RESUMO
Arterioles are key determinants of the total peripheral vascular resistance, which, in turn, is a key determinant of arterial blood pressure. However, the amount of protein available from one isolated human arteriole may be less than 5 µg, making proteomic analysis challenging. In addition, obtaining human arterioles requires manual dissection of unfrozen clinical specimens. This limits its feasibility, especially for powerful multicenter clinical studies in which clinical specimens need to be shipped overnight to a research laboratory for arteriole isolation. We performed a study to address low-input, test overnight tissue storage and develop a reference human arteriolar proteomic profile. In tandem mass tag proteomics, use of a booster channel consisting of human induced pluripotent stem cell-derived endothelial and vascular smooth muscle cells (1:5 ratio) increased the number of proteins detected in a human arteriole segment with a false discovery rate of <0.01 from 1051 to more than 3000. The correlation coefficient of proteomic profile was similar between replicate arterioles isolated freshly, following cold storage, or before and after the cold storage (1-way analysis of variance; P = .60). We built a human arteriolar proteomic profile consisting of 3832 proteins based on the analysis of 12 arteriole samples from 3 subjects. Of 1945 blood pressure-relevant proteins that we curated, 476 (12.5%) were detected in the arteriolar proteome, which was a significant overrepresentation (χ2 test; P < .05). These findings demonstrate that proteomic analysis is feasible with arterioles isolated from human adipose tissue following cold overnight storage and provide a reference human arteriolar proteome profile highly valuable for studies of arteriole-related traits.
Assuntos
Tecido Adiposo , Proteômica , Humanos , Arteríolas/metabolismo , Proteômica/métodos , Tecido Adiposo/metabolismo , Tecido Adiposo/irrigação sanguínea , Proteoma/metabolismo , Proteoma/análise , Feminino , Masculino , Adulto , Pessoa de Meia-IdadeRESUMO
Pregnancy is characterized by longitudinal maternal, physiological adaptations to support the development of a fetus. One of the cardinal maternal adaptations during a healthy pregnancy is a progressive increase in uterine artery blood flow. This facilitates sufficient blood supply for the development of the placenta and the growing fetus. Regional hemodynamic changes in the uterine circulation, such as a vast reduction in uterine artery resistance, are mainly facilitated by changes in uterine artery reactivity and myogenic tone along with remodeling of the uterine arteries. These regional changes in vascular reactivity have been attributed to pregnancy-induced adaptations of cell-to-cell communication mechanisms, with an emphasis on the interaction between endothelial and vascular smooth muscle cells. Perivascular adipose tissue (PVAT) is considered the fourth layer of the vascular wall and contributes to the regulation of vascular reactivity in most vascular beds and most species. This review focuses on mechanisms of uterine artery reactivity and the role of PVAT in pregnancy-induced maternal vascular adaptations, with an emphasis on the uterine circulation.
Assuntos
Adaptação Fisiológica , Tecido Adiposo , Artéria Uterina , Feminino , Gravidez , Humanos , Artéria Uterina/fisiologia , Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/fisiologia , Adaptação Fisiológica/fisiologia , AnimaisRESUMO
PURPOSE: The aim of this study is to investigate the origin and course of the orbital fat arterial supply in the lower eyelid using traditional anatomy and three-dimensional computed tomography (CT). METHODS: Twenty-seven cadaver heads were infused with mercury sulfide contrast media through the ophthalmic artery, maxillary artery, transverse facial artery, and facial artery. CT images were obtained after contrast agent injection, three-dimensional CT scans were reconstructed, and the cadaver heads were dissected. RESULTS: Forty-five qualified hemifaces showed that the orbital fat arterial supply in the lower eyelid originates primarily from the inferomedial muscular trunk (IMT) of the ophthalmic artery and the orbital branch of the infraorbital artery. The medial branch of the IMT terminated at the medial fat pad (35.6%) or the orbital floor (64.4%). The lateral branch terminated at the inferior oblique (IO) muscle (28.9%) or the central and lateral fat pads (17.8%). In 53.3%, the lateral branch extended to the anterior part of the lateral fat pad and terminated in the orbital wall or the zygomaticoorbital foramina. The orbital branch of the infraorbital artery coursed between the orbital floor and the orbital fat, providing supply to the IO muscle, inferior rectus (IR) muscle, nasolacrimal duct, and orbital fat. CONCLUSION: This study elucidated the origin and course of the orbital fat arterial supply in the lower eyelid, which may help to avoid reducing the blood supply of the orbital fat pedicles during surgery. LEVEL OF EVIDENCE IV: This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Assuntos
Tecido Adiposo , Cadáver , Pálpebras , Órbita , Tomografia Computadorizada por Raios X , Humanos , Pálpebras/irrigação sanguínea , Pálpebras/anatomia & histologia , Pálpebras/diagnóstico por imagem , Feminino , Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/anatomia & histologia , Órbita/irrigação sanguínea , Órbita/diagnóstico por imagem , Órbita/anatomia & histologia , Masculino , Imageamento Tridimensional , Pessoa de Meia-Idade , Adulto , Relevância ClínicaRESUMO
OBJECTIVE: The objective of the study is to demonstrate the innovation and utility of mesenteric tissue culture for discovering the microvascular growth dynamics associated with adipose-derived stromal vascular fraction (SVF) transplantation. Understanding how SVF cells contribute to de novo vessel growth (i.e., neovascularization) and host network angiogenesis motivates the need to make observations at single-cell and network levels within a tissue. METHODS: Stromal vascular fraction was isolated from the inguinal adipose of adult male Wistar rats, labeled with DiI, and seeded onto adult Wistar rat mesentery tissues. Tissues were then cultured in MEM + 10% FBS for 3 days and labeled for BSI-lectin to identify vessels. Alternatively, SVF and tissues from green fluorescent-positive (GFP) Sprague Dawley rats were used to track SVF derived versus host vasculature. RESULTS: Stromal vascular fraction-treated tissues displayed a dramatically increased vascularized area compared to untreated tissues. DiI and GFP+ tracking of SVF identified neovascularization involving initial segment formation, radial outgrowth from central hub-like structures, and connection of segments. Neovascularization was also supported by the formation of segments in previously avascular areas. New segments characteristic of SVF neovessels contained endothelial cells and pericytes. Additionally, a subset of SVF cells displayed the ability to associate with host vessels and the presence of SVF increased host network angiogenesis. CONCLUSIONS: The results showcase the use of the rat mesentery culture model as a novel tool for elucidating SVF cell transplant dynamics and highlight the impact of model selection for visualization.
Assuntos
Células Endoteliais , Células Estromais , Ratos , Masculino , Animais , Fração Vascular Estromal , Ratos Sprague-Dawley , Ratos Wistar , Microvasos , Tecido Adiposo/irrigação sanguínea , Neovascularização Patológica , MesentérioRESUMO
Lyl1 encodes a hematopoietic- and endothelial-specific bHLH transcription factor. Lyl1-deficient mice are viable, but they display mild hematopoietic and vascular defects. Specifically, LYL1 is required for the maturation and stabilization of blood vessel endothelial adherens junctions. Here, we report that young adult Lyl1-/- mice exhibit transient overweight associated with general expansion of adipose tissue, without signs of metabolic disorder and unrelated to food intake. The increased fat tissue development in Lyl1-/- mice resulted from earlier differentiation of adipose stem cells (ASCs) into adipocytes through noncell autonomous mechanisms. Specifically, we found that in Lyl1-/- mice, the adipose tissue vascular structures are immature, as indicated by their high permeability, reduced coverage by pericytes, lower recruitment of VE-cadherin and ZO1 at cell junctions, and more prone to angiogenesis. Together, our data show that in Lyl1-/- mice, the impaired vascular compartment of the adipose niche promotes ASC differentiation, leading to early adipocyte expansion and premature ASC depletion. Our study highlights the major structural role of the adipose tissue vascular niche in coordinating stem cell self-renewal and differentiation into adipocytes.
Assuntos
Tecido Adiposo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Proteínas de Neoplasias/deficiência , Neovascularização Patológica , Nicho de Células-Tronco , Células-Tronco/metabolismo , Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Células-Tronco/patologiaRESUMO
OBJECTIVE: Coronary artery disease (CAD) is associated with a compensatory switch in mechanism of flow-mediated dilation (FMD) from nitric oxide (NO) to H2O2. The underlying mechanism responsible for the pathological shift is not well understood, and recent reports directly implicate telomerase and indirectly support a role for autophagy. We hypothesize that autophagy is critical for shear stress-induced release of NO and is a crucial component of for the pathway by which telomerase regulates FMD. Approach and Results: Human left ventricular, atrial, and adipose resistance arterioles were collected for videomicroscopy and immunoblotting. FMD and autophagic flux were measured in arterioles treated with autophagy modulators alone, and in tandem with telomerase-activity modulators. LC3B II/I was higher in left ventricular tissue from patients with CAD compared with non-CAD (2.8±0.2 versus 1.0±0.2-fold change; P<0.05), although p62 was similar between groups. Shear stress increased Lysotracker fluorescence in non-CAD arterioles, with no effect in CAD arterioles. Inhibition of autophagy in non-CAD arterioles induced a switch from NO to H2O2, while activation of autophagy restored NO-mediated vasodilation in CAD arterioles. In the presence of an autophagy activator, telomerase inhibitor prevented the expected switch (Control: 82±4%; NG-Nitro-l-arginine methyl ester: 36±5%; polyethylene glycol catalase: 80±3). Telomerase activation was unable to restore NO-mediated FMD in the presence of autophagy inhibition in CAD arterioles (control: 72±7%; NG-Nitro-l-arginine methyl ester: 79±7%; polyethylene glycol catalase: 38±9%). CONCLUSIONS: We provide novel evidence that autophagy is responsible for the pathological switch in dilator mechanism in CAD arterioles, demonstrating that autophagy acts downstream of telomerase as a common denominator in determining the mechanism of FMD.
Assuntos
Tecido Adiposo/irrigação sanguínea , Arteríolas/enzimologia , Autofagia , Doença da Artéria Coronariana/enzimologia , Vasos Coronários/enzimologia , Telomerase/metabolismo , Vasodilatação , Adulto , Idoso , Arteríolas/patologia , Arteríolas/fisiopatologia , Estudos de Casos e Controles , Doença da Artéria Coronariana/patologia , Doença da Artéria Coronariana/fisiopatologia , Vasos Coronários/patologia , Vasos Coronários/fisiopatologia , Feminino , Humanos , Peróxido de Hidrogênio/metabolismo , Lisossomos/enzimologia , Lisossomos/patologia , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Pessoa de Meia-Idade , Óxido Nítrico/metabolismo , Transdução de SinaisRESUMO
BACKGROUND/OBJECTIVES: Ghrelin is an orexigenic hormone that increases food intake, adiposity, and insulin resistance through its receptor Growth Hormone Secretagogue Receptor (GHS-R). We previously showed that ghrelin/GHS-R signaling has important roles in regulation of energy homeostasis, and global deletion of GHS-R reduces obesity and improves insulin sensitivity by increasing thermogenesis. However, it is unknown whether GHS-R regulates thermogenic activation in adipose tissues directly. METHODS: We generated a novel adipose tissue-specific GHS-R deletion mouse model and characterized the mice under regular diet (RD) and high-fat diet (HFD) feeding. Body composition was measured by Echo MRI. Metabolic profiling was determined by indirect calorimetry. Response to environmental stress was assessed using a TH-8 temperature monitoring system. Insulin sensitivity was evaluated by glucose and insulin tolerance tests. Tissue histology was analyzed by hematoxylin/eosin and immunofluorescent staining. Expression of genes involved in thermogenesis, angiogenesis and fibrosis in adipose tissues were analyzed by real-time PCR. RESULTS: Under RD feeding, adipose tissue-specific GHS-R deletion had little or no impact on metabolic parameters. However, under HFD feeding, adipose tissue-specific GHS-R deletion attenuated diet-induced obesity and insulin resistance, showing elevated physical activity and heat production. In addition, adipose tissue-specific GHS-R deletion increased expression of master adipose transcription regulator of peroxisome proliferator-activated receptor (PPAR) γ1 and adipokines of adiponectin and fibroblast growth factor (FGF) 21; and differentially modulated angiogenesis and fibrosis evident in both gene expression and histological analysis. CONCLUSIONS: These results show that GHS-R has cell-autonomous effects in adipocytes, and suppression of GHS-R in adipose tissues protects against diet-induced obesity and insulin resistance by modulating adipose angiogenesis and fibrosis. These findings suggest adipose GHS-R may constitute a novel therapeutic target for treatment of obesity and metabolic syndrome.
Assuntos
Tecido Adiposo/metabolismo , Resistência à Insulina/genética , Obesidade/metabolismo , Receptores de Grelina , Termogênese/genética , Adipócitos/citologia , Adipócitos/metabolismo , Adiponectina/metabolismo , Tecido Adiposo/irrigação sanguínea , Animais , Dieta Hiperlipídica , Fibrose/metabolismo , Masculino , Camundongos , Receptores de Grelina/genética , Receptores de Grelina/metabolismoRESUMO
BACKGROUND: Though autologous fat transplantation is regularly and successfully performed in plastic surgery, little is known about the factors that contribute to the rise of preadipocytes and how the viability of adipocytes is regulated. As sufficient blood supply is a key parameter for the transplant's survival, we opted to analyse the development of preadipocytes within the fat transplant via stimulation of tissue perfusion with the angiogenesis enhancing hormone leptin. METHODS: In a murine (C57BL/6N) model inguinal fat was autologously transplanted into a dorsal skinfold chamber. In the intervention group the fat transplant was treated with local administration of leptin (3 µg/ml) at days 3, 7 and 10 after transplantation. Saline solution was administered respectively in the control group. On the postoperative days 3, 7, 10, and 15 intra vital microscopy was done to assess the functional vessel density, vessel diameter, adipocyte survival and preadipocyte development. The study was completed by histological tissue analysis on days 15 after transplantation. RESULTS: Leptin administration leads to an increase of angiogenesis, which starts from day 7 after implantation and elevates perfusion as well as functional vessel density FVD at days 10 and 15 after transplantation. Perfusion develops first from the border zones of the transplant. Histological evaluation showed that the percentage of perilipin positive adipocytes increased markedly in the study group of mice. Moreover, fat transplants of mice of the leptin group disclosed significantly higher Pref-1 positive cells than fat transplants of the control group. The findings reported in this study indicate that the leptin can enhance the survival and the quality of grafted fat tissue, which may be due to induction of angiogenesis. CONCLUSION: Leptin administration to fat transplants induced an increase in angiogenesis in the transplanted tissue and may play a role in reducing the resorption rate of lipoaspirates.
Assuntos
Adipócitos/transplante , Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/transplante , Indutores da Angiogênese/farmacologia , Leptina/farmacologia , Transplante de Células-Tronco Mesenquimais , Neovascularização Fisiológica/efeitos dos fármacos , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Sobrevivência Celular , Feminino , Canal Inguinal , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Microcirculação , Fluxo Sanguíneo Regional , Transplante AutólogoRESUMO
The perivascular adipose tissue (PVAT) refers to an ectopic local deposit of connective tissue that anatomically surrounds most of the blood vessels. While it was initially known only as a structural support for vasculature, the landmark findings of Soltis and Cassis (1991), first demonstrating that PVAT reduces the contractions of norepinephrine in the isolated rat aorta, brought the potential vascular role of PVAT into the limelight. This seminal work implied the potential ability of PVAT to influence vascular responsiveness. Several vasoactive/vasocrine substances influencing vascular homeostasis were successively shown to be released from PVAT that include both adipocyte-derived relaxing and contracting factors. The PVAT is currently recognized as a metabolically active endocrine organ and is eventually considered as the 'protagonist' in vascular homeostasis. It plays prominent defending and opposing roles in vascular function, while the actual vascular influences of PVAT vary with an increase in adiposity. Recent studies have presented compelling evidence implicating the pivotal role of PVAT in the local activation of the renin-angiotensin system (RAS), which substantially impacts vascular physiology and physiopathology. Current findings have advanced our understanding of the role of PVAT in favorably or adversely modulating the vascular function through differential RAS activation. Given that adipocytes also produce major RAS components locally to influence vascular function, this review provides a scientific basis to distinctly understand the key role of PVAT in regulating the autocrine and paracrine functions of vascular RAS components and its potential as an emerging therapeutic target for mitigating cardiovascular complications.
Assuntos
Tecido Adiposo/irrigação sanguínea , Sistema Renina-Angiotensina , Tecido Adiposo/fisiologia , Animais , HumanosRESUMO
OBJECTIVE: In mice fed a high-fat diet, impairment of insulin signaling in endothelium is an early phenomenon that precedes decreased insulin sensitivity of skeletal muscle, adipose tissue, and liver. We assessed in humans whether short-term overfeeding affects insulin-induced microvascular recruitment in skeletal muscle and adipose tissue before changes occur in glucose uptake and lipolysis. Approach and Results: Fifteen healthy males underwent a hypercaloric and subsequent hypocaloric diet intervention. Before, during, and after the hypercaloric diet, and upon return to baseline weight, all participants underwent (1) a hyperinsulinemic-euglycemic clamp to determine insulin-induced glucose uptake and suppression of lipolysis (2) contrast-enhanced ultrasonography to measure insulin-induced microvascular recruitment in skeletal muscle and adipose tissue. In addition, we assessed insulin-induced vasodilation of isolated skeletal muscle resistance arteries by pressure myography after the hypercaloric diet in study participants and controls (n=5). The hypercaloric diet increased body weight (3.5 kg; P<0.001) and fat percentage (3.5%; P<0.001) but did not affect glucose uptake nor lipolysis. The hypercaloric diet increased adipose tissue microvascular recruitment (P=0.041) and decreased the ratio between skeletal muscle and adipose tissue microvascular blood volume during hyperinsulinemia (P=0.019). Insulin-induced vasodilation of isolated skeletal muscle arterioles was significantly lower in participants compared with controls (P<0.001). The hypocaloric diet reversed all of these changes, except the increase in adipose tissue microvascular recruitment. CONCLUSIONS: In lean men, short-term overfeeding reduces insulin-induced vasodilation of skeletal muscle resistance arteries and shifts the distribution of tissue perfusion during hyperinsulinemia from skeletal muscle to adipose tissue without affecting glucose uptake and lipolysis. Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT02628301.
Assuntos
Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/metabolismo , Arteríolas/efeitos dos fármacos , Glicemia/efeitos dos fármacos , Restrição Calórica , Ingestão de Energia , Insulina/administração & dosagem , Lipólise/efeitos dos fármacos , Microcirculação/efeitos dos fármacos , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/metabolismo , Adiposidade , Adolescente , Adulto , Arteríolas/fisiologia , Glicemia/metabolismo , Estudos de Casos e Controles , Voluntários Saudáveis , Humanos , Resistência à Insulina , Masculino , Fatores de Tempo , Vasodilatação/efeitos dos fármacos , Aumento de Peso , Redução de Peso , Adulto JovemRESUMO
Investigations into the mechanisms regulating obesity are frantic and novel translational approaches are needed. The raccoon dog (Nyctereutes procyonoides) is a canid species representing a promising model to study metabolic regulation in a species undergoing cycles of seasonal obesity and fasting. To understand the molecular mechanisms of metabolic regulation in seasonal adaptation, we analyzed key central nervous system and peripheral signals regulating food intake and metabolism from raccoon dogs after autumnal fattening and winter fasting. Expressions of neuropeptide Y (NPY), orexin-2 receptor (OX2R), pro-opiomelanocortin (POMC) and leptin receptor (ObRb) were analyzed as examples of orexigenic and anorexigenic signals using qRT-PCR from raccoon dog hypothalamus samples. Plasma metabolic profiles were measured with 1H NMR-spectroscopy and LC-MS. Circulating hormones and cytokines were determined with canine specific antibody assays. Surprisingly, NPY and POMC were not affected by the winter fasting nor autumn fattening and the metabolic profiles showed a remarkable equilibrium, indicating conserved homeostasis. However, OX2R and ObRb expression changes suggested seasonal regulation. Circulating cytokine levels were not increased, demonstrating that the autumn fattening did not induce subacute inflammation. Thus, the raccoon dog developed seasonal regulatory mechanisms to accommodate the autumnal fattening and prolonged fasting making the species unique in coping with the extreme environmental challenges.
Assuntos
Adiposidade , Jejum/metabolismo , Metaboloma , Cães Guaxinins/metabolismo , Estações do Ano , Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/patologia , Animais , Biomarcadores/metabolismo , Peso Corporal , Análise Discriminante , Feminino , Hormônios/sangue , Hipotálamo/metabolismo , Inflamação/patologia , Análise dos Mínimos Quadrados , Limite de Detecção , Análise Multivariada , Peptídeos/genética , Peptídeos/metabolismo , Espectroscopia de Prótons por Ressonância Magnética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Cães Guaxinins/sangue , Receptores de Peptídeos/metabolismoRESUMO
BACKGROUND: Cross-linked hyaluronic acid (HA) is an active anti-aging cosmetic filler. The combination of cross-linked HA and preadipocytes or adipose-derived stem cells has been previously investigated, but the effects of agglomerated cross-linked HA injection on the vascularization of fat grafts remain unclear. OBJECTIVES: The aim of this study was to explore the effects of agglomerated cross-linked HA injection on the vascularization of fat grafts. METHODS: The backs of nude mice were divided into 4 regions that received different treatments: nothing (control group), agglomerated Biohyalux (HA group), agglomerated fat (FAT group), and lumps formed by the sequential injection of Biohyalux and fat (HA/FAT group). Samples were collected after 1 month for weighing and hematoxylin and eosin staining, immunohistochemistry, image analysis, and Western blotting. RESULTS: The weight of fat and the mean number of adipocytes in the HA/FAT group did not significantly differ from those in the FAT group. No living tissue was found in agglomerated HA. Some tiny HA particles were surrounded by tissue rich in blood vessels. The expression levels of CD31 and vascular endothelial growth factor (VEGF) in the HA/FAT group were higher than those in the FAT group, but the difference was only significant for VEGF expression. CONCLUSIONS: Cross-linked HA had minimal effect on the early retention rate of surrounding fat grafts, but enhanced their vascularization. Fat grafts should be not injected into lumps of cross-linked HA. Therefore, agglomerated cross-linked HA should be dissolved before fat transplantation.
Assuntos
Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/transplante , Ácido Hialurônico , Neovascularização Fisiológica , Adipócitos , Animais , Sobrevivência de Enxerto , Humanos , Camundongos , Camundongos Nus , Fator A de Crescimento do Endotélio VascularRESUMO
Tumor-associated lymphangiogenesis correlates with lymph node metastasis and poor outcome in several human malignancies. In addition, the presence of functional lymphatic vessels regulates the formation of tumor inflammatory and immune microenvironments. Although lymphatic structures are often found deeply integrated into the fabric of adipose tissue, the impact of lymphangiogenesis on tumor-associated adipose tissue (AT) has not yet been investigated. Using K14-VEGFR3-Ig mice that constitutively express soluble vascular endothelial growth factor receptor (VEGFR) 3-Ig in the skin, scavenging VEGF-C and VEGF-D, the role of lymphangiogenesis in the generation of an inflammatory response within tumor-associated AT was studied. Macrophages expressing lymphatic vessel endothelial hyaluronan receptor-1 were found within peritumoral adipose tissue from melanoma-bearing K14-VEGFR3-Ig mice, which were further enriched with alternatively activated macrophages based on surface marker CD301/C-type lectin domain family 10 member A expression. The blockade of lymphangiogenesis also resulted in accumulation of the cytokine IL-6, which correlated with enhanced macrophage proliferation of the alternatively activated phenotype. Furthermore, melanomas co-implanted with freshly isolated adipose tissue macrophages grew more robustly than melanomas growing alone. In human cutaneous melanomas, adipocyte-selective FABP4 transcripts closely correlated with gene signatures of CLEC10A and were associated with poor overall survival. These data suggest that the blockade of pathways regulating lymphatic vessel formation shapes an inflammatory response within tumor-associated AT by facilitating accumulation of tumor-promoting alternatively activated macrophages.
Assuntos
Tecido Adiposo/patologia , Inflamação/patologia , Linfangiogênese , Melanoma Experimental/patologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/imunologia , Animais , Feminino , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Tumorais Cultivadas , Microambiente TumoralRESUMO
Despite being considered present in most vascularised tissues, lymphatic vessels have not been properly shown in human adipose tissue (AT). Our goal in this study is to investigate an unanswered question in AT biology, regarding lymphatic network presence in tissue parenchyma. Using human subcutaneous (S-) and visceral (V-) AT samples with whole mount staining for lymphatic specific markers and three-dimensional imaging, we showed lymphatic capillaries and larger lymphatic vessels in the human VAT. Conversely, in the human SAT, microcirculatory lymphatic vascular structures were rarely detected and no initial lymphatics were found.
Assuntos
Tecido Adiposo/anatomia & histologia , Vasos Linfáticos/anatomia & histologia , Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/fisiologia , Feminino , Humanos , Imageamento Tridimensional , Imuno-Histoquímica , Gordura Intra-Abdominal/anatomia & histologia , Gordura Intra-Abdominal/irrigação sanguínea , Gordura Intra-Abdominal/fisiologia , Vasos Linfáticos/irrigação sanguínea , Vasos Linfáticos/fisiologia , Masculino , Pessoa de Meia-Idade , Gordura Subcutânea/anatomia & histologia , Gordura Subcutânea/irrigação sanguínea , Gordura Subcutânea/fisiologiaRESUMO
Perivascular adipose tissue (PVAT) dysfunction is associated with vascular damage in cardiometabolic diseases. Although heart failure (HF)-induced endothelial dysfunction is associated with renin-angiotensin system (RAS) activation, no data have correlated this syndrome with PVAT dysfunction. Thus, the aim of the present study was to investigate whether the hyperactivation of the RAS in PVAT participates in the vascular dysfunction observed in rats with HF after myocardial infarction surgery. Wire myograph studies were carried out in thoracic aorta rings in the presence and absence of PVAT. An anticontractile effect of PVAT was observed in the rings of the control rats in the presence (33%) or absence (11%) of endothelium. Moreover, this response was substantially reduced in animals with HF (5%), and acute type 1 angiotensin II receptor (AT1R) and type 2 angiotensin II receptor (AT2R) blockade restored the anticontractile effect of PVAT. In addition, the angiotensin-converting enzyme 1 (ACE1) activity (26%) and angiotensin II levels (51%), as well as the AT1R and AT2R gene expression, were enhanced in the PVAT of rats with HF. Associated with these alterations, HF-induced lower nitric oxide bioavailability, oxidative stress and whitening of the PVAT, which suggests changes in the secretory function of this tissue. The ACE1/angiotensin II/AT1R and AT2R axes are involved in thoracic aorta PVAT dysfunction in rats with HF. These results suggest PVAT as a target in the pathophysiology of vascular dysfunction in HF and provide new perspectives for the treatment of this syndrome.
Assuntos
Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/fisiopatologia , Insuficiência Cardíaca/fisiopatologia , Sistema Renina-Angiotensina , Angiotensina II/metabolismo , Animais , Aorta Torácica/patologia , Disponibilidade Biológica , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Insuficiência Cardíaca/complicações , Hemodinâmica , Masculino , Modelos Biológicos , Infarto do Miocárdio/complicações , Infarto do Miocárdio/fisiopatologia , Óxido Nítrico/metabolismo , Estresse Oxidativo , Peptidil Dipeptidase A/metabolismo , Ratos Wistar , Receptor Tipo 1 de Angiotensina/metabolismo , VasoconstriçãoRESUMO
Tissue constructs of physiologically relevant scale require a vascular system to maintain cell viability. However, in vitro vascularization of engineered tissues is still a major challenge. Successful approaches are based on a feeder layer (FL) to support vascularization. Here, we investigated whether the supporting effect on the self-assembled formation of prevascular-like structures by microvascular endothelial cells (mvECs) originates from the FL itself or from its extracellular matrix (ECM). Therefore, we compared the influence of ECM, either derived from adipose-derived stem cells (ASCs) or adipogenically differentiated ASCs, with the classical cell-based FL. All cell-derived ECM (cdECM) substrates enabled mvEC growth with high viability. Prevascular-like structures were visualized by immunofluorescence staining of endothelial surface protein CD31 and could be observed on all cdECM and FL substrates but not on control substrate collagen I. On adipogenically differentiated ECM, longer and higher branched structures could be found compared with stem cell cdECM. An increased concentration of proangiogenic factors was found in cdECM substrates and FL approaches compared with controls. Finally, the expression of proteins associated with tube formation (E-selectin and thrombomodulin) was confirmed. These results highlight cdECM as promising biomaterial for adipose tissue engineering by inducing the spontaneous formation of prevascular-like structures by mvECs.
Assuntos
Tecido Adiposo/citologia , Materiais Biocompatíveis/química , Células Endoteliais/citologia , Matriz Extracelular/química , Células-Tronco/citologia , Tecido Adiposo/irrigação sanguínea , Diferenciação Celular/fisiologia , Células Cultivadas , Matriz Extracelular/metabolismo , Humanos , Neovascularização FisiológicaRESUMO
Methylglyoxal was shown to impair adipose tissue capillarization and insulin sensitivity in obese models. We hypothesized that glyoxalase-1 (GLO-1) activity could be diminished in the adipose tissue of type 2 diabetic obese patients. Moreover, we assessed whether such activity could be increased by GLP-1-based therapies in order to improve adipose tissue capillarization and insulin sensitivity. GLO-1 activity was assessed in visceral adipose tissue of a cohort of obese patients. The role of GLP-1 in modulating GLO-1 was assessed in type 2 diabetic GK rats submitted to sleeve gastrectomy or Liraglutide treatment, in the adipose tissue angiogenesis assay and in the HUVEC cell line. Glyoxalase-1 activity was decreased in visceral adipose tissue of pre-diabetic and diabetic obese patients, together with other markers of adipose tissue dysfunction and correlated with increased HbA1c levels. Decreased adipose tissue GLO-1 levels in GK rats were increased by sleeve gastrectomy and Liraglutide, being associated with overexpression of angiogenic and vasoactive factors, as well as insulin receptor phosphorylation (Tyr1161). Moreover, GLP-1 increased adipose tissue capillarization and HUVEC proliferation in a glyoxalase-dependent manner. Lower adipose tissue GLO-1 activity was observed in dysmetabolic patients, being a target for GLP-1 in improving adipose tissue capillarization and insulin sensitivity.
Assuntos
Tecido Adiposo/irrigação sanguínea , Capilares/efeitos dos fármacos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Hipoglicemiantes/farmacologia , Incretinas/farmacologia , Resistência à Insulina , Lactoilglutationa Liase/metabolismo , Liraglutida/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Adulto , Idoso , Animais , Capilares/enzimologia , Capilares/fisiopatologia , Células Cultivadas , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Feminino , Gastrectomia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Masculino , Pessoa de Meia-Idade , Obesidade/enzimologia , Obesidade/fisiopatologia , Obesidade/cirurgia , Ratos Wistar , Transdução de SinaisRESUMO
OBJECTIVE: Infrapatellar fat pad (IPFP) fat-suppressed T2 (T2FS) hyperintense regions on MRI are an important imaging feature of knee osteoarthritis (OA) and are thought to represent inflammation. These regions are also common in non-OA subjects, and may not always be linked to inflammation. Our aim was to evaluate quantitative blood perfusion parameters, as surrogate measure of inflammation, within T2FS-hyperintense regions in patients with OA, with patellofemoral pain (PFP) (supposed OA precursor), and control subjects. METHODS: Twenty-two knee OA patients, 35 PFP patients and 43 healthy controls were included and underwent MRI, comprising T2 and DCE-MRI sequences. T2FS-hyperintense IPFP regions were delineated and a reference region was drawn in adjacent IPFP tissue with normal signal intensity. After fitting the extended Tofts pharmacokinetic model, quantitative DCE-MRI perfusion parameters were compared between the two regions within subjects in each subgroup, using a paired Wilcoxon signed-rank test. RESULTS: T2FS-hyperintense IPFP regions were present in 16 of 22 (73%) OA patients, 13 of 35 (37%) PFP patients, and 14 of 43 (33%) controls. DCE-MRI perfusion parameters were significantly different between regions with and without a T2FS-hyperintense signal in OA patients, demonstrating higher Ktrans compared to normal IFPF tissue (0.039 min-1 versus 0.025 min-1, p = 0.017) and higher Ve (0.157 versus 0.119, p = 0.010). For PFP patients and controls no significant differences were found. CONCLUSIONS: IPFP T2FS-hyperintense regions are associated with higher perfusion in knee OA patients in contrast to identically appearing regions in PFP patients and controls, pointing towards an inflammatory pathogenesis in OA only. KEY POINTS: ⢠Morphologically identical appearing T2FS-hyperintense infrapatellar fat pad regions show different perfusion in healthy subjects, subjects with patellofemoral pain, and subjects with knee osteoarthritis. ⢠Elevated DCE-MRI perfusion parameters within T2FS-hyperintense infrapatellar fat pad regions in patients with osteoarthritis suggest an inflammatory pathogenesis in osteoarthritis, but not in patellofemoral pain and healthy subjects.
Assuntos
Tecido Adiposo/diagnóstico por imagem , Articulação do Joelho/diagnóstico por imagem , Osteoartrite do Joelho/diagnóstico por imagem , Síndrome da Dor Patelofemoral/diagnóstico por imagem , Tecido Adiposo/irrigação sanguínea , Adulto , Idoso , Estudos de Casos e Controles , Meios de Contraste , Feminino , Humanos , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Adulto JovemRESUMO
OBJECTIVE: Endothelial cells (EC) in obese adipose tissue (AT) are exposed to a chronic proinflammatory environment that may induce a mesenchymal-like phenotype and altered function. The objective of this study was to establish whether endothelial-to-mesenchymal transition (EndoMT) is present in human AT in obesity and to investigate the effect of such transition on endothelial function and the endothelial particulate secretome represented by extracellular vesicles (EV). Approach and Results: We identified EndoMT in obese human AT depots by immunohistochemical co-localization of CD31 or vWF and α-SMA (alpha-smooth muscle actin). We showed that AT EC exposed in vitro to TGF-ß (tumor growth factor-ß), TNF-α (tumor necrosis factor-α), and IFN-γ (interferon-γ) undergo EndoMT with progressive loss of endothelial markers. The phenotypic change results in failure to maintain a tight barrier in culture, increased migration, and reduced angiogenesis. EndoMT also reduced mitochondrial oxidative phosphorylation and glycolytic capacity of EC. EVs produced by EC that underwent EndoMT dramatically reduced angiogenic capacity of the recipient naïve ECs without affecting their migration or proliferation. Proteomic analysis of EV produced by EC in the proinflammatory conditions showed presence of several pro-inflammatory and immune proteins along with an enrichment in angiogenic receptors. CONCLUSIONS: We demonstrated the presence of EndoMT in human AT in obesity. EndoMT in vitro resulted in production of EV that transferred some of the functional and metabolic features to recipient naïve EC. This result suggests that functional and molecular features of EC that underwent EndoMT in vivo can be disseminated in a paracrine or endocrine fashion and may induce endothelial dysfunction in distant vascular beds.
Assuntos
Tecido Adiposo/irrigação sanguínea , Transição Epitelial-Mesenquimal/genética , Neovascularização Patológica/genética , Obesidade/genética , Fator de Crescimento Transformador beta1/farmacologia , Tecido Adiposo/metabolismo , Análise de Variância , Biomarcadores/metabolismo , Estudos de Casos e Controles , Movimento Celular/genética , Proliferação de Células/genética , Células Cultivadas , Células Endoteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Citometria de Fluxo/métodos , Humanos , Obesidade/fisiopatologia , Proteômica/métodosRESUMO
Decompression sickness (DCS) occurs when nitrogen gas (N2) comes out of solution too quickly, forming bubbles in the blood and tissues. These bubbles can be a serious condition; thus it is of extreme interest in the dive community to model DCS risk. Diving models use tissue compartments to calculate tissue partial pressures, often using data obtained from other mammalian species (i.e., pigs). Adipose tissue is an important compartment in these models because N2 is five times more soluble in fat than in blood; at any blood/tissue interface N2 will diffuse into the fat and can lead to bubble formation on ascent. Little is known about many characteristics of adipose tissue relevant to diving physiology. Therefore, we measured microvessel density and morphology, lipid composition, and N2 solubility in adipose tissue from humans and pigs. Human adipose tissue has significantly higher microvascular density (1.79 ± 0.04 vs. 1.21 ± 0.30%), vessel diameter (10.25 ± 0.28 vs. 6.72 ± 0.60 µm), total monounsaturated fatty acids (50.1 vs. 41.2 mol%) and N2 solubility (0.061 ± 0.003 vs. 0.054 ± 0.004 mL N2 mLâ» ¹ oil) compared to pig tissue. Pig adipose tissue has significantly higher lipid content (76.1 ± 4.9 vs. 64.6 ± 5.1%) and total saturated fatty acids (38.8 vs. 29.5 mol%). Though two important components in gas kinetics within adipose tissue during diving (blood flow rates and degree of perfusion) are not well understood, our results indicate differences between the adipose tissue of humans and pigs. This suggests data from swine may not exactly predict gas dynamics for estimating DCS in humans.