Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Theranostics ; 12(2): 558-573, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34976201

RESUMO

Background: Microcirculation is essential for skin homeostasis and repair. A variety of growth factors have been identified as important regulators of wound healing. However, direct observation and longitudinal monitoring of skin remodeling in an unperturbed in vivo environment remains challenging. Methods: We report on non-invasive longitudinal imaging of the wound healing process in transgenic mice overexpressing vascular endothelial growth factor A (VEGF-A) in keratinocytes by means of large-scale optoacoustic microscopy (LSOM). This rapid, label-free, high throughput intravital microscopy method averts the use of dorsal skin-fold chambers, allowing for fully non-invasive repeated imaging of intact wounds with capillary resolution over field-of-view spanning several centimeters. Results: We observed VEGF-driven enhancement of dermal vascularization in ears, dorsal skin and healing wounds and quantified the hemoglobin content, fill fraction, vessel diameter and tortuosity. The in vivo findings were further corroborated by detailed side-by-side classical histological whole-mount vascular stainings and pan-endothelial CD31 immunofluorescence. Conclusion: The new approach is suitable for supplementing or replacing the cumbersome histological procedures in a broad range of skin regeneration and tissue engineering applications.


Assuntos
Pele/lesões , Fator A de Crescimento do Endotélio Vascular/fisiologia , Cicatrização/fisiologia , Animais , Feminino , Estudos Longitudinais , Camundongos , Camundongos Transgênicos , Microscopia/métodos , Microvasos/diagnóstico por imagem , Microvasos/crescimento & desenvolvimento , Neovascularização Fisiológica , Técnicas Fotoacústicas , Pele/diagnóstico por imagem , Fenômenos Fisiológicos da Pele , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
PLoS Comput Biol ; 17(6): e1009164, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34170925

RESUMO

The vasculature is a dynamic structure, growing and regressing in response to embryonic development, growth, changing physiological demands, wound healing, tumor growth and other stimuli. At the microvascular level, network geometry is not predetermined, but emerges as a result of biological responses of each vessel to the stimuli that it receives. These responses may be summarized as angiogenesis, remodeling and pruning. Previous theoretical simulations have shown how two-dimensional vascular patterns generated by these processes in the mesentery are consistent with experimental observations. During early development of the brain, a mesh-like network of vessels is formed on the surface of the cerebral cortex. This network then forms branches into the cortex, forming a three-dimensional network throughout its thickness. Here, a theoretical model is presented for this process, based on known or hypothesized vascular response mechanisms together with experimentally obtained information on the structure and hemodynamics of the mouse cerebral cortex. According to this model, essential components of the system include sensing of oxygen levels in the midrange of partial pressures and conducted responses in vessel walls that propagate information about metabolic needs of the tissue to upstream segments of the network. The model provides insights into the effects of deficits in vascular response mechanisms, and can be used to generate physiologically realistic microvascular network structures.


Assuntos
Córtex Cerebral/irrigação sanguínea , Modelos Cardiovasculares , Modelos Neurológicos , Neovascularização Fisiológica , Animais , Córtex Cerebral/crescimento & desenvolvimento , Biologia Computacional , Simulação por Computador , Hemodinâmica/fisiologia , Camundongos , Microcirculação/fisiologia , Microvasos/anatomia & histologia , Microvasos/crescimento & desenvolvimento , Microvasos/fisiologia , Consumo de Oxigênio
3.
Biosci Rep ; 40(10)2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-33015716

RESUMO

As a negative immune checkpoint molecule, T-cell immunoglobulin domain and mucin domain containing molecule-3 (Tim-3) has been found to serve a crucial role in immune escape and tumour progression. Previous studies have reported that Tim-3 is important to endothelial cells and it has also been demonstrated to be involved in numerous types of human diseases, including melanoma, lymphoma, rickettsial infection and atherosclerosis; however, its exact mechanism of action remains largely unknown. In the present study, Tim-3 was overexpressed in vascular endothelial human lung microvascular endothelial cells (HMVECs) and human umbilical vein endothelial cells (HUVECs), and in vitro assays were used to determine that Tim-3 promoted cell proliferation, migration, invasion and tube formation through activating cyclin D1 (CCND1), Ras homolog gene family member A and vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2). Additionally, Tim-3 decreased tight junction (TJ) formation and the transepithelial resistance (TER) of endothelial cells by decreasing the expression levels of TJ protein 2, Occludin and claudin 1 (CLND1). In conclusion, these findings suggested that Tim-3 may exert a positive role in angiogenesis and a negative role in TJ formation in vascular endothelial cells, which may provide novel strategies for the treatment of Tim-3-associated diseases.


Assuntos
Endotélio Vascular/crescimento & desenvolvimento , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Neovascularização Fisiológica , Junções Íntimas/metabolismo , Linhagem Celular , Movimento Celular , Proliferação de Células , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Receptor Celular 2 do Vírus da Hepatite A/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Pulmão/irrigação sanguínea , Microvasos/citologia , Microvasos/crescimento & desenvolvimento , Microvasos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção
4.
J Exp Clin Cancer Res ; 39(1): 193, 2020 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-32958011

RESUMO

BACKGROUND: Angiogenesis, a basic requirement for tumor cell survival, is considered to be a malignant characteristic of small cell lung cancer (SCLC) and is closely related to the poor outcomes of SCLC patients. miR-141 has been found to play pro- and antiangiogenic roles in different cancers, but its role in SCLC angiogenesis has never been explored. METHODS: Total RNA was isolated from plasm exosomes and serum of SCLC patients to examine the expression of miR-141 by qRT-PCR. Cell proliferation, invasion, migration, tube formation assay, aortic ring assay and mouse tumor model were used to investigate the effect of exosomal miR-141 in angiogenesis in vitro and in vivo. Dual-luciferase assay was conducted to explore the target gene of miR-141. RESULTS: Circulating miR-141 was upregulated in samples from 122 SCLC patients compared with those from normal volunteers and that the increase in miR-141 was significantly associated with advanced TNM stages, implying the potential oncogenic role of miR-141 in SCLC malignancy. In vitro, miR-141 that was packaged into SCLC cell-secreted exosomes and delivered to human umbilical vein vascular endothelial cells (HUVECs) via exosomes facilitated HUVEC proliferation, invasion, migration and tube formation and promoted microvessel sprouting from mouse aortic rings. Matrigel plug assays demonstrated that SCLC cell-derived exosomal miR-141 induced neoangiogenesis in vivo. Furthermore, mouse subcutaneous tumor nodules that were developed from miR-141-overexpressing SCLC cells had a higher microvessel density (MVD) and grew faster than those developed from negative control cells. KLF12 was found to be the direct target gene of miR-141 and that the proangiogenic effect of miR-141 on HUVECs was abrogated by KLF12 overexpression. CONCLUSIONS: Our results demonstrate the specific function of the exosomal miR-141/KLF12 pathway in SCLC angiogenesis for the first time and provide potential novel targets for antiangiogenic therapies for SCLC patients.


Assuntos
Fatores de Transcrição Kruppel-Like/genética , MicroRNAs/genética , Neovascularização Patológica/genética , Carcinoma de Pequenas Células do Pulmão/genética , Animais , Movimento Celular/genética , Proliferação de Células/genética , Exossomos/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Xenoenxertos , Células Endoteliais da Veia Umbilical Humana , Humanos , Fatores de Transcrição Kruppel-Like/sangue , Masculino , Camundongos , MicroRNAs/sangue , Microvasos/crescimento & desenvolvimento , Microvasos/patologia , Pessoa de Meia-Idade , Neovascularização Patológica/sangue , Neovascularização Patológica/patologia , Transdução de Sinais/genética , Carcinoma de Pequenas Células do Pulmão/sangue , Carcinoma de Pequenas Células do Pulmão/patologia
5.
Sci Rep ; 10(1): 15562, 2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32968145

RESUMO

Revascularization of ischemic tissues is a major barrier to restoring tissue function in many pathologies. Delivery of pro-angiogenic factors has shown some benefit, but it is difficult to recapitulate the complex set of factors required to form stable vasculature. Cell-based therapies and pre-vascularized tissues have shown promise, but the former require time for vascular assembly in situ while the latter require invasive surgery to implant vascularized scaffolds. Here, we developed cell-laden fibrin microbeads that can be pre-cultured to form primitive vascular networks within the modular structures. These microbeads can be delivered in a minimally invasive manner and form functional microvasculature in vivo. Microbeads containing endothelial cells and stromal fibroblasts were pre-cultured for 3 days in vitro and then injected within a fibrin matrix into subcutaneous pockets on the dorsal flanks of SCID mice. Vessels deployed from these pre-cultured microbeads formed functional connections to host vasculature within 3 days and exhibited extensive, mature vessel coverage after 7 days in vivo. Cellular microbeads showed vascularization potential comparable to bulk cellular hydrogels in this pilot study. Furthermore, our findings highlight some potentially advantageous characteristics of pre-cultured microbeads, such as volume preservation and vascular network distribution, which may be beneficial for treating ischemic diseases.


Assuntos
Fibrina/farmacologia , Hidrogéis/farmacologia , Neovascularização Fisiológica , Engenharia Tecidual , Animais , Células Cultivadas , Fibrina/química , Fibroblastos/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Hidrogéis/química , Camundongos , Microesferas , Microvasos/efeitos dos fármacos , Microvasos/crescimento & desenvolvimento , Alicerces Teciduais/química
6.
Cancer Res ; 80(19): 4288-4301, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32665356

RESUMO

In solid tumors, vascular structure and function varies from the core to the periphery. This structural heterogeneity has been proposed to influence the mechanisms by which tumor cells enter the circulation. Blood vessels exhibit regional defects in endothelial coverage, which can result in cancer cells directly exposed to flow and potentially promoting intravasation. Consistent with prior reports, we observed in human breast tumors and in a mouse model of breast cancer that approximately 6% of vessels consisted of both endothelial cells and tumor cells, so-called mosaic vessels. Due, in part, to the challenges associated with observing tumor-vessel interactions deep within tumors in real-time, the mechanisms by which mosaic vessels form remain incompletely understood. We developed a tissue-engineered model containing a physiologically realistic microvessel in coculture with mammary tumor organoids. This approach allows real-time and quantitative assessment of tumor-vessel interactions under conditions that recapitulate many in vivo features. Imaging revealed that tumor organoids integrate into the endothelial cell lining, resulting in mosaic vessels with gaps in the basement membrane. While mosaic vessel formation was the most frequently observed interaction, tumor organoids also actively constricted and displaced vessels. Furthermore, intravasation of cancer cell clusters was observed following the formation of a mosaic vessel. Taken together, our data reveal that cancer cells can rapidly reshape, destroy, or integrate into existing blood vessels, thereby affecting oxygenation, perfusion, and systemic dissemination. Our novel assay also enables future studies to identify targetable mechanisms of vascular recruitment and intravasation. SIGNIFICANCE: A tissue-engineered microdevice that recapitulates the tumor-vascular microenvironment enables real-time imaging of the cellular mechanisms of mosaic vessel formation and vascular defect generation.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Microvasos/crescimento & desenvolvimento , Engenharia Tecidual/métodos , Animais , Morte Celular , Proliferação de Células , Técnicas de Cocultura , Células Endoteliais/patologia , Endotélio Vascular/citologia , Endotélio Vascular/patologia , Feminino , Imunofluorescência , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos Endogâmicos NOD , Microvasos/patologia , Modelos Biológicos , Células Neoplásicas Circulantes/patologia , Organoides/crescimento & desenvolvimento , Engenharia Tecidual/instrumentação
7.
Nat Commun ; 10(1): 4602, 2019 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-31601796

RESUMO

The success of engineered cell or tissue implants is dependent on vascular regeneration to meet adequate metabolic requirements. However, development of a broadly applicable strategy for stable and functional vascularization has remained challenging. We report here highly organized and resilient microvascular meshes fabricated through a controllable anchored self-assembly method. The microvascular meshes are scalable to centimeters, almost free of defects and transferrable to diverse substrates, ready for transplantation. They promote formation of functional blood vessels, with a density as high as ~220 vessels mm-2, in the poorly vascularized subcutaneous space of SCID-Beige mice. We further demonstrate the feasibility of fabricating microvascular meshes from human induced pluripotent stem cell-derived endothelial cells, opening a way to engineer patient-specific microvasculature. As a proof-of-concept for type 1 diabetes treatment, we combine microvascular meshes and subcutaneously transplanted rat islets and achieve correction of chemically induced diabetes in SCID-Beige mice for 3 months.


Assuntos
Técnicas de Cultura de Células/instrumentação , Diabetes Mellitus Experimental/terapia , Transplante das Ilhotas Pancreáticas/métodos , Microvasos/crescimento & desenvolvimento , Animais , Bioengenharia , Técnicas de Cultura de Células/métodos , Diabetes Mellitus Experimental/complicações , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Hiperglicemia/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Transplante das Ilhotas Pancreáticas/instrumentação , Masculino , Camundongos SCID , Microvasos/citologia , Microvasos/fisiologia , Neovascularização Fisiológica , Ratos Sprague-Dawley
8.
Tissue Eng Part C Methods ; 25(8): 447-458, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31280703

RESUMO

IMPACT STATEMENT: Microvascular remodeling, or angiogenesis, plays a central role in multiple pathological conditions, including cancer, diabetes, and ischemia. Tissue-engineered in vitro models have emerged as tools to elucidate the mechanisms that drive the angiogenic process. However, a major challenge with model development is recapitulating the physiological complexity of real microvascular networks, including incorporation of the entire vascular tree and hemodynamics. This study establishes a bioreactor system that incorporates real microvascular networks with physiological flow as a novel ex vivo tissue culture model, thereby providing a platform to evaluate angiogenesis in a physiologically relevant environment.


Assuntos
Reatores Biológicos , Microvasos/crescimento & desenvolvimento , Neovascularização Fisiológica , Animais , Microvasos/citologia , Ratos , Ratos Wistar , Técnicas de Cultura de Tecidos
9.
J Cell Biochem ; 120(10): 16786-16797, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31104315

RESUMO

Cerebral ischemic stroke is a devastating neurological disease with high rates of morbidity, disability, and mortality. Lentiviral-mediated mast cell-expressed membrane protein 1 (MCEMP1) has been shown to function in ischemic stroke. Hence, this study aims to explore the function of MCEMP1 specifically in angiogenesis, neuronal proliferation, and apoptosis in rats with cerebral ischemic stroke. Initially, stroke-related genes were obtained through microarray-based gene expression analysis, followed by the construction of a lentiviral vector for MCEMP1 shRNA and establishment of the middle cerebral artery occlusion model. After rats were transfected with MCEMP1 shRNA lentivirus, microvessel density (MVD), expression of MCEMP1, caspase-3, and vascular endothelial growth factor (VEGF), and neuronal proliferation and apoptosis were measured to explore the role of MCEMP1 in cerebral ischemic stroke. MCEMP1 was found to be highly expressed in rats with cerebral ischemic stroke. Silencing of MCEMP1 led to upregulation of VEGF, while downregulation of caspase-3, and resulted in the promotion of MVD in rats with ischemic stroke. Moreover, MCEMP1 silencing could increase Ki67 positive cells and reduce terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling positive cells in the marginal zone of cortical infarction in rats. Our study provides evidence that silenced MCEMP1 could enhance angiogenesis and suppress neuronal apoptosis in rats with cerebral ischemic stroke, highlighting that MCEMP1 silencing could serve as a therapeutic target for cerebral ischemic stroke treatment.


Assuntos
Mastócitos/metabolismo , Proteínas de Membrana/fisiologia , Microvasos/crescimento & desenvolvimento , Neovascularização Fisiológica/genética , Acidente Vascular Cerebral/patologia , Animais , Apoptose/genética , Caspase 3/biossíntese , Proliferação de Células/genética , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/patologia , Lentivirus/genética , Masculino , Proteínas de Membrana/genética , Neurônios/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/genética , Fator A de Crescimento do Endotélio Vascular/biossíntese
10.
Sci Rep ; 9(1): 2190, 2019 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-30778117

RESUMO

Regulating the intrinsic interactions between blood vessels and nerve cells has the potential to enhance repair and regeneration of the central nervous system. Here, we evaluate the efficacy of aligned microvessels to induce and control directional axon growth from neural progenitor cells in vitro and host axons in a rat spinal cord injury model. Interstitial fluid flow aligned microvessels generated from co-cultures of cerebral-derived endothelial cells and pericytes in a three-dimensional scaffold. The endothelial barrier function was evaluated by immunostaining for tight junction proteins and quantifying the permeability coefficient (~10-7 cm/s). Addition of neural progenitor cells to the co-culture resulted in the extension of Tuj-positive axons in the direction of the microvessels. To validate these findings in vivo, scaffolds were transplanted into an acute spinal cord hemisection injury with microvessels aligned with the rostral-caudal direction. At three weeks post-surgery, sagittal sections indicated close alignment between the host axons and the transplanted microvessels. Overall, this work demonstrates the efficacy of exploiting neurovascular interaction to direct axon growth in the injured spinal cord and the potential to use this strategy to facilitate central nervous system regeneration.


Assuntos
Orientação de Axônios/fisiologia , Regeneração Nervosa/fisiologia , Animais , Técnicas de Cocultura , Modelos Animais de Doenças , Células Endoteliais/fisiologia , Feminino , Regeneração Tecidual Guiada , Técnicas In Vitro , Microvasos/crescimento & desenvolvimento , Microvasos/fisiologia , Células-Tronco Neurais/fisiologia , Células-Tronco Neurais/transplante , Ratos , Ratos Sprague-Dawley , Medula Espinal/irrigação sanguínea , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Alicerces Teciduais
11.
Macromol Biosci ; 19(4): e1800346, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30624848

RESUMO

Modulation of material properties and growth factor application are critical in constructing suitable cell culture environments to induce desired cellular functions. Sulfonated polyrotaxane (PRX) surfaces with immobilized vascular endothelial growth factors (VEGFs) are prepared to improve network formation in vascular endothelial cells. Sulfonated PRXs, whereby sulfonated α-cyclodextrins (α-CDs) are threaded onto a linear poly(ethylene glycol) chain capped with bulky groups at both terminals, are coated onto surfaces. The molecular mobility of sulfonated PRX surfaces is modulated by tuning the number of threading α-CDs. VEGF is immobilized onto surfaces with varying mobility. Low mobility and VEGF-immobilization reinforce cell proliferation, yes-associated protein activity, and rhoA, pdgf, ang-1, and pecam-1 gene expression. Highly mobile surfaces and soluble VEGF weakly affect these cell responses. Network formation is strongly stimulated in vascular endothelial cells only on low-mobility VEGF-immobilized surfaces, suggesting that molecular mobility and VEGF immobilization synergistically control cell function.


Assuntos
Materiais Biocompatíveis/química , Ciclodextrinas/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Proteínas Imobilizadas , Microvasos/crescimento & desenvolvimento , Poloxâmero/química , Rotaxanos/química , Fator A de Crescimento do Endotélio Vascular , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Proteínas Imobilizadas/química , Proteínas Imobilizadas/farmacologia , Polietilenoglicóis/química , Propriedades de Superfície , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/farmacologia , alfa-Ciclodextrinas/química
12.
Acta Biomater ; 77: 201-211, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30030175

RESUMO

Insufficient vascularization is a major cause for the development of non-unions. To overcome this problem, adipose tissue-derived microvascular fragments (MVF) may serve as vascularization units. However, their application into bone defects needs a carrier system. Herein, we analyzed whether this is achieved by a thermoresponsive hydrogel (TRH). MVF were isolated from CD-1 mice and cultivated after incorporation into TRH, while non-incorporated MVF served as controls. Viability of MVF was assessed immunohistochemically over a 7-day period. Moreover, osteotomies were induced in femurs of CD-1 mice. The osteotomy gaps were filled with MVF-loaded TRH (TRH + MVF), unloaded TRH (TRH) or no material (control). Bone healing was evaluated 14 and 35 days postoperatively. MVF incorporated into TRH exhibited less apoptotic cells and showed a stable vessel morphology compared to controls. Micro-computed tomography revealed a reduced bone volume in TRH + MVF femurs. Histomorphometry showed less bone and more fibrous tissue after 35 days in TRH + MVF femurs compared to controls. Accordingly, TRH + MVF femurs exhibited a lower osseous bridging score and a reduced bending stiffness. Histology and Western blot analysis revealed an increased vascularization and CD31 expression, whereas vascular endothelial growth factor (VEGF) expression was reduced in TRH + MVF femurs. Furthermore, the callus of TRH + MVF femurs showed increased receptor activator of NF-κB ligand expression and higher numbers of osteoclasts. These findings indicate that TRH is an appropriate carrier system for MVF. Application of TRH + MVF increases the vascularization of bone defects. However, this impairs bone healing, most likely due to lower VEGF expression during the early course of bone healing. STATEMENT OF SIGNIFICANCE: In the present study we analyzed for the first time the in vivo performance of a thermoresponsive hydrogel (TRH) as a delivery system for bioactive microvascular fragments (MVF). We found that TRH represents an appropriate carrier for MVF as vascularization units and maintains their viability. Application of MVF-loaded TRH impaired bone formation in an established murine model of bone healing, although vascularization was improved. This unexpected outcome was most likely due to a reduced VEGF expression in the early phase bone healing.


Assuntos
Tecido Adiposo/citologia , Regeneração Óssea , Hidrogéis/química , Microcirculação , Microvasos/crescimento & desenvolvimento , Animais , Calo Ósseo/patologia , Elasticidade , Fêmur/patologia , Consolidação da Fratura , Masculino , Camundongos , Neovascularização Fisiológica , Osteoclastos/metabolismo , Osteotomia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Resistência ao Cisalhamento , Fator A de Crescimento do Endotélio Vascular/metabolismo , Viscosidade , Microtomografia por Raio-X
13.
JCI Insight ; 3(4)2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29467323

RESUMO

Von Hippel-Lindau (VHL) gene mutations induce neural tissue hemangioblastomas, as well as highly vascularized clear cell renal cell carcinomas (ccRCCs). Pathological vessel remodeling arises from misregulation of HIFs and VEGF, among other genes. Variation in disease penetrance has long been recognized in relation to genotype. We show Vhl mutations also disrupt Notch signaling, causing mutation-specific vascular abnormalities, e.g., type 1 (null) vs. type 2B (murine G518A representing human R167Q). In conditional mutation retina vasculature, Vhl-null mutation (i.e., UBCCreER/+Vhlfl/fl) had little effect on initial vessel branching, but it severely reduced arterial and venous branching at later stages. Interestingly, this mutation accelerated arterial maturation, as observed in retina vessel morphology and aberrant α-smooth muscle actin localization, particularly in vascular pericytes. RNA sequencing analysis identified gene expression changes within several key pathways, including Notch and smooth muscle cell contractility. Notch inhibition failed to reverse later-stage branching defects but rescued the accelerated arterialization. Retinal vessels harboring the type 2B Vhl mutation (i.e., UBCCreER/+Vhlfl/2B) displayed stage-specific changes in vessel branching and an advanced progression toward an arterial phenotype. Disrupting Notch signaling in type 2B mutants increased both artery and vein branching and restored arterial maturation toward nonmutant levels. By revealing differential effects of the null and type 2B Vhl mutations on vessel branching and maturation, these data may provide insight into the variability of VHL-associated vascular changes - particularly the heterogeneity and aggressiveness in ccRCC vessel growth - and also suggest Notch pathway targets for treating VHL syndrome.


Assuntos
Células Endoteliais/patologia , Hemangioblastoma/patologia , Receptores Notch/metabolismo , Artéria Retiniana/crescimento & desenvolvimento , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Doença de von Hippel-Lindau/patologia , Animais , Diaminas/farmacologia , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Hemangioblastoma/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Microvasos/citologia , Microvasos/crescimento & desenvolvimento , Microvasos/patologia , Mutação , Receptores Notch/antagonistas & inibidores , Artéria Retiniana/citologia , Artéria Retiniana/patologia , Tiazóis/farmacologia , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Doença de von Hippel-Lindau/genética
14.
Cancer Chemother Pharmacol ; 81(1): 207-216, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29196964

RESUMO

PURPOSE: To investigate the optimal starting time point of sorafenib therapy in suppressing the tumor-promoting effects of VEGF up-regulation, which is frequently found after local therapy in clinical practice. METHODS: VEGF was intravenously injected to imitate the evaluated expression after local tumor therapy, such as TACE. A total of 40 SD rats bearing hepatic tumors were randomly divided into four groups and sorafenib was administered at different timepoints: (A) control group: VEGF injection only; (B) initiating sorafenib 72 h prior to VEGF injection; (C) initiating sorafenib simultaneously with VEGF injection; (D) initiating sorafenib 72 h post-VEGF injection. The rate of tumor growth, median survival time, expression of VEGF, and microvessel density (MVD), as determined by immunohistochemical (IHC) examination, were compared. RESULTS: The results revealed that the tumor size and median survival time were significantly different between the three sorafenib groups compared to the control group (p < 0.05). Median survival times were 19.6 ± 1.78, 31.2 ± 6.99, 27.4 ± 4.9, and 26.5 ± 4.6 days in group A, B, C, and D, respectively. Furthermore, there was a difference in statistical significance between the two sorafenib groups B and D (p = 0.04). Tumors were collected for HE staining and IHC examination. The expression levels of VEGF in B, C, and D were 42.8 ± 7.96, 71.9 ± 15.73, and 73.6 ± 13.73, and all of them were significantly lower than that in the control group (88.3 ± 13.61). Furthermore, the level of MVD was 109.2 ± 8.98 in the control group, which was significantly higher than in the three sorafenib groups (45.7 ± 16.92, 77.1 ± 16.29, and 93.6 ± 12.87, all p < 0.05). CONCLUSIONS: According to our results, the most suitable regimen for the administration of sorafenib is before the increased expression of VEGF, which showed a potential advantage for controlling the tumor growth and prolonging the survival time of test animal via inhibiting VEGF-receptor expression through the bifunction of VEGF, and the reduction of tumor angiogenesis.


Assuntos
Antineoplásicos/administração & dosagem , Carcinogênese/efeitos dos fármacos , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Neoplasias Hepáticas Experimentais/metabolismo , Sorafenibe/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Esquema de Medicação , Xenoenxertos , Imuno-Histoquímica , Neoplasias Hepáticas Experimentais/irrigação sanguínea , Neoplasias Hepáticas Experimentais/patologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Microvasos/crescimento & desenvolvimento , Ratos , Ratos Sprague-Dawley , Sorafenibe/uso terapêutico , Análise de Sobrevida , Fatores de Tempo , Regulação para Cima
15.
J Tissue Eng Regen Med ; 12(3): e1325-e1336, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28714140

RESUMO

Self-sustainability after implantation is one of the critical obstacles facing large engineered tissues. A preformed functional vascular network provides an effective solution for solving the mass transportation problem. With the support of mural cells, endothelial cells (ECs) can form microvessels within engineered tissues. As an important mural cell, human mesenchymal stem cells (hMSCs) not only stabilize the engineered microvessel network, but also preserve their multi-potency when grown under optimal culture conditions. A prevascularized hMSC/extracellular matrix (ECM) sheet fabricated by the combination of hMSCs, ECs and a naturally derived nanofibrous ECM scaffold offers great opportunity for engineering mechanically strong and completely biological three-dimensional prevascularized tissues. The objective of this study was to create a prevascularized hMSC/ECM sheet by co-culturing ECs and hMSCs on a nanofibrous ECM scaffold. Physiologically low oxygen (2% O2 ) was introduced during the 7 day hMSC culture to preserve the stemness of hMSCs and thereby their capability to secrete angiogenic factors. The ECs were then included to form microvessels under normal oxygen (20% O2 ) for up to 7 days. The results showed that a branched and mature vascular network was formed in the co-culture condition. Angiogenic factors vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF) and angiopoietin-1 (Ang-1) were significantly increased by low-oxygen culture of hMSCs, which further stabilized and supported the maturation of microvessels. A differentiation assay of the prevascularized ECM scaffold demonstrated a retained hMSC multi-potency in the hypoxia cultured samples. The prevascularized hMSC/ECM sheet holds great promise for engineering three-dimensional prevascularized tissues for diverse applications.


Assuntos
Matriz Extracelular/metabolismo , Nanofibras/química , Neovascularização Fisiológica , Engenharia Tecidual/métodos , Indutores da Angiogênese/metabolismo , Diferenciação Celular , Hipóxia Celular , Linhagem da Célula , Proliferação de Células , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Microvasos/crescimento & desenvolvimento , Osteogênese , Permeabilidade
16.
Int J Mol Sci ; 18(5)2017 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-28467355

RESUMO

The formation of brain vasculature is an essential step during central nervous system development. The molecular mechanism underlying brain angiogenesis remains incompletely understood. The role of Atg7, an autophagy-related protein, in brain angiogenesis was investigated in this study. We found that the microvessel density in mice brains with endothelial-specific knockout of Atg7 (Atg7 EKO) was significantly decreased compared to wild-type control. Consistently, in vitro angiogenesis assays showed that Atg7 knockdown impaired angiogenesis in brain microvascular endothelial cells. Further results indicated that knockdown of Atg7 reduced interleukin-6 (IL-6) expression in brain microvascular endothelial cells, which is mediated by NF-κB-dependent transcriptional control. Interestingly, exogenous IL-6 restored the impaired angiogenesis and reduced cell motility caused by Atg7 knockdown. These results demonstrated that Atg7 has proangiogenic activity in brain angiogenesis which is mediated by IL-6 production in a NF-κB-dependent manner.


Assuntos
Proteína 7 Relacionada à Autofagia/metabolismo , Encéfalo/irrigação sanguínea , Interleucina-6/metabolismo , NF-kappa B/metabolismo , Neovascularização Fisiológica/fisiologia , Análise de Variância , Animais , Proteína 7 Relacionada à Autofagia/genética , Movimento Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais , Humanos , Camundongos , Camundongos Knockout , Microvasos/crescimento & desenvolvimento , Microvasos/metabolismo , Neovascularização Fisiológica/genética
17.
Int J Oncol ; 50(6): 2136-2144, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28498425

RESUMO

The significance of indoleamine 2,3-dioxygenase-1 (IDO1) has been studied in various types of tumors, but the relationship between IDO1 and tumor angiogenesis needs further delineation. We aimed to clarify the relationship between tumor angiogenesis and IDO1 expression, and to explore the possibility of IDO1-targeting molecular therapy for lung cancer. For the first time, we found that silencing the IDO1 gene using small interfering RNA (siRNA) inhibits in vitro cancer cell invasion and migration. We further demonstrated that knockdown of IDO1 decreased the formation of vasculogenic mimicry. In addition to these in vitro findings, we also demonstrated that in vivo IDO1 gene silencing using short hairpin RNA (shRNA) delayed tumor onset and inhibited tumor growth in the mouse model. Immunostaining showed that IDO1 gene silencing inhibited tumor angiogenesis. Moreover, the expression of IDO1 was associated with microvessel density (MVD) labeled by CD34 and CD146. These findings indicate that IDO1 has the potential to participate in or contribute to the formation of new capillaries, supporting the applicability of IDO1-targeting molecular therapy in lung cancer.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase/genética , Terapia de Alvo Molecular , Neovascularização Patológica/genética , Neovascularização Patológica/terapia , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Inativação Gênica , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Camundongos , Microvasos/crescimento & desenvolvimento , Microvasos/patologia , Invasividade Neoplásica/genética , Neovascularização Patológica/patologia , RNA Interferente Pequeno/genética , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Sci Rep ; 7: 44687, 2017 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-28317868

RESUMO

We report a pivotal role for IL-5 as an angiogenic activator. IL-5 increased proliferation, migration and colony tube formation in HUVECs associated with the phosphorylation of ERK and AKT/eNOS, and promoted microvessel sprouting from an angiogenesis animal model. The angiogenic effects were confirmed in IL-5-deficient mice and addition of IL-5 antibody. HSP70-1 was identified via expression profiling following IL-5 stimulation. A siRNA knockdown of HSP70-1 suppressed angiogenic responses and eNOS phosphorylation induced by IL-5. HSP70-1 overexpression enhanced IL-5-induced angiogenic responses. In addition, IL-5-induced neo-vascular formation was verified in both HSP70-1 knockout and HSP70-1 transgenic mice. Furthermore, transcription factor AP-1 was a main factor in IL-5-induced HSP70-1 in response to ERK and AKT signaling pathway. Angiogenic responses induced by VEGF had no effect in either HSP70-1 siRNA in vitro or HSP70-1 knockout mice. IL-5-induced angiogenic responses depended on the binding of IL-5Rα. Our data demonstrate that binding of IL-5 to IL-5Rα receptors enhances angiogenic responses by stimulating the expression of HSP70-1 via the eNOS signaling pathway.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Interleucina-5/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/metabolismo , Transdução de Sinais , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Perfilação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Interleucina-5/deficiência , Interleucina-5/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microvasos/efeitos dos fármacos , Microvasos/crescimento & desenvolvimento , Neovascularização Fisiológica/genética , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Cicatrização/efeitos dos fármacos
19.
Stem Cell Res Ther ; 8(1): 52, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28279225

RESUMO

BACKGROUND: Myocardial microvascular loss after myocardial infarction (MI) remains a therapeutic challenge. Autologous stem cell therapy was considered as an alternative; however, it has shown modest benefits due to the impairing effects of cardiovascular risk factors on stem cells. Allogenic adipose-derived stem cells (ASCs) may overcome such limitations, and because of their low immunogenicity and paracrine potential may be good candidates for cell therapy. In the present study we investigated the effects of allogenic ASCs and their released products on cardiac rarefaction post MI. METHODS: Pig subcutaneous adipose tissue ASCs were isolated, expanded and GFP-labeled. ASC angiogenic function was assessed by the in-vivo chick chorioallantoic membrane (CAM) model. Pigs underwent MI induction and 7 days after were randomized to receive: allogenic ASCs (intracoronary infusion); conditioned media (CM; intravenous infusion); ASCs + CM; or PBS/placebo (control). Cardiac damage and function were monitored by 3-T cardiac magnetic resonance imaging upon infusion (baseline CMR) and 1 and 3 weeks thereafter. We assessed in the myocardium: microvessel density; angiogenic markers (CD105, CD31, TF, VEGFR2, VEGFR1, vWF, eNOS, CD62); collagen deposition; and reparative fibrosis (TGFß/TßRII/collagen). Differential proteomics of ASCs and CM was performed to characterize the ASC protein signature. RESULTS: CAM indicated a significant ASC proangiogenic capacity. In pigs after MI, only PBS/placebo animals displayed an impaired cardiac function 3 weeks after infusion (p < 0.05 vs baseline). Administration of ASCs + CM significantly enhanced neovessel formation and favored cardiac repair post MI (p < 0.05 vs the other groups). Molecular markers of angiogenesis were significantly upregulated both at transcriptional and protein levels (p < 0.05). The in-silico bioinformatics analysis of the ASC and CM proteome (interactome) indicated activation of a coordinated protein network involved in the formation of microvessels and the resolution of rarefaction. CONCLUSION: Coadministration of allogenic ASCs and their CM synergistically contribute to the neovascularization of the infarcted myocardium through a coordinated upregulation of the proangiogenic protein interactome.


Assuntos
Infarto do Miocárdio/terapia , Isquemia Miocárdica/terapia , Transplante de Células-Tronco , Transplante Autólogo , Tecido Adiposo/citologia , Animais , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Microvasos/crescimento & desenvolvimento , Microvasos/fisiopatologia , Infarto do Miocárdio/fisiopatologia , Isquemia Miocárdica/fisiopatologia , Miocárdio/patologia , Fatores de Risco , Suínos , Biologia de Sistemas
20.
Sci Rep ; 7: 39848, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-28045067

RESUMO

Inflammaging is defined as low-grade, chronic, systemic inflammation in aging, in the absence of overt infection. Age-associated deterioration of gastrointestinal function could be ascribed to the inflammaging, although evidence is yet to emerge. Here we show that microvessels in aging mouse intestine were progressively deprived of supportive structures, microvessel-associated pericytes and adherens junction protein vascular endothelial (VE)-cadherin, and became leaky. This alteration was ascribed to up-regulation of angiopoetin-2 in microvascular endothelial cells. Up-regulation of the angiopoietin-2 was by TNF-α, originated from M2-like residential CD206+ macrophages, proportion of which increases as animal ages. It was concluded that antigenic burdens encountered in intestine throughout life create the condition of chronic stage of inflammation, which accumulates M2-like macrophages expressing TNF-α. The TNF-α induces vascular leakage to facilitate recruitment of immune cells into intestine under the chronic inflammatory setting.


Assuntos
Envelhecimento/patologia , Mucosa Intestinal/metabolismo , Microvasos/metabolismo , Remodelação Vascular , Junções Aderentes/metabolismo , Angiopoietina-2/metabolismo , Animais , Caderinas/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Intestinos/citologia , Intestinos/crescimento & desenvolvimento , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/citologia , Microvasos/crescimento & desenvolvimento , Pericitos/metabolismo , Receptores de Superfície Celular/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA