Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Microcirculation ; 31(4): e12852, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38619428

RESUMO

The microvasculature is integral to nearly every tissue in the body, providing not only perfusion to and from the tissue, but also homing sites for immune cells, cellular niches for tissue dynamics, and cooperative interactions with other tissue elements. As a microtissue itself, the microvasculature is a composite of multiple cell types exquisitely organized into structures (individual vessel segments and extensive vessel networks) capable of considerable dynamics and plasticity. Consequently, it has been challenging to include a functional microvasculature in assembled or fabricated tissues. Isolated fragments of intact microvessels, which retain the cellular composition and structures of native microvessels, are proving effective in a variety of vascularization applications including tissue in vitro disease modeling, vascular biology, mechanistic discovery, and tissue prevascularization in regenerative therapeutics and grafting. In this review, we will discuss the importance of recapitulating native tissue biology and the successful vascularization applications of isolated microvessels.


Assuntos
Microvasos , Neovascularização Fisiológica , Humanos , Microvasos/fisiologia , Animais
2.
J Surg Res ; 295: 28-40, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37979234

RESUMO

INTRODUCTION: Graft loss in vascularized composite allotransplantation (VCA) is more often associated with vasculopathy and chronic rejection (CR) than acute cellular rejection (ACR). We present a rat osteomyocutaneous flap model using titrated tacrolimus administration that mimics the graft rejection patterns in our clinical hand transplant program. Comparison of outcomes in these models support a role for ischemia reperfusion injury (IRI) and microvascular changes in CR of skin and large-vessel vasculopathy. The potential of the surgical models for investigating mechanisms of rejection and vasculopathy in VCA and treatment interventions is presented. MATERIALS AND METHODS: Four rodent groups were evaluated: syngeneic controls (Group 1), allogeneic transient immunosuppression (Group 2), allogeneic suboptimal immunosuppression (Group 3), and allogeneic standard immunosuppression (Group 4). Animals were monitored for ACR, vasculopathy, and CR of the skin. RESULTS: Transient immunosuppression resulted in severe ACR within 2 wk of tacrolimus discontinuation. Standard immunosuppression resulted in minimal rejection but subclinical microvascular changes, including capillary thrombosis and luminal narrowing in arterioles in the donor skin. Further reduction in tacrolimus dose led to femoral vasculopathy and CR of the skin. Surprisingly, femoral vasculopathy was also observed in the syngeneic control group. CONCLUSIONS: Titration of tacrolimus in the allogeneic VCA model resulted in presentations of rejection and vasculopathy similar to those in patients and suggests vasculopathy starts at the microvascular level. This adjustable experimental model will allow the study of variables and interventions, such as external trauma or complement blockade, that may initiate or mitigate vasculopathy and CR in VCA.


Assuntos
Tacrolimo , Alotransplante de Tecidos Compostos Vascularizados , Humanos , Ratos , Animais , Alotransplante de Tecidos Compostos Vascularizados/efeitos adversos , Alotransplante de Tecidos Compostos Vascularizados/métodos , Retalhos Cirúrgicos , Terapia de Imunossupressão , Tolerância Imunológica , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/etiologia , Sobrevivência de Enxerto
3.
Ann Biomed Eng ; 51(8): 1835-1846, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37149511

RESUMO

The formation of new vascular networks via angiogenesis is a crucial biological mechanism to balance tissue metabolic needs, yet the coordination of factors that influence the guidance of growing neovessels remain unclear. This study investigated the influence of extracellular cues within the immediate environment of sprouting tips over multiple hours and obtained quantitative relationships describing their effects on the growth trajectories of angiogenic neovessels. Three distinct microenvironmental cues-fibril tracks, ECM density, and the presence of nearby cell bodies-were extracted from 3D time series image data. The prominence of each cue was quantified along potential sprout trajectories to predict the response to multiple microenvironmental factors simultaneously. Sprout trajectories significantly correlated with the identified microenvironmental cues. Specifically, ECM density and nearby cellular bodies were the strongest predictors of the trajectories taken by neovessels (p < 0.001 and p = 0.016). Notwithstanding, direction changing trajectories, deviating from the initial neovessel orientation, were significantly correlated with fibril tracks (p = 0.003). Direction changes also occurred more frequently with strong microenvironmental cues. This provides evidence for the first time that local matrix fibril alignment influences changes in sprout trajectories but does not materially contribute to persistent sprouting. Together, our results suggest the microenvironmental cues significantly contribute to guidance of sprouting trajectories. Further, the presented methods quantitatively distinguish the influence of individual microenvironmental stimuli during guidance.


Assuntos
Sinais (Psicologia) , Neovascularização Fisiológica , Morfogênese , Fenômenos Fisiológicos Cardiovasculares , Imageamento Tridimensional , Matriz Extracelular/fisiologia
4.
Bioengineering (Basel) ; 10(2)2023 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-36829618

RESUMO

Tissue organoids hold enormous potential as tools for a variety of applications, including disease modeling and drug screening. To effectively mimic the native tissue environment, it is critical to integrate a microvasculature with the parenchyma and stroma. In addition to providing a means to physiologically perfuse the organoids, the microvasculature also contributes to the cellular dynamics of the tissue model via the cells of the perivascular niche, thereby further modulating tissue function. In this review, we discuss current and developing strategies for vascularizing organoids, consider tissue-specific vascularization approaches, discuss the importance of perfusion, and provide perspectives on the state of the field.

5.
SLAS Discov ; 27(6): 358-368, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35772696

RESUMO

Advanced in vitro tissue models better reflect healthy and disease tissue conditions in the body. However, complex tissue models are often manufactured using custom solutions and can be challenging to manufacture to scale. Here, we describe the automated fabrication of a cell-dense, thick (≤ 1 cm), human vascularized liver tissue model using a robotic biomanufacturing platform and off-the-shelf components to build, culture, and sample liver tissues hands-free without compromising tissue health or function. Fabrication of the tissue involved 3D bioprinting and incorporation of primary human hepatocytes, primary human non-parenchymal cells, and isolated fragments of intact human microvessels as vascular precursors. No differences were observed in select assessments of the liver tissues fabricated by hand or via automation. Furthermore, constant media exchange, via perfusion, improved urea output and elevated tissue metabolism. Interestingly, inclusion of adipose-derived human microvessels enhanced functional gene expression, including an enhanced response to a drug challenge. Our results describe the fabrication of a thick liver tissue environment useful for a variety of applications including liver disease modeling, infectious agent studies, and cancer investigations. We expect the automated fabrication of the vascularized liver tissue, at the point of use and using off-the-shelf platforms, eases fabrication of the complex model and increases its utility.


Assuntos
Bioimpressão , Engenharia Tecidual , Bioimpressão/métodos , Hepatócitos/metabolismo , Humanos , Fígado , Microvasos , Engenharia Tecidual/métodos
6.
Mamm Genome ; 33(3): 437-450, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35333952

RESUMO

Tissue organoids or "mini organs" can be invaluable tools for understanding health and disease biology, modeling tissue dynamics, or screening potential drug candidates. Effective vascularization of these models is critical for truly representing the in vivo tissue environment. Not only is the formation of a vascular network, and ultimately a microcirculation, essential for proper distribution and exchange of oxygen and nutrients throughout larger organoids, but vascular cells dynamically communicate with other cells to modulate overall tissue behavior. Additionally, interstitial fluid flow, mediated by a perfused microvasculature, can have profound influences on tissue biology. Thus, a truly functionally and biologically relevant organoid requires a vasculature. Here, we review existing strategies for fabricating and incorporating vascular elements and perfusion within tissue organoids.


Assuntos
Organoides , Oxigênio , Perfusão
7.
Am J Physiol Heart Circ Physiol ; 322(5): H806-H818, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35333118

