Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Life Sci ; 335: 122285, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37995934

RESUMO

AIMS: The goal of this study was to identify mediators in peri-lymphatic adipose tissue (PLAT) that are altered in obese versus lean Zucker rats, with focus on potential sex differences MAIN METHODS: Mesenteric PLAT was analyzed with protein and lncRNA arrays. Additional RT-PCR confirmation was performed with epididymal/ovarian fat. KEY FINDINGS: MCP-1, TCK-1, Galectin-1, Galectin-3, and neuropilin-1 were elevated in PLAT from obese rats of both sexes. However, 11 additional proteins were elevated only in obese males while 24 different proteins were elevated in obese females. Profiling of lncRNAs revealed lean males have elevated levels of NEAT1, MALAT1 and GAS5 compared to lean females. NEAT1, MALAT1, and GAS5 were significantly reduced with obesity in males but not in females. Another lncRNA, HOTAIR, was higher in lean females compared to males, and its levels in females were reduced with obesity. Obese rats of both sexes had similar histologic findings of mesenteric macrophage crown-like structures and hepatocyte fat accumulation. SIGNIFICANCE: While obese male and female Zucker rats both have increased inflammation, they have distinct signals. Future studies of the proteome and lncRNA landscape of obese males vs. females in various animal models and in human subjects are warranted to better guide development of therapeutics for obesity-induced inflammation.


Assuntos
RNA Longo não Codificante , Feminino , Masculino , Ratos , Humanos , Animais , RNA Longo não Codificante/genética , Ratos Zucker , Obesidade/genética , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Inflamação/genética , Inflamação/metabolismo , Mediadores da Inflamação
2.
Am J Physiol Heart Circ Physiol ; 324(5): H610-H623, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36867447

RESUMO

Microvascular hyperpermeability is a hallmark of inflammation. Many negative effects of hyperpermeability are due to its persistence beyond what is required for preserving organ function. Therefore, we propose that targeted therapeutic approaches focusing on mechanisms that terminate hyperpermeability would avoid the negative effects of prolonged hyperpermeability while retaining its short-term beneficial effects. We tested the hypothesis that inflammatory agonist signaling leads to hyperpermeability and initiates a delayed cascade of cAMP-dependent pathways that causes inactivation of hyperpermeability. We applied platelet-activating factor (PAF) and vascular endothelial growth factor (VEGF) to induce hyperpermeability. We used an Epac1 agonist to selectively stimulate exchange protein activated by cAMP (Epac1) and promote inactivation of hyperpermeability. Stimulation of Epac1 inactivated agonist-induced hyperpermeability in the mouse cremaster muscle and in human microvascular endothelial cells (HMVECs). PAF induced nitric oxide (NO) production and hyperpermeability within 1 min and NO-dependent increased cAMP concentration in about 15-20 min in HMVECs. PAF triggered phosphorylation of vasodilator-stimulated phosphoprotein (VASP) in a NO-dependent manner. Epac1 stimulation promoted cytosol-to-membrane eNOS translocation in HMVECs and in myocardial microvascular endothelial (MyEnd) cells from wild-type mice, but not in MyEnd cells from VASP knockout mice. We demonstrate that PAF and VEGF cause hyperpermeability and stimulate the cAMP/Epac1 pathway to inactivate agonist-induced endothelial/microvascular hyperpermeability. Inactivation involves VASP-assisted translocation of eNOS from the cytosol to the endothelial cell membrane. We demonstrate that hyperpermeability is a self-limiting process, whose timed inactivation is an intrinsic property of the microvascular endothelium that maintains vascular homeostasis in response to inflammatory conditions.NEW & NOTEWORTHY Termination of microvascular hyperpermeability has been so far accepted to be a passive result of the removal of the applied proinflammatory agonists. We provide in vivo and in vitro evidence that 1) inactivation of hyperpermeability is an actively regulated process, 2) proinflammatory agonists (PAF and VEGF) stimulate microvascular hyperpermeability and initiate endothelial mechanisms that terminate hyperpermeability, and 3) eNOS location-translocation is critical in the activation-inactivation cascade of endothelial hyperpermeability.


Assuntos
Células Endoteliais , Fator A de Crescimento do Endotélio Vascular , Camundongos , Humanos , Animais , Células Endoteliais/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Inflamação/metabolismo , Fator de Ativação de Plaquetas/metabolismo , Fator de Ativação de Plaquetas/farmacologia , Camundongos Knockout , Endotélio/metabolismo , Permeabilidade Capilar , Endotélio Vascular/metabolismo
3.
Am J Physiol Heart Circ Physiol ; 321(6): H1083-H1095, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34652985

RESUMO

Nitric oxide (NO) is a key factor in inflammation. Endothelial nitric oxide synthase (eNOS), whose activity increases after stimulation with proinflammatory cytokines, produces NO in endothelium. NO activates two pathways: 1) soluble guanylate cyclase-protein kinase G and 2) S-nitrosylation (NO-induced modification of free-thiol cysteines in proteins). S-nitrosylation affects phosphorylation, localization, and protein interactions. NO is classically described as a negative regulator of leukocyte adhesion to endothelial cells. However, agonists activating NO production induce a fast leukocyte adhesion, which suggests that NO might positively regulate leukocyte adhesion. We tested the hypothesis that eNOS-induced NO promotes leukocyte adhesion through the S-nitrosylation pathway. We stimulated leukocyte adhesion to endothelium in vitro and in vivo using tumor necrosis factor-α (TNF-α) as proinflammatory agonist. ICAM-1 changes were evaluated by immunofluorescence, subcellular fractionation, immunoprecipitation, and fluorescence recovery after photobleaching (FRAP). Protein kinase Cζ (PKCζ) activity and S-nitrosylation were evaluated by Western blot analysis and biotin switch method, respectively. TNF-α, at short times of stimulation, activated the eNOS S-nitrosylation pathway and caused leukocyte adhesion to endothelial cells in vivo and in vitro. TNF-α-induced NO led to changes in ICAM-1 at the cell surface, which are characteristic of clustering. TNF-α-induced NO also produced S-nitrosylation and phosphorylation of PKCζ, association of PKCζ with ICAM-1, and ICAM-1 phosphorylation. The inhibition of PKCζ blocked leukocyte adhesion induced by TNF-α. Mass spectrometry analysis of purified PKCζ identified cysteine 503 as the only S-nitrosylated residue in the kinase domain of the protein. Our results reveal a new eNOS S-nitrosylation-dependent mechanism that induces leukocyte adhesion and suggests that S-nitrosylation of PKCζ may be an important regulatory step in early leukocyte adhesion in inflammation.NEW & NOTEWORTHY Contrary to the well-established inhibitory role of NO in leukocyte adhesion, we demonstrate a positive role of nitric oxide in this process. We demonstrate that NO induced by eNOS after TNF-α treatment induces early leukocyte adhesion activating the S-nitrosylation pathway. Our data suggest that PKCζ S-nitrosylation may be a key step in this process.


Assuntos
Músculos Abdominais/irrigação sanguínea , Adesão Celular , Células Endoteliais/efeitos dos fármacos , Leucócitos/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Linhagem Celular , Técnicas de Cocultura , Células Endoteliais/enzimologia , Ativação Enzimática , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fosforilação , Proteína Quinase C/metabolismo , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Fatores de Tempo
4.
Am J Physiol Gastrointest Liver Physiol ; 318(4): G705-G716, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32090604

RESUMO

Anemia is a frequent diagnosis in critically ill infants, but the clinical implications of severe anemia in these patients remain unclear. In this study, we examined preweaned mice to investigate the effects of severe anemia during early infancy on gut mucosal permeability. C57BL/6 mice were subjected to timed phlebotomy between postnatal days (P) 2-10 to induce severe anemia (hematocrits 20%-24%), and intestinal permeability was tracked longitudinally between P10 and P20 as intestine-to-plasma translocation of enteral macromolecules and bacterial translocation. Epithelial junctions were evaluated by electron microscopy, polymerase chain reactions, immunohistochemistry, and/or enzyme immunoassays on intestinal tissues, Caco-2 intestinal epithelial-like cells, and colonic organoids. Preweaned mouse pups showed an age-related susceptibility to severe anemia, with increased intestinal permeability to enteral macromolecules (dextran, ovalbumin, ß-lactoglobulin) and luminal bacteria. Electron micrographs showed increased paracellular permeability and ultrastructural abnormalities of the adherens junctions. These findings were explained by the loss of E-cadherin in epithelial cells, which was caused by destabilization of the E-cadherin (Cdh1) mRNA because of microRNA let-7e-5p binding to the 3'-untranslated region. Severe anemia resulted in a disproportionate and persistent increase in intestinal permeability in preweaned mice because of the disruption of epithelial adherens junctions. These changes are mediated via microRNA let-7e-mediated depletion of Cdh1 mRNA.NEW & NOTEWORTHY This research article shows that newborn infants with severe anemia show an age-related susceptibility to developing increased intestinal permeability to ingested macromolecules. This abnormal permeability develops because of abnormalities in intestinal epithelial junctions caused by a deficiency of the molecule E-cadherin in epithelial cells. The deficiency of E-cadherin is caused by destabilization of its mRNA precursor because of increased expression and binding of another molecule, the microRNA let-7e-5p, to the E-cadherin mRNA.


Assuntos
Junções Aderentes/patologia , Anemia Neonatal/patologia , Mucosa Intestinal/patologia , Intestinos/patologia , Junções Aderentes/ultraestrutura , Animais , Animais Recém-Nascidos , Células CACO-2 , Caderinas/genética , Caderinas/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , MicroRNAs/metabolismo , Permeabilidade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
5.
J Physiol ; 597(13): 3255-3279, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31077371

RESUMO

KEY POINTS: We investigated the cardiovascular and respiratory responses of the normotensive Wistar-Kyoto (WKY) rat and the spontaneously hypertensive (SH) rat to inhalation and intravenous injection of the noxious stimuli allyl isothiocyanate (AITC). AITC inhalation evoked atropine-sensitive bradycardia in conscious WKY rats, and evoked atropine-sensitive bradycardia and atenolol-sensitive tachycardia with premature ventricular contractions (PVCs) in conscious SH rats. Intravenous injection of AITC evoked bradycardia but no tachycardia/PVCs in conscious SHs, while inhalation and injection of AITC caused similar bradypnoea in conscious SH and WKY rats. Anaesthesia (inhaled isoflurane) inhibited the cardiac reflexes evoked by inhaled AITC but not injected AITC. Data indicate the presence of a de novo nociceptive pulmonary-cardiac reflex triggering sympathoexcitation in SH rats, and this reflex is dependent on vagal afferents but is not due to steady state blood pressure or due to remodelling of vagal efferent function. ABSTRACT: Inhalation of noxious irritants/pollutants activates airway nociceptive afferents resulting in reflex bradycardia in healthy animals. Nevertheless, noxious pollutants evoke sympathoexcitation (tachycardia, hypertension) in cardiovascular disease patients. We hypothesize that cardiovascular disease alters nociceptive pulmonary-cardiac reflexes. Here, we studied reflex responses to irritants in normotensive Wistar-Kyoto (WKY) rats and spontaneously hypertensive (SH) rats. Inhaled allyl isothiocyanate (AITC) evoked atropine-sensitive bradycardia with atrial-ventricular (AV) block in conscious WKY rats, thus indicating a parasympathetic reflex. Conversely, inhaled AITC in conscious SH rats evoked complex brady-tachycardia with both AV block and premature ventricular contractions (PVCs). Atropine abolished the bradycardia and AV block, but the atropine-insensitive tachycardia and PVCs were abolished by the ß1 -adrenoceptor antagonist atenolol. The aberrant AITC-evoked reflex in SH rats was not reduced by acute blood pressure reduction by captopril. Surprisingly, intravenous AITC only evoked bradycardia in conscious SH and WKY rats. Furthermore, anaesthesia reduced the cardiac reflexes evoked by inhaled but not injected AITC. Nevertheless, anaesthesia had little effect on AITC-evoked respiratory reflexes. Such data suggest distinct differences in nociceptive reflex pathways dependent on cardiovascular disease, administration route and downstream effector. AITC-evoked tachycardia in decerebrate SH rats was abolished by vagotomy. Finally, there was no difference in the cardiac responses of WKY and SH rats to vagal efferent electrical stimulation. Our data suggest that AITC inhalation in SH rats evokes de novo adrenergic reflexes following vagal afferent activation. This aberrant reflex is independent of steady state hypertension and is not evoked by intravenous AITC. We conclude that pre-existing hypertension aberrantly shifts nociceptive pulmonary-cardiac reflexes towards sympathoexcitation.


Assuntos
Pressão Sanguínea/fisiologia , Coração/fisiopatologia , Hipertensão/fisiopatologia , Pulmão/fisiopatologia , Nociceptores/fisiologia , Reflexo/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Bradicardia/tratamento farmacológico , Bradicardia/fisiopatologia , Captopril/farmacologia , Coração/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Isotiocianatos/farmacologia , Pulmão/efeitos dos fármacos , Masculino , Nociceptores/efeitos dos fármacos , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Reflexo/efeitos dos fármacos , Taquicardia/tratamento farmacológico , Taquicardia/fisiopatologia , Nervo Vago/efeitos dos fármacos , Nervo Vago/fisiopatologia
6.
Microcirculation ; 26(1): e12506, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30281888

RESUMO

OBJECTIVE: S1P has known endothelial barrier-protective properties, but whether this extends to the BBB is unclear. We hypothesized that alcohol-induced disruption of brain microvascular endothelial barrier function and junctional protein organization can be ameliorated by S1P treatment. METHODS: Cultured primary HBMEC monolayers were used to characterize endothelial-specific mechanisms of BBB regulation. TER and apparent permeability coefficients for albumin, dextran-4 kDa, and sodium fluorescein were used as indices of barrier function. Junctional localization of Claudin-5, VE-cadherin, and ß-catenin was determined by immunofluorescence confocal microscopy. S1P was applied following treatment with alcohol. RESULTS: Alcohol significantly impaired HBMEC TER. Application of S1P after alcohol treatment resulted in a hastened recovery to the baseline HBMEC TER. Alcohol-treated HBMEC had a significantly higher mean permeability than control that was reversed by S1P. Alcohol caused the formation of gaps between cells. Treatment with S1P (after alcohol) increased junctional localization of VE-Cadherin, Claudin-5, and ß-catenin. CONCLUSIONS: Alcohol impairs the barrier function and junctional organization of HBMEC monolayers. S1P enhanced barrier function and restored junctions in the presence of alcohol, and thus may be useful for restoring BBB function during alcohol intoxication.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/irrigação sanguínea , Endotélio Vascular/química , Etanol/toxicidade , Lisofosfolipídeos/fisiologia , Proteínas Musculares/efeitos dos fármacos , Esfingosina/análogos & derivados , Antígenos CD , Barreira Hematoencefálica/patologia , Caderinas , Células Cultivadas , Claudina-5 , Endotélio Vascular/citologia , Humanos , Microcirculação , Proteínas Musculares/química , Permeabilidade/efeitos dos fármacos , Esfingosina/fisiologia , beta Catenina
7.
Shock ; 52(4): 423-433, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30339634

RESUMO

Excessive microvascular permeability is a serious complication following hemorrhagic shock and resuscitation (HSR). S1P has been shown to ameliorate microvascular leakage in a model of combined alcohol intoxication and HSR. In the current study, we tested the hypothesis that S1P reduces HSR-induced microvascular leakage by preserving endothelial cell junctional structure and the endothelial glycocalyx through the protection of mitochondrial function. We used an established in vivo rat model of conscious HSR and assessed microvascular leakage, endothelial glycocalyx integrity, and mitochondrial function by intravital microscopy. Junctional integrity in the mesenteric microcirculation was assessed by confocal microscopy. Cultured rat intestinal microvascular endothelial cells monolayers were used to test the ability of S1P to protect against glycocalyx shedding and endothelial barrier dysfunction caused by direct disruption of mitochondrial integrity due to inhibition of mitochondrial complex III. The results show that in vivo, S1P protects against HSR-induced hyperpermeability, preserves the expression of adherens junctional proteins, and protects against glycocalyx degradation. S1P treatment during HSR also protects against mitochondrial membrane depolarization. S1P also protects against mitochondrial dysfunction-induced endothelial barrier dysfunction and glycocalyx degradation by acting through mitochondrial complex III. Taken together, our data indicate that S1P protects against HSR-induced mitochondrial dysfunction in endothelial cells, which in turn improves the structure of the endothelial glycocalyx after HSR and allows for better junctional integrity to the prevention of excess microvascular permeability.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Endotélio Vascular , Lisofosfolipídeos/farmacologia , Mitocôndrias , Ressuscitação , Choque Hemorrágico , Esfingosina/análogos & derivados , Junções Aderentes/metabolismo , Junções Aderentes/patologia , Animais , Linhagem Celular , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Microvasos/metabolismo , Microvasos/patologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/patologia , Ratos , Ratos Sprague-Dawley , Choque Hemorrágico/tratamento farmacológico , Choque Hemorrágico/metabolismo , Choque Hemorrágico/patologia , Esfingosina/farmacologia
8.
Curr Top Membr ; 82: 93-140, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30360784

RESUMO

Endothelial cells of the microcirculation form a semi-permeable diffusion barrier between the blood and tissues. This permeability of the endothelium, particularly in the capillaries and postcapillary venules, is a normal physiological function needed for blood-tissue exchange in the microcirculation. During inflammation, microvascular permeability increases dramatically and can lead to tissue edema, which in turn can lead to dysfunction of tissues and organs. The molecular mechanisms that control the barrier function of endothelial cells have been under investigation for several decades and remain an important topic due to the potential for discovery of novel therapeutic strategies to reduce edema. This review highlights current knowledge of the cellular and molecular mechanisms that lead to endothelial hyperpermeability during inflammatory conditions associated with injury and disease. This includes a discussion of recent findings demonstrating temporal protrusions by endothelial cells that may contribute to intercellular junction integrity between endothelial cells and affect the diffusion distance for solutes via the paracellular pathway.


Assuntos
Permeabilidade Capilar , Endotélio Vascular/metabolismo , Animais , Humanos , Microcirculação , Microfluídica/métodos , Microvasos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Choque Hemorrágico/metabolismo , Choque Hemorrágico/patologia
9.
Methods Mol Biol ; 1717: 61-81, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29468584

RESUMO

Hypotension, cardiac depression, and elevated microvascular permeability are known problems that complicate resuscitation of patients following traumatic injury, particularly those who are also intoxicated from alcohol consumption. A conscious rat model of combined alcohol intoxication and hemorrhagic shock has been used to study the hemodynamic mechanisms involved. Here, we describe using this model to study microvascular leakage and cardiac electrical activity.


Assuntos
Intoxicação Alcoólica , Permeabilidade Capilar , Microcirculação , Ressuscitação/métodos , Choque Hemorrágico , Intoxicação Alcoólica/patologia , Intoxicação Alcoólica/fisiopatologia , Intoxicação Alcoólica/terapia , Animais , Modelos Animais de Doenças , Humanos , Ratos , Choque Hemorrágico/patologia , Choque Hemorrágico/fisiopatologia , Choque Hemorrágico/terapia
10.
Sci Rep ; 7(1): 4078, 2017 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-28642485

RESUMO

Fluid resuscitation following hemorrhagic shock is often problematic, with development of prolonged hypotension and edema. In addition, many trauma patients are also intoxicated, which generally worsens outcomes. We directly investigated how alcohol intoxication impacts hemorrhagic shock and resuscitation-induced microvascular leakage using a rat model with intravital microscopic imaging. We also tested the hypothesis that an endothelial barrier-protective bioactive lipid, sphingosine-1-phosphate (S1P), could ameliorate the microvascular leakage following alcohol intoxication plus hemorrhagic shock and resuscitation. Our results show that alcohol intoxication exacerbated hemorrhagic shock and resuscitation-induced hypotension and microvascular leakage. We next found that S1P effectively could reverse alcohol-induced endothelial barrier dysfunction using both cultured endothelial cell monolayer and in vivo models. Lastly, we observed that S1P administration ameliorated hypotension and microvascular leakage following combined alcohol intoxication and hemorrhagic shock, in a dose-related manner. These findings suggest the viability of using agonists that can improve microvascular barrier function to ameliorate trauma-induced hypotension, offering a novel therapeutic opportunity for potentially improving clinical outcomes in patients with multi-hit injuries.


Assuntos
Intoxicação Alcoólica/complicações , Intoxicação Alcoólica