , Ratones
3.
FASEB J ; 33(2): 2171-2186, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30252532

RESUMEN

Dysregulated neutrophil extravasation contributes to the pathogenesis of many inflammatory disorders. Pericytes (PCs) have been implicated in the regulation of neutrophil transmigration, and previous work demonstrates that endothelial cell (EC)-derived signals reduce PC barrier function; however, the signaling mechanisms are unknown. Here, we demonstrate a novel role for EC-derived macrophage migration inhibitory factor (MIF) in inhibiting PC contractility and facilitating neutrophil transmigration. With the use of micro-ELISAs, RNA sequencing, quantitative PCR, and flow cytometry, we found that ECs secrete MIF, and PCs upregulate CD74 in response to TNF-α. We demonstrate that EC-derived MIF decreases PC contractility on 2-dimensional silicone substrates via reduction of phosphorylated myosin light chain. With the use of an in vitro microvascular model of the human EC-PC barrier, we demonstrate that MIF decreases the PC barrier to human neutrophil transmigration by increasing intercellular PC gap formation. For the first time, an EC-specific MIF knockout mouse was used to investigate the effects of selective deletion of EC MIF. In a model of acute lung injury, selective deletion of EC MIF decreases neutrophil infiltration to the bronchoalveolar lavage and tissue and simultaneously decreases PC relaxation by increasing myosin light-chain phosphorylation. We conclude that paracrine signals from EC via MIF decrease PC contraction and enhance PC-regulated neutrophil transmigration.-Pellowe, A. S., Sauler, M., Hou, Y., Merola, J., Liu, R., Calderon, B., Lauridsen, H. M., Harris, M. R., Leng, L., Zhang, Y., Tilstam, P. V., Pober, J. S., Bucala, R., Lee, P. J., Gonzalez, A. L. Endothelial cell-secreted MIF reduces pericyte contractility and enhances neutrophil extravasation.


Asunto(s)
Endotelio Vascular/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Neutrófilos/citología , Pericitos/citología , Animales , Líquido del Lavado Bronquioalveolar , Células Cultivadas , Endotelio Vascular/citología , Ensayo de Inmunoadsorción Enzimática , Humanos , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , Ratones Noqueados
4.
Am J Pathol ; 187(8): 1893-1906, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28609645

RESUMEN

Sweet syndrome (SS) is a prototypical neutrophilic dermatosis, a class of inflammatory diseases marked by elevated levels of tumor necrosis factor (TNF)-α and IL-17A, pathologic neutrophil recruitment, and microvascular remodeling. Histologic analyses of four matrix proteins-collagen I and IV, laminin, and fibronectin-in skin biopsies of patients with SS reveal that the basement membrane of dermal postcapillary venules undergoes changes in structure and composition. Increased neutrophil recruitment in vivo was associated with increases in collagen IV, decreases in laminin, and varied changes in fibronectin. In vitro studies using TNF-α and IL-17A were conducted to dissect basement membrane remodeling. Prolonged dual activation of cultured human pericytes with TNF-α and IL-17A augmented collagen IV production, similar to in vivo remodeling. Co-activation of pericytes with TNF-α and IL-17A also elevated fibronectin levels with little direct effect on laminin. However, the expression of fibronectin- and laminin-specific matrix metalloproteinases (MMPs), particularly MMP-3, was significantly up-regulated. Interactions between pericytes and neutrophils in culture yielded even higher levels of active MMPs, facilitating fibronectin and laminin degradation, and likely contributing to the varied levels of detectable fibronectin and the decreases in laminin observed in vivo. These data indicate that pericyte-neutrophil interactions play a role in mediating microvascular changes in SS and suggest that targeting MMP-3 may be effective in protecting vascular wall integrity.


Asunto(s)
Membrana Basal/efectos de los fármacos , Interleucina-17/farmacología , Neutrófilos/metabolismo , Pericitos/efectos de los fármacos , Síndrome de Sweet/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Anciano , Membrana Basal/metabolismo , Membrana Basal/patología , Células Cultivadas , Colágeno Tipo IV/metabolismo , Femenino , Fibronectinas/metabolismo , Humanos , Laminina/metabolismo , Masculino , Metaloproteinasa 3 de la Matriz/metabolismo , Persona de Mediana Edad , Neutrófilos/patología , Pericitos/metabolismo , Pericitos/patología , Síndrome de Sweet/patología
5.
J Immunol ; 197(6): 2400-8, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27534549

RESUMEN

A classical hallmark of acute inflammation is neutrophil infiltration of tissues, a multistep process that involves sequential cell-cell interactions of circulating leukocytes with IL-1- or TNF-activated microvascular endothelial cells (ECs) and pericytes (PCs) that form the wall of the postcapillary venules. The initial infiltrating cells accumulate perivascularly in close proximity to PCs. IL-17, a proinflammatory cytokine that acts on target cells via a heterodimeric receptor formed by IL-17RA and IL-17RC subunits, also promotes neutrophilic inflammation but its effects on vascular cells are less clear. We report that both cultured human ECs and PCs strongly express IL-17RC and, although neither cell type expresses much IL-17RA, PCs express significantly more than ECs. IL-17, alone or synergistically with TNF, significantly alters inflammatory gene expression in cultured human PCs but not ECs. RNA sequencing analysis identifies many IL-17-induced transcripts in PCs encoding proteins known to stimulate neutrophil-mediated immunity. Conditioned media from IL-17-activated PCs, but not ECs, induce pertussis toxin-sensitive neutrophil polarization, likely mediated by PC-secreted chemokines, and they also stimulate neutrophil production of proinflammatory molecules, including TNF, IL-1α, IL-1ß, and IL-8. Furthermore, IL-17-activated PCs, but not ECs, can prolong neutrophil survival by producing G-CSF and GM-CSF, delaying the mitochondrial outer membrane permeabilization and caspase-9 activation. Importantly, neutrophils exhibit enhanced phagocytic capacity after activation by conditioned media from IL-17-treated PCs. We conclude that PCs, not ECs, are the major target of IL-17 within the microvessel wall and that IL-17-activated PCs can modulate neutrophil functions within the perivascular tissue space.


Asunto(s)
Endotelio Vascular/fisiología , Interleucina-17/inmunología , Neutrófilos/inmunología , Pericitos/fisiología , Receptores de Interleucina-17/inmunología , Caspasa 9/metabolismo , Células Cultivadas , Medios de Cultivo , Citocinas/biosíntesis , Citocinas/inmunología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Factor Estimulante de Colonias de Granulocitos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Interleucina-17/genética , Interleucina-17/farmacología , Infiltración Neutrófila , Neutrófilos/fisiología , Pericitos/efectos de los fármacos , Pericitos/inmunología , Receptores de Interleucina-17/fisiología , Análisis de Secuencia de ARN , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/farmacología , Vénulas/citología , Vénulas/inmunología
6.
Am J Respir Crit Care Med ; 196(12): 1571-1581, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-28783377

RESUMEN

RATIONALE: Idiopathic pulmonary fibrosis (IPF) involves the accumulation of α-smooth muscle actin-expressing myofibroblasts arising from interactions with soluble mediators such as transforming growth factor-ß1 (TGF-ß1) and mechanical influences such as local tissue stiffness. Whereas IPF fibroblasts are enriched for aerobic glycolysis and innate immune receptor activation, innate immune ligands related to mitochondrial injury, such as extracellular mitochondrial DNA (mtDNA), have not been identified in IPF. OBJECTIVES: We aimed to define an association between mtDNA and fibroblast responses in IPF. METHODS: We evaluated the response of normal human lung fibroblasts (NHLFs) to stimulation with mtDNA and determined whether the glycolytic reprogramming that occurs in response to TGF-ß1 stimulation and direct contact with stiff substrates, and spontaneously in IPF fibroblasts, is associated with excessive levels of mtDNA. We measured mtDNA concentrations in bronchoalveolar lavage (BAL) from subjects with and without IPF, as well as in plasma samples from two longitudinal IPF cohorts and demographically matched control subjects. MEASUREMENTS AND MAIN RESULTS: Exposure to mtDNA augments α-smooth muscle actin expression in NHLFs. The metabolic changes in NHLFs that are induced by interactions with TGF-ß1 or stiff hydrogels are accompanied by the accumulation of extracellular mtDNA. These findings replicate the spontaneous phenotype of IPF fibroblasts. mtDNA concentrations are increased in IPF BAL and plasma, and in the latter compartment, they display robust associations with disease progression and reduced event-free survival. CONCLUSIONS: These findings demonstrate a previously unrecognized and highly novel connection between metabolic reprogramming, mtDNA, fibroblast activation, and clinical outcomes that provides new insight into IPF.


Asunto(s)
ADN Mitocondrial/metabolismo , Fibroblastos/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/mortalidad , Anciano , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino
7.
Microcirculation ; 22(1): 54-67, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25214363

RESUMEN

OBJECTIVE: Neutrophil extravasation at post-capillary venules, consisting of EC, PC, and the shared ECM, increases following fibrotic remodeling in the lung, liver, and skin. The role of fibrotic pericyte-derived ECM in regulating EC activation and neutrophil recruitment remains unexplored. METHODS: To elucidate the role of human pericyte-derived ECM in EC activation, we characterized PC-derived ECM following transforming growth factor-ß1, IL-1ß, CCL2, or bleomycin activation, and examined surface adhesion molecule expression and neutrophil recruitment by EC cultured on PC-ECM. RESULTS: Pro-inflammatory activation of PC-induced deposition of compositionally distinct ECM compared with non-activated control. Bleomycin activation induced fibronectin-rich and collagen-poor ECM remodeling by PC, facilitating increased neutrophil transendothelial migration when compared with non-activated pericyte ECM (49.9 ± 3.4% versus 29.7 ± 1.4%). Increases in fibronectin compared to collagen I, are largely responsible for ECM-regulated neutrophil recruitment, as EC cultured on fibronectin supported increased neutrophil transmigration compared to collagen I (51.6 ± 6.2% versus 28.0 ± 4.8%). We attribute this difference to increased expression of ICAM-1 and its redistribution to EC borders. CONCLUSIONS: This is the first demonstration of human pericyte sensitivity to inflammatory stimuli, inducing fibrotic matrix deposition that regulates EC adhesion molecule expression and neutrophil recruitment.


Asunto(s)
Membrana Basal/metabolismo , Matriz Extracelular/metabolismo , Molécula 1 de Adhesión Intercelular/biosíntesis , Neutrófilos/metabolismo , Pericitos/metabolismo , Migración Transendotelial y Transepitelial/fisiología , Antibióticos Antineoplásicos/farmacología , Bleomicina/farmacología , Quimiocina CCL2/metabolismo , Humanos , Interleucina-1beta/metabolismo , Neutrófilos/citología , Pericitos/citología , Migración Transendotelial y Transepitelial/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo
8.
FASEB J ; 28(3): 1166-80, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24297702

RESUMEN

Neutrophil extravasation occurs across postcapillary venules, structures composed of endothelial cells (ECs), pericytes (PCs), and basement membrane (BM). We constructed composite models of the human postcapillary venule, combining ECs with PCs or PC-deposited BM, to better study this process. Quiescent and tumor necrosis factor α (TNF-α)-activated composites demonstrated in situ-like expression of cadherins, E-selectin, intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), platelet-endothelial cell adhesion molecule 1 (PECAM-1), CD99, and interleukin 8 (IL-8). After TNF-α activation, the ECs supported greater neutrophil adhesion (66.1 vs. 23.7% of input cells) and transmigration (35.1 vs. 7.20% of input cells) than did the PCs, but the composites behaved comparably (no significant difference) to ECs in both assays. TNF-α-activated EC-conditioned medium (CM) increased transmigration across the PCs, whereas TNF-α-activated PC-CM decreased transmigration across the ECs, and culturing on PC-derived BM decreased both adhesion to and transmigration across the ECs. Anti-very late antigen 4 (VLA-4; on neutrophils) inhibited adhesion to TNF-α-activated composites, but not to ECs alone. Anti-CD99 (expressed on all 3 cell types) inhibited transmigration across the composites (14.5% of control) more than across the ECs (39.0% of control), and venular shear stress reduced transmigration across the ECs (17.3% of static) more than across the composites (36.7% of static). These results provide proof of concept that our composite human EC/PC/BM venular construct can reveal new interactions in the inflammatory cascade.


Asunto(s)
Leucocitos/citología , Modelos Biológicos , Vénulas/anatomía & histología , Adhesión Celular , Movimiento Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Humanos , Microscopía Electrónica de Rastreo , Vénulas/citología
9.
Biotechnol Bioeng ; 110(3): 999-1003, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23055258

RESUMEN

The ability to discriminate cell adhesion molecule expression between healthy and inflamed endothelium is critical for therapeutic intervention in many diseases. This study explores the effect of laminar flow on TNFα-induced E-selectin surface expression levels in human umbilical vein endothelial cells (HUVECs) relative to IL-1ß-induced expression via flow chamber assays. HUVECs grown in static culture were either directly (naïve) activated with cytokine in the presence of laminar shear or pre-exposed to 12 h of laminar shear (shear-conditioned) prior to simultaneous shear and cytokine activation. Naïve cells activated with cytokine in static served as control. Depending on the cell shear history, fluid shear is found to differently affect TNFα-induced relative to IL-1ß-induced HUVEC expression of E-selectin. Specifically, E-selectin surface expression by naïve HUVECs is enhanced in the 8-12 h activation time range with simultaneous exposure to shear and TNFα (shear-TNFα) relative to TNFα static control whereas enhanced E-selectin expression is observed in the 4-24 h range for shear-IL-1ß treatment relative to IL-1ß static control. While exposure of HUVECs to shear preconditioning mutes shear-TNFα-induced E-selectin expression, it enhances or down-regulates shear-IL-1ß-induced expression dependent on the activation period. Under dual-cytokine-shear conditions, IL-1ß signaling dominates. Overall, a better understanding of E-selectin expression pattern by human ECs relative to the combined interaction of cytokines, shear profile and history can help elucidate many disease pathologies.


Asunto(s)
Selectina E/biosíntesis , Células Endoteliales/fisiología , Interleucina-1beta/metabolismo , Fenómenos Mecánicos , Estrés Fisiológico , Factor de Necrosis Tumoral alfa/metabolismo , Humanos
10.
Yale J Biol Med ; 86(4): 537-54, 2013 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-24348218

RESUMEN

Fibrosis is characterized by excessive extracellular matrix deposition and is the pathological outcome of repetitive tissue injury in many disorders. The accumulation of matrix disrupts the structure and function of the native tissue and can affect multiple organs including the lungs, heart, liver, and skin. Unfortunately, current therapies against the deadliest and most common fibrosis are ineffective. The pathogenesis of fibrosis is the result of aberrant wound healing, therefore, the microvasculature plays an important role, contributing through regulation of leukocyte recruitment, inflammation, and angiogenesis. Further exacerbating the condition, microvascular endothelial cells and pericytes can transdifferentiate into matrix depositing myofibroblasts. The contribution of the microvasculature to fibrotic progression makes its cellular components and acellular products attractive therapeutic targets. In this review, we examine many of the cytokine, matrix, and cellular microvascular components involved in fibrosis and discuss their potential as targets for fibrotic therapies with a particular focus on developing nanotechnologies.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Microvasos/efectos de los fármacos , Nanopartículas/química , Nanotecnología/métodos , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibrosis , Humanos , Microvasos/metabolismo , Microvasos/patología , Modelos Biológicos , Terapia Molecular Dirigida/métodos , Nanopartículas/administración & dosificación
11.
Yale J Biol Med ; 85(2): 167-85, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22737047

RESUMEN

Polymorphoneuclear leukocytes or neutrophils, a major component of white blood cells, contribute to the innate immune response in humans. Upon sensing changes in the microenvironment, neutrophils adhere to the vascular wall, migrate through the endothelial cell (EC)-pericyte bilayer, and subsequently through the extracellular matrix to reach the site of inflammation. These cells are capable of destroying microbes, cell debris, and foreign proteins by oxidative and non-oxidative processes. While primarily mediators of tissue homeostasis, there are an increasing number of studies indicating that neutrophil recruitment and transmigration can also lead to host-tissue injury and subsequently inflammation-related diseases. Neutrophil-induced tissue injury is highly regulated by the microenvironment of the infiltrated tissue, which includes cytokines, chemokines, and the provisional extracellular matrix, remodeled through increased vascular permeability and other cellular infiltrates. Thus, investigation of the effects of matrix proteins on neutrophil-EC interaction and neutrophil transmigration may help identify the proteins that induce pro- or anti-inflammatory responses. This area of research presents an opportunity to identify therapeutic targets in inflammation-related diseases. This review will summarize recent literature on the role of neutrophils and the effects of matrix proteins on neutrophil-EC interactions, with focus on three different disease models: 1) atherosclerosis, 2) COPD, and 3) tumor growth and progression. For each disease model, inflammatory molecules released by neutrophils, important regulatory matrix proteins, current anti-inflammatory treatments, and the scope for further research will be summarized.


Asunto(s)
Comunicación Celular/inmunología , Células Endoteliales/inmunología , Proteínas de la Matriz Extracelular/inmunología , Neutrófilos/inmunología , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Células Endoteliales/patología , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Modelos Inmunológicos , Neoplasias/inmunología , Neoplasias/metabolismo , Neutrófilos/patología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo
12.
Ann N Y Acad Sci ; 1518(1): 183-195, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36177947

RESUMEN

The ability to engineer complex multicellular systems has enormous potential to inform our understanding of biological processes and disease and alter the drug development process. Engineering living systems to emulate natural processes or to incorporate new functions relies on a detailed understanding of the biochemical, mechanical, and other cues between cells and between cells and their environment that result in the coordinated action of multicellular systems. On April 3-6, 2022, experts in the field met at the Keystone symposium "Engineering Multicellular Living Systems" to discuss recent advances in understanding how cells cooperate within a multicellular system, as well as recent efforts to engineer systems like organ-on-a-chip models, biological robots, and organoids. Given the similarities and common themes, this meeting was held in conjunction with the symposium "Organoids as Tools for Fundamental Discovery and Translation".


Asunto(s)
Ingeniería , Organoides , Humanos , Ingeniería de Tejidos
13.
Stem Cell Res ; 53: 102318, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33836422

RESUMEN

Although delivery of neural stem cell (NSC) as a therapeutic treatment for intracerebral hemorrhage (ICH) provides promise, NSC delivery typically has extremely low survival rates. Here, we investigate endothelial cell (EC) and pericyte (PC) interactions with NSC, where our results demonstrate that EC, and not PC, promote NSC cell proliferation and reduce cytotoxicity under glucose deprivation (GD). Additionally, NSC proliferation was increased upon treatment with EC conditioned media, inhibited with antagonism of VEGFR3. In an NSC + EC co-culture we detected elevated levels of VEGF-C, not seen for NSC cultured alone. Exogenous VEGF-C induced NSC upregulation of VEGFR3, promoted proliferation, and reduced cytotoxicity. Finally, we delivered microbeads containing NSC + EC into a murine ICH cavity, where VEGF-C was increasingly present in the injury site, not seen upon delivery NSC encapsulated alone. These studies demonstrate that EC-secreted VEGF-C may promote NSC survival during injury, enhancing the potential for cell delivery therapies for stroke.


Asunto(s)
Células-Madre Neurales , Factor C de Crecimiento Endotelial Vascular , Animales , Diferenciación Celular , Medios de Cultivo Condicionados , Células Endoteliales , Ratones
14.
Brain Res Bull ; 168: 120-137, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33373665

RESUMEN

Intracerebral implantation of neural stem cells (NSCs) to treat stroke remains an inefficient process with <5% of injected cells being retained. To improve the retention and distribution of NSCs after a stroke, we investigated the utility of NSCs' encapsulation in polyethylene glycol (PEG) microspheres. We first characterized the impact of the physical properties of different syringes and needles, as well as ejection speed, upon delivery of microspheres to the stroke injured rat brain. A 20 G needle size at a 10 µL/min flow rate achieved the most efficient microsphere ejection. Secondly, we optimized the delivery vehicles for in vivo implantation of PEG microspheres. The suspension of microspheres in extracellular matrix (ECM) hydrogel showed superior retention and distribution in a cortical stroke caused by photothrombosis, as well as in a striatal and cortical cavity ensuing middle cerebral artery occlusion (MCAo). Thirdly, NSCs or NSCs + endothelial cells (ECs) encapsulated into biodegradable microspheres were implanted into a large stroke cavity. Cells in microspheres exhibited a high viability, survived freezing and transport. Implantation of 110 cells/microsphere suspended in ECM hydrogel produced a highly efficient delivery that resulted in the widespread distribution of NSCs in the tissue cavity and damaged peri-infarct tissues. Co-delivery of ECs enhanced the in vivo survival and distribution of ∼1.1 million NSCs. The delivery of NSCs and ECs can be dramatically improved using microsphere encapsulation combined with suspension in ECM hydrogel. These biomaterial innovations are essential to advance clinical efforts to improve the treatment of stroke using intracerebral cell therapy.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Hidrogeles/farmacología , Microesferas , Células-Madre Neurales/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Matriz Extracelular/metabolismo , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Polietilenglicoles/farmacología , Accidente Cerebrovascular/metabolismo
15.
Sci Rep ; 10(1): 621, 2020 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-31932638

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

16.
Curr Stem Cell Rep ; 5(3): 109-114, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32864301

RESUMEN

PURPOSE OF REVIEW: Neural stem cells (NSCs) have the potential to proliferate and differentiate into functional neurons, heightening their potential use for therapeutic applications. This review explores bioengineered systems which recapitulate NSC niche cell-cell and cell-matrix interactions. RECENT FINDINGS: Delivery of NSCs to the cytotoxic injured brain is limited by low cell survival rates post-transplantation and poor maintenance of native niche bioactive components. The use of biomaterial platforms can mimic in vivo the environment of the two germinal areas of the adult brain in which NSCs thrive. An environmental mimic that includes extracellular proteins and moieties, along with appropriate biomechanical cues has recently demonstrated promising results in enhancing neurogenesis, aiding the production of a bioengineered niche. SUMMARY: Biocomposition, biomechanics, and biostructure can be manipulated through engineered platforms to re-create the biofunctionality of an NSC niche. Upon transplantation and delivery with biomimetic scaffolds, NSCs show potential to promote functional recovery and rebuild neural circuitry post neurological trauma.

17.
Sci Rep ; 9(1): 17798, 2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-31780709

RESUMEN

Stem cell therapies demonstrate promising results as treatment for neurological disease and injury, owing to their innate ability to enhance endogenous neural tissue repair and promote functional recovery. However, delivery of undifferentiated and viable neuronal stem cells requires an engineered delivery system that promotes integration of transplanted cells into the inflamed and cytotoxic region of damaged tissue. Within the brain, endothelial cells (EC) of the subventricular zone play a critical role in neural stem cell (NSC) maintenance, quiescence and survival. Therefore, here, we describe the use of polyethylene glycol microbeads for the coincident delivery of EC and NSC as a means of enhancing appropriate NSC quiescence and survival during transplantation into the mouse brain. We demonstrate that EC and NSC co-encapsulation maintained NSC quiescence, enhanced NSC viability, and facilitated NSC extravasation in vitro, as compared to NSC encapsulated alone. In addition, co-encapsulated cells delivered to an in vivo non-injury model reduced inflammatory response compared to freely injected NSC. These results suggest the strong potential of a biomimetic engineered niche for NSC delivery into the brain following neurological injury.


Asunto(s)
Encapsulación Celular/métodos , Ventrículos Laterales/cirugía , Microesferas , Células-Madre Neurales/trasplante , Trasplante de Células Madre/métodos , Animales , Diferenciación Celular , Línea Celular , Proliferación Celular , Supervivencia Celular , Células Endoteliales/metabolismo , Ventrículos Laterales/metabolismo , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/fisiología , Neuronas , Polietilenglicoles/química , Recuperación de la Función , Nicho de Células Madre
18.
J Leukoc Biol ; 81(3): 686-95, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17164427

RESUMEN

Transendothelial migration of neutrophils induces phenotypic changes that influence the interactions of neutrophils with extravascular tissue components. To assess the influence of transmigration on neutrophil chemokinetic motility, we used polyethylene glycol hydrogels covalently modified with specific peptide sequences relevant to extracellular matrix proteins. We evaluated fMLP-stimulated human neutrophil motility on peptides Arg-Gly-Asp-Ser (RGDS) and TMKIIPFNRTLIGG (P2), alone and in combination. RGDS is a bioactive sequence found in a number of proteins, and P2 is a membrane-activated complex-1 (Mac-1) ligand located in the gamma-chain of the fibrinogen protein. We evaluated, via video microscopy, cell motility by measuring cell displacement from origin and total accumulated distance traveled and then calculated average velocity. Results indicate that although adhesion and shape change were supported by hydrogels containing RGD alone, motility was not. Mac-1-dependent motility was supported on hydrogels containing P2 alone. Motility was enhanced through combined presentation of RGD and P2, engaging Mac-1, alpha(V)beta(3), and beta(1) integrins. Naïve neutrophil motility on combined peptide substrates was dependent on Mac-1, and alpha(4)beta(1) while alpha(6)beta(1) contributed to speed and linear movement. Transmigrated neutrophil motility was dependent on alpha(v)beta(3) and alpha(5)beta(1), and alpha(4)beta(1), alpha(6)beta(1), and Mac-1 contributed to speed and linear motion. Together, the data demonstrate that efficient neutrophil migration, dependent on multi-integrin interaction, is enhanced after transendothelial migration.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Células Endoteliales/inmunología , Integrinas/inmunología , Neutrófilos/inmunología , Movimiento Celular/inmunología , Células Cultivadas , Células Endoteliales/citología , Humanos , Hidrogeles , N-Formilmetionina Leucil-Fenilalanina/farmacología , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Oligopéptidos/farmacología , Péptidos/farmacología , Relación Estructura-Actividad
19.
PLoS One ; 12(2): e0171386, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28234918

RESUMEN

The vascular basement membrane-a thin, elastic layer of extracellular matrix separating and encasing vascular cells-provides biological and mechanical cues to endothelial cells, pericytes, and migrating leukocytes. In contrast, experimental scaffolds typically used to replicate basement membranes are stiff and bio-inert. Here, we present thin, porated polyethylene glycol hydrogels to replicate human vascular basement membranes. Like commercial transwells, our hydrogels are approximately 10µm thick, but like basement membranes, the hydrogels presented here are elastic (E: 50-80kPa) and contain a dense network of small pores. Moreover, the inclusion of bioactive domains introduces receptor-mediated biochemical signaling. We compare elastic hydrogels to common culture substrates (E: >2GPa) for human endothelial cell and pericyte monolayers and bilayers to replicate postcapillary venules in vitro. Our data demonstrate that substrate elasticity facilitates differences in vascular phenotype, supporting expression of vascular markers that are increasingly replicative of venules. Endothelial cells differentially express vascular markers, like EphB4, and leukocyte adhesion molecules, such as ICAM-1, with decreased mechanical stiffness. With porated PEG hydrogels we demonstrate the ability to evaluate and observe leukocyte recruitment across endothelial cell and pericyte monolayers and bilayers, reporting that basement membrane scaffolds can significantly alter the rate of vascular migration in experimental systems. Overall, this study demonstrates the creation and utility of a new and accessible method to recapture the mechanical and biological complexity of human basement membranes in vitro.


Asunto(s)
Membrana Basal/química , Células Endoteliales/citología , Neutrófilos/citología , Pericitos/citología , Ingeniería de Tejidos/métodos , Membrana Basal/metabolismo , Membrana Basal/ultraestructura , Biomarcadores/metabolismo , Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Movimiento Celular , Módulo de Elasticidad , Elasticidad , Células Endoteliales/metabolismo , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Expresión Génica , Humanos , Hidrogeles/química , Hidrogeles/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Neutrófilos/metabolismo , Pericitos/metabolismo , Polietilenglicoles/química , Porosidad , Cultivo Primario de Células , Receptor EphB4/genética , Receptor EphB4/metabolismo , Transducción de Señal
20.
J Vis Exp ; (130)2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29364202

RESUMEN

The basement membrane is a critical component of cellular bilayers that can vary in stiffness, composition, architecture, and porosity. In vitro studies of endothelial-epithelial bilayers have traditionally relied on permeable support models that enable bilayer culture, but permeable supports are limited in their ability to replicate the diversity of human basement membranes. In contrast, hydrogel models that require chemical synthesis are highly tunable and allow for modifications of both the material stiffness and the biochemical composition via incorporation of biomimetic peptides or proteins. However, traditional hydrogel models are limited in functionality because they lack pores for cell-cell contacts and functional in vitro migration studies. Additionally, due to the thickness of traditional hydrogels, incorporation of pores that span the entire thickness of hydrogels has been challenging. In the present study, we use poly-(ethylene-glycol) (PEG) hydrogels and a novel zinc oxide templating method to address the previous shortcomings of biomimetic hydrogels. As a result, we present an ultrathin, basement membrane-like hydrogel that permits the culture of confluent cellular bilayers on a customizable scaffold with variable pore architectures, mechanical properties, and biochemical composition.


Asunto(s)
Membrana Basal/metabolismo , Materiales Biomiméticos/química , Biomimética/métodos , Técnicas de Cultivo de Célula/métodos , Hidrogeles/química , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Polietilenglicoles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA