Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Physiol ; 11: 1026, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33013445

RESUMO

Vascular connectivity between adjacent vessel beds within and between tissue compartments is essential to any successful neovascularization process. To establish new connections, growing neovessels must locate other vascular elements during angiogenesis, often crossing matrix and other tissue-associated boundaries and interfaces. How growing neovessels traverse any tissue interface, whether part of the native tissue structure or secondary to a regenerative procedure (e.g., an implant), is not known. In this study, we developed an experimental model of angiogenesis wherein growing neovessels must interact with a 3D interstitial collagen matrix interface that separates two distinct tissue compartments. Using this model, we determined that matrix interfaces act as a barrier to neovessel growth, deflecting growing neovessels parallel to the interface. Computational modeling of the neovessel/matrix biomechanical interactions at the interface demonstrated that differences in collagen fibril density near and at the interface are the likely mechanism of deflection, while fibril alignment guides deflected neovessels along the interface. Interestingly, stromal cells facilitated neovessel interface crossing during angiogenesis via a vascular endothelial growth factor (VEGF)-A dependent process. However, ubiquitous addition of VEGF-A in the absence of stromal cells did not promote interface invasion. Therefore, our findings demonstrate that vascularization of a tissue via angiogenesis involves stromal cells providing positional cues to the growing neovasculature and provides insight into how a microvasculature is organized within a tissue.

2.
Immunol Cell Biol ; 98(8): 650-666, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32392367

RESUMO

The metabolically dynamic nature of healthy adipose places this tissue under regular inflammatory stress. A network of adipose-resident anti-inflammatory immune cells modulates and resolves this endogenous inflammation. Previous work in our laboratory identified a CD11b+ Gr1+ subset of these immunosuppressive adipose stromal cells in healthy mice. Myeloid-derived suppressor cells (MDSCs), typically associated with cancer and chronic inflammation, have a similar surface marker phenotype and accumulate in adipose of high-fat diet-fed mice. Given the routine inflammatory stresses on healthy adipose and the suppressive nature of the tissue-resident immune cells, we hypothesized that these CD11b+ Gr1+ cells were a genuine population of MDSCs involved in regulating tissue homeostasis. Flow cytometric analysis of these cells found that they were CD11b+ CD301- Ly6C+ Ly6G+/- and did not express traditional macrophage markers. Moreover, in vitro functional assays demonstrated that these cells suppressed αCD3/αCD28-induced T-cell proliferation, solidifying their identity as bona fide adipose-resident MDSCs. Systemic MDSC depletion altered adipose immune cell dynamics in otherwise healthy mice, increasing the number of CD4+ effector memory T cells and modifying the surface markers expressed by adipose-resident macrophages. In addition, transcription of various immunomodulatory cytokines was clearly dysregulated in the adipose of MDSC-depleted animals compared with controls. Altogether, our findings indicate that there is a population of bona fide MDSCs in the adipose of otherwise healthy mice that actively contribute to the health and immune homeostasis of this tissue.


Assuntos
Tecido Adiposo/imunologia , Homeostase/imunologia , Células Supressoras Mieloides , Animais , Antígeno CD11b , Citocinas , Ativação Linfocitária , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Células Supressoras Mieloides/imunologia , Linfócitos T
3.
Stem Cells Transl Med ; 4(4): 369-80, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25722428

RESUMO

Vasoactivity, an important aspect of tissue healing, is often compromised in disease and tissue injury. Dysfunction in the smaller vasoactive arteries is most impactful, given the role of these vessels in controlling downstream tissue perfusion. The adipose stromal vascular fraction (SVF) is a mix of homeostatic cells shown to promote tissue healing. Our objective was to test the hypothesis that autologous SVF cells therapeutically modulate peripheral artery vasoactivity in syngeneic mouse models of small artery function. Analysis of vasoactivity of saphenous arteries isolated from normal mice 1 week after intravenous injection of freshly isolated SVF cells revealed that pressure-dependent artery vasomotor tone was decreased by the SVF cell isolate, but not one depleted of CD11b(+) cells. Scavenging hydrogen peroxide in the vessel wall abrogated the artery relaxation promoted by the SVF cell isolate. Consistent with a CD11b(+) cell being the relevant cell type, SVF-derived F4/80-positive macrophages were present within the adventitia of the artery wall coincident with vasorelaxation. In a model of artery inflammation mimicking a common disease condition inducing vasoactive dysfunction, the SVF cells potentiated relaxation of saphenous arteries without structurally remodeling the artery via a CD11b(+) cell-dependent manner. Our findings demonstrate that freshly isolated, adipose SVF cells promote vasomotor relaxation in vasoactive arteries via a hydrogen peroxide-dependent mechanism that required CD11b(+) cells (most likely macrophages). Given the significant impact of small artery dysfunction in disease, we predict that the intravenous delivery of this therapeutic cell preparation would significantly improve tissue perfusion, particularly in diseases with diffuse vascular involvement.


Assuntos
Tecido Adiposo/citologia , Artérias/citologia , Células Estromais/citologia , Sistema Vasomotor/metabolismo , Adipócitos/citologia , Animais , Artérias/metabolismo , Antígeno CD11b/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos
4.
Cell Transplant ; 24(10): 2029-39, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25397993

RESUMO

Adipose-derived stromal vascular fraction (SVF) cells have been shown to self-associate to form vascular structures under both in vitro and in vivo conditions. The angiogenic (new vessels from existing vessels) and vasculogenic (new vessels through self-assembly) potential of the SVF cell population may provide a cell source for directly treating (i.e., point of care without further cell isolation) ischemic tissues. However the correct dosage of adipose SVF cells required to achieve a functional vasculature has not been established. Accordingly, in vitro and in vivo dose response assays were performed evaluating the SVF cell vasculogenic potential. Serial dilutions of freshly isolated rat adipose SVF cells were plated on growth factor reduced Matrigel and vasculogenesis, assessed as cellular tube-like network assembly, was quantified after 3 days of culture. This in vitro vasculogenesis assay indicated that rat SVF cells reached maximum network length at a concentration of 2.5 × 10(5) cells/ml and network maintained at the higher concentrations tested. The same concentrations of rat and human SVF cells were used to evaluate vasculogenesis in vivo. SVF cells were incorporated into collagen gels and subcutaneously implanted into Rag1 immunodeficient mice. The 3D confocal images of harvested constructs were evaluated to quantify dose dependency of SVF cell vasculogenesis potential. Rat- and human-derived SVF cells yielded a maximum vasculogenic potential at 1 × 10(6) and 4 × 10(6) cells/ml, respectively. No adverse reactions (e.g., toxicity, necrosis, tumor formation) were observed at any concentration tested. In conclusion, the vasculogenic potential of adipose-derived SVF cell populations is dose dependent.


Assuntos
Adipócitos/citologia , Tecido Adiposo/citologia , Diferenciação Celular/fisiologia , Neovascularização Patológica/patologia , Células Estromais/citologia , Adiposidade/fisiologia , Animais , Células Cultivadas , Humanos , Camundongos , Neovascularização Patológica/metabolismo , Ratos
5.
Tissue Eng Part C Methods ; 19(4): 307-15, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22966764

RESUMO

The three-dimensional culture of blood vessel wall cells now permits the construction of a human blood vessel mimic (BVM). Previous studies have used the human BVM as a tool to perform in vitro testing of medical devices and imaging instrumentation. The purpose of the current study was to enhance this technology through both the elimination of animal serum and the modification of scaffold properties in human BVM preparation. Additionally, BVMs were implanted with vascular stents to observe a potential cellular response to the devices in a serum-free environment. Serum-free culture of human adipose-derived stromal vascular fraction (SVF) cells was accomplished through sequential adaptation from a serum-supplemented medium. The adipose-derived SVF serves as a source of both human endothelium and human smooth muscle cells. Utilizing established pressure-sodding technologies, these cells were incorporated into the luminal surface of either expanded polytetrafluoroethylene (ePTFE) tubular scaffolds or electrospun poly(l-lactide-co-caprolactone) scaffolds, and the resulting constructs were cultivated in a perfusion bioreactor using a serum-free medium. Histological analysis of BVMs created using ePTFE scaffolds indicated that a complete lining of cells had formed on the inner surfaces of the grafts. Vessel mimics were also established under serum-free conditions on the highly porous electrospun tubes, resulting in cellularization throughout the scaffold wall in addition to inner and outer surfaces. Neither endothelial cells nor smooth muscles cells were identified among the mesenchymal cells present in each type of BVM. Bare metal stents were deployed within the electrospun BVMs, and after bioreactor perfusion, scanning electron microscopy and nuclear-specific bisbenzimide staining confirmed the presence of cells on stent surfaces. The outcomes of this study support the hypothesis that BVMs developed using serum-free conditions are affected by scaffold variations and exhibit tissue growth over implanted medical devices. Ultimately, employing serum-free methods could lead to controlled, reproducible BVM production and interpretable, human-specific results in studies of device-tissue interaction, toxicity, and other vascular phenomena; however, comparisons to in vivo biological responses and incorporation of defined blood vessel cells will be critical to validating the serum-free BVM as an appropriate device testing alternative.


Assuntos
Bioengenharia , Vasos Sanguíneos , Meios de Cultura Livres de Soro , Equipamentos e Provisões , Mimetismo Molecular , Alicerces Teciduais , Animais , Reatores Biológicos , Humanos , Imuno-Histoquímica , Microscopia Eletrônica de Varredura
6.
Microcirculation ; 17(7): 557-67, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21040121

RESUMO

UNLABELLED: We have demonstrated that MFs isolated from adipose retain angiogenic potential in vitro and form a mature, perfused network when implanted. However, adipose-derived microvessels are rich in provascularizing cells that could uniquely drive neovascularization in adipose-derived MFs implants. OBJECTIVE: Investigate the ability of MFs from a different vascular bed to recapitulate adipose-derived microvessel angiogenesis and network formation and analyze adipose-derived vessel plasticity by assessing whether vessel function could be modulated by astrocyte-like cells. METHODS: MFs were isolated by limited collagenase digestion from rodent brain or adipose and assembled into 3D collagen gels in the presence or absence of GRPs. The resulting neovasculatures that formed following implantation were assessed by measuring 3D vascularity and vessel permeability to small and large molecular tracers. RESULTS: Similar to adipose-derived MFs, brain-derived MFs can sprout and form a perfused neovascular network when implanted. Furthermore, when co-implanted in the constructs, GRPs caused adipose-derived vessels to express the brain endothelial marker glucose transporter-1 and to significantly reduce microvessel permeability. CONCLUSION: Neovascularization involving isolated microvessel elements is independent of the tissue origin and degree of vessel specialization. In addition, adipose-derived vessels have the ability to respond to environmental signals and change vessel characteristics.


Assuntos
Microvasos/crescimento & desenvolvimento , Microvasos/transplante , Neovascularização Fisiológica , Adipócitos/citologia , Adipócitos/transplante , Animais , Astrócitos/citologia , Permeabilidade Capilar , Separação Celular , Córtex Cerebral/irrigação sanguínea , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/transplante , Epididimo/irrigação sanguínea , Epididimo/citologia , Técnicas In Vitro , Masculino , Microvasos/citologia , Microvasos/fisiologia , Neuroglia/citologia , Neuroglia/transplante , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...