RESUMO

Angiogenesis is necessary for wound healing, tumorigenesis, implant inosculation, and homeostasis. In each situation, matrix structure and mechanics play a role in determining whether new vasculatures can establish transport to new or hypoxic tissues. Neovessel growth and directional guidance are sensitive to three-dimensional (3-D) matrix anisotropy and density, although the individual and integrated roles of these matrix features have not been fully recapitulated in vitro. We developed a tension-based method to align 3-D collagen constructs seeded with microvessel fragments in matrices of three levels of collagen fibril anisotropy and two levels of collagen density. The extent and direction of neovessel growth from the parent microvessel fragments increased with matrix anisotropy and decreased with density. The proangiogenic effects of anisotropy were attenuated at higher matrix densities. We also examined the impact of matrix anisotropy in an experimental model of neovessel invasion across a tissue interface. Matrix density was found to dictate the success of interface crossing, whereas interface curvature and fibril alignment were found to control directional guidance. Our findings indicate that complex configurations of matrix density and alignment can facilitate or complicate the establishment or maintenance of vascular networks in pathological and homeostatic angiogenesis. Furthermore, we extend preexisting methods for tuning collagen anisotropy in thick constructs. This approach addresses gaps in tissue engineering and cell culture by supporting the inclusion of large multicellular structures in prealigned constructs.NEW & NOTEWORTHY Matrix anisotropy and density have a considerable effect on angiogenic vessel growth and directional guidance. However, the current literature relies on 2-D and simplified models of angiogenesis (e.g., tubulogenesis and vasculogenesis). We present a method to align 3-D collagen scaffolds embedded with microvessel fragments to different levels of anisotropy. Neovessel growth increases with anisotropy and decreases with density, which may guide angiogenic neovessels across tissue interfaces such as during implant inosculation and tumorigenesis.


Assuntos
Colágeno , Neovascularização Fisiológica , Anisotropia , Carcinogênese , Matriz Extracelular/química , Humanos , Morfogênese , Neovascularização Patológica
8.
Front Bioeng Biotechnol ; 10: 838415, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35356783

RESUMO

Critical-sized defects of irregular bones requiring bone grafting, such as in craniofacial reconstruction, are particularly challenging to repair. With bone-grafting procedures growing in number annually, there is a reciprocal growing interest in bone graft substitutes to meet the demand. Autogenous osteo(myo)cutaneous grafts harvested from a secondary surgical site are the gold standard for reconstruction but are associated with donor-site morbidity and are in limited supply. We developed a bone graft strategy for irregular bone-involved reconstruction that is customizable to defect geometry and patient anatomy, is free of synthetic materials, is cellularized, and has an outer pre-vascularized tissue layer to enhance engraftment and promote osteogenesis. The graft, comprised of bioprinted human-derived demineralized bone matrix blended with native matrix proteins containing human mesenchymal stromal cells and encased in a simple tissue shell containing isolated, human adipose microvessels, ossifies when implanted in rats. Ossification follows robust vascularization within and around the graft, including the formation of a vascular leash, and develops mechanical strength. These results demonstrate an early feasibility animal study of a biofabrication strategy to manufacture a 3D printed patient-matched, osteoconductive, tissue-banked, bone graft without synthetic materials for use in craniofacial reconstruction. The bone fabrication workflow is designed to be performed within the hospital near the Point of Care.

9.
Am J Physiol Heart Circ Physiol ; 322(4): H647-H680, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35179976

RESUMO

Pathologies of the vasculature including the microvasculature are often complex in nature, leading to loss of physiological homeostatic regulation of patency and adequate perfusion to match tissue metabolic demands. Microvascular dysfunction is a key underlying element in the majority of pathologies of failing organs and tissues. Contributing pathological factors to this dysfunction include oxidative stress, mitochondrial dysfunction, endoplasmic reticular (ER) stress, endothelial dysfunction, loss of angiogenic potential and vascular density, and greater senescence and apoptosis. In many clinical settings, current pharmacologic strategies use a single or narrow targeted approach to address symptoms of pathology rather than a comprehensive and multifaceted approach to address their root cause. To address this, efforts have been heavily focused on cellular therapies and cell-free therapies (e.g., exosomes) that can tackle the multifaceted etiology of vascular and microvascular dysfunction. In this review, we discuss 1) the state of the field in terms of common therapeutic cell population isolation techniques, their unique characteristics, and their advantages and disadvantages, 2) common molecular mechanisms of cell therapies to restore vascularization and/or vascular function, 3) arguments for and against allogeneic versus autologous applications of cell therapies, 4) emerging strategies to optimize and enhance cell therapies through priming and preconditioning, and, finally, 5) emerging strategies to bolster therapeutic effect. Relevant and recent clinical and animal studies using cellular therapies to restore vascular function or pathologic tissue health by way of improved vascularization are highlighted throughout these sections.


Assuntos
Microvasos , Doenças Vasculares , Animais , Endotélio Vascular/metabolismo , Estresse Oxidativo , Regeneração , Doenças Vasculares/metabolismo
10.
Methods Mol Biol ; 2441: 311-320, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35099747

RESUMO

Angiogenesis, the formation of new vessel elements from existing vessels, is important in homeostasis and tissue repair. Dysfunctional angiogenesis can contribute to numerous pathologies, including cancer, ischemia, and chronic wounds. In many instances, growing vessels must navigate along or across tissue-associated boundaries and interfaces tissue interfaces. To understand this dynamic, we developed a new model for studying angiogenesis at tissue interfaces utilizing intact microvessel fragments isolated from adipose tissue. Isolated microvessels retain their native structural and cellular complexity. When embedded in a 3D matrix, microvessels, sprout, grow, and connect to form a neovasculature. Here, we discuss and describe methodology for one application of our microvessel-based angiogenesis model, studying neovessel behavior at tissue interfaces.


Assuntos
Microvasos , Neovascularização Fisiológica , Tecido Adiposo , Humanos , Morfogênese , Neovascularização Patológica
11.
Front Physiol ; 12: 650714, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33986691

RESUMO

Given the considerable research efforts in understanding and manipulating the vasculature in tissue health and function, making effective measurements of vascular density is critical for a variety of biomedical applications. However, because the vasculature is a heterogeneous collection of vessel segments, arranged in a complex three-dimensional architecture, which is dynamic in form and function, it is difficult to effectively measure. Here, we developed a semi-automated method that leverages machine learning to identify and quantify vascular metrics in an angiogenesis model imaged with different modalities. This software, BioSegment, is designed to make high throughput vascular density measurements of fluorescent or phase contrast images. Furthermore, the rapidity of assessments makes it an ideal tool for incorporation in tissue manufacturing workflows, where engineered tissue constructs may require frequent monitoring, to ensure that vascular growth benchmarks are met.

12.
Biofabrication ; 13(3)2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33513595

RESUMO

Tissue organoids are proving valuable for modeling tissue health and disease in a variety of applications. This is due, in part, to the dynamic cell-cell interactions fostered within the 3D tissue-like space. To this end, the more that organoids recapitulate the different cell-cell interactions found in native tissue, such as that between parenchyma and the microvasculature, the better the fidelity of the model. The microvasculature, which is comprised of a spectrum of cell types, provides not only perfusion in its support of tissue health, but also important cellular interactions and biochemical dynamics important in tissue phenotype and function. Here, we incorporate whole, intact human microvessel fragments isolated from adipose tissue into organoids to form both mesenchymal stem cell (MSC) and adipocyte vascularized organoids. Isolated microvessels retain their native structure and cell composition, providing a more complete representation of the microvasculature within the organoids. Microvessels expanded via sprouting angiogenesis within organoids comprised of either MSCs or MSC-derived adipocytes grew out of the organoids when placed in a 3D collagen matrix. In MSC organoids, a ratio of 50 MSCs to 1 microvessel fragment created the optimal vascularization response. We developed a new differentiation protocol that enabled the differentiation of MSCs into adipocytes while simultaneously promoting microvessel angiogenesis. The adipocyte organoids contained vascular networks, were responsive in a lipolysis assay, and expressed the functional adipocyte markers adiponectin and peroxisome proliferator-activated receptor gamma. The presence of microvessels promoted insulin receptor expression by adipocytes and modified interleukin-6 secretion following a tumor necrosis factor alpha challenge. Overall, we demonstrate a robust method for vascularizing high cell-density organoids with potential implications for other tissues as well.


Assuntos
Células-Tronco Mesenquimais , Organoides , Adipócitos , Tecido Adiposo , Humanos , Microvasos
13.
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.

14.
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
16.
Cell Mol Bioeng ; 12(5): 443-454, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31719926

RESUMO

INTRODUCTION: The biggest bottleneck for cell-based regenerative therapy is the lack of a functional vasculature to support the grafts. This problem is exacerbated in diabetic patients, where vessel growth is inhibited. To address this issue, we aim to identify the causes of poor vascularization in 3D engineered tissues in diabetes and to reverse its negative effects. METHODS: We used 3D vascularized constructs composed of microvessel fragments containing all cells present in the microcirculation, embedded in collagen type I hydrogels. Constructs were either cultured in vitro or implanted subcutaneously in non-diabetic or in a type I diabetic (streptozotocin-injected) mouse model. We used qPCR, ELISA, immunostaining, FACs and co-culture assays to characterize the effect of diabetes in engineered constructs. RESULTS: We demonstrated in 3D vascularized constructs that perivascular cells secrete hepatocyte growth factor (HGF), driving microvessel sprouting. Blockage of HGF or HGF receptor signaling in 3D constructs prevented vessel sprouting. Moreover, HGF expression in 3D constructs in vivo is downregulated in diabetes; while no differences were found in HGF receptor, VEGF or VEGF receptor expression. Low HGF expression in diabetes delayed the inosculation of graft and host vessels, decreasing blood perfusion and preventing tissue engraftment. Supplementation of HGF in 3D constructs, restored vessel sprouting in a diabetic milieu. CONCLUSION: We show for the first time that diabetes affects HGF secretion in microvessels, which in turn prevents the engraftment of engineered tissues. Exogenous supplementation of HGF, restores angiogenic growth in 3D constructs showing promise for application in cell-based regenerative therapies.

17.
Front Physiol ; 10: 1011, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31507428

RESUMO

Thorough understanding of growth and evolution of tissue vasculature is fundamental to many fields of medicine including cancer therapy, wound healing, and tissue engineering. Angiogenesis, the growth of new vessels from existing ones, is dynamically influenced by a variety of environmental factors, including mechanical and biophysical factors, chemotactic factors, proteolysis, and interaction with stromal cells. Yet, dynamic interactions between neovessels and their environment are difficult to study with traditional fixed time imaging techniques. Advancements in imaging technologies permit time-series and volumetric imaging, affording the ability to visualize microvessel growth over 3D space and time. Time-lapse imaging has led to more informative investigations of angiogenesis. The environmental factors implicated in angiogenesis span a wide range of signals. Neovessels advance through stromal matrices by forming attachments and pulling and pushing on their microenvironment, reorganizing matrix fibers, and inducing large deformations of the surrounding stroma. Concurrently, neovessels secrete proteolytic enzymes to degrade their basement membrane, create space for new vessels to grow, and release matrix-bound cytokines. Growing neovessels also respond to a host of soluble and matrix-bound growth factors, and display preferential growth along a cytokine gradient. Lastly, stromal cells such as macrophages and mesenchymal stem cells (MSCs) interact directly with neovessels and their surrounding matrix to facilitate sprouting, vessel fusion, and tissue remodeling. This review highlights how time-lapse imaging techniques advanced our understanding of the interaction of blood vessels with their environment during sprouting angiogenesis. The technology provides means to characterize the evolution of microvessel behavior, providing new insights and holding great promise for further research on the process of angiogenesis.

18.
J Vis Exp ; (146)2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-31081823

RESUMO

Vascularized composite allotransplantation (VCA) is a relatively new field in the reconstructive surgery. Clinical achievements in human VCA include hand and face transplants and, more recently, abdominal wall, uterus, and urogenital transplants. Functional outcomes have exceeded initial expectations, and most recipients enjoy an improved quality of life. However, as clinical experience accumulates, chronic rejection and complications from the immunosuppression must be addressed. In many cases where grafts have failed, the causative pathology has been ischemic vasculopathy. The biological mechanisms of the acute and chronic rejection associated with VCA, especially ischemic vasculopathy, are important areas of research. However, due to the very small number of VCA patients, the evaluation of proposed mechanisms is better addressed in an experimental model. Multiple groups have used animal models to address some of the relevant unsolved questions in VCA rejection and vasculopathy. Several model designs involving a variety of species are described in the literature. Here we present a reproducible model of VCA heterotopic hindlimb osteomyocutaneous flap in the rat that can be utilized for translational VCA research. This model allows for the serial evaluation of the graft, including biopsies and different imaging modalities, while maintaining a low level of morbidity.


Assuntos
Coristoma/cirurgia , Membro Posterior/cirurgia , Retalhos Cirúrgicos , Pesquisa Translacional Biomédica , Animais , Membro Posterior/transplante , Modelos Animais , Ratos , Transplante Homólogo , Alotransplante de Tecidos Compostos Vascularizados/métodos
19.
PLoS One ; 13(8): e0202934, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30142193

RESUMO

An early manifestation of coronary artery disease in advanced age is the development of microvascular dysfunction leading to deficits in diastolic function. Our lab has previously shown that epicardial treatment with adipose-derived stromal vascular fraction (SVF) preserves microvascular function following coronary ischemia in a young rodent model. Follow-up studies showed intravenous (i.v.) delivery of SVF allows the cells to migrate to the walls of small vessels and reset vasomotor tone. Therefore we tested the hypothesis that the i.v. cell injection of SVF would reverse the coronary microvascular dysfunction associated with aging in a rodent model. Fischer 344 rats were divided into 4 groups: young control (YC), old control (OC), old + rat aortic endothelial cells (O+EC) and old + GFP+ SVF cells (O+SVF). After four weeks, cardiac function and coronary flow reserve (CFR) were measured via echocardiography, and hearts were explanted either for histology or isolation of coronary arterioles for vessel reactivity studies. In a subgroup of animals, microspheres were injected during resting and dobutamine-stimulated conditions to measure coronary blood flow. GFP+ SVF cells engrafted and persisted in the myocardium and coronary vasculature four weeks following i.v. injection. Echocardiography showed age-related diastolic dysfunction without accompanying systolic dysfunction; diastolic function was improved in old rats after SVF treatment. Ultrasound and microsphere data both showed increased stimulated coronary blood flow in O+SVF rats compared to OC and O+EC, while isolated vessel reactivity was mostly unchanged. I.v.-injected SVF cells were capable of incorporating into the vasculature of the aging heart and are shown in this study to improve CFR and diastolic function in a model of advanced age. Importantly, SVF injection did not lead to arrhythmias or increased mortality in aged rats. SVF cells provide an autologous cell therapy option for treatment of microvascular and cardiac dysfunction in aged populations.


Assuntos
Adipócitos/citologia , Arteríolas/fisiologia , Circulação Coronária , Células Estromais/citologia , Função Ventricular Esquerda , Acetilcolina/farmacologia , Animais , Arteríolas/efeitos dos fármacos , Circulação Coronária/efeitos dos fármacos , Feminino , Nitroprussiato/farmacologia , Ratos , Função Ventricular Esquerda/efeitos dos fármacos
20.
Hepatol Commun ; 1(9): 886-898, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29130076

RESUMO

Familial hypercholesterolemia (FH) is a hereditary disease primarily due to mutations in the low-density lipoprotein receptor (LDLR) that lead to elevated cholesterol and premature development of cardiovascular disease. Homozygous FH patients (HoFH) with two dysfunctional LDLR alleles are not as successfully treated with standard hypercholesterol therapies, and more aggressive therapeutic approaches to control cholesterol levels must be considered. Liver transplant can resolve HoFH, and hepatocyte transplantation has shown promising results in animals and humans. However, demand for donated livers and high-quality hepatocytes overwhelm the supply. Human pluripotent stem cells can differentiate to hepatocyte-like cells (HLCs) with the potential for experimental and clinical use. To be of future clinical use as autologous cells, LDLR genetic mutations in derived FH-HLCs need to be corrected. Genome editing technology clustered-regularly-interspaced-short-palindromic-repeats/CRISPR-associated 9 (CRISPR/Cas9) can repair pathologic genetic mutations in human induced pluripotent stem cells. CONCLUSION: We used CRISPR/Cas9 genome editing to permanently correct a 3-base pair homozygous deletion in LDLR exon 4 of patient-derived HoFH induced pluripotent stem cells. The genetic correction restored LDLR-mediated endocytosis in FH-HLCs and demonstrates the proof-of-principle that CRISPR-mediated genetic modification can be successfully used to normalize HoFH cholesterol metabolism deficiency at the cellular level.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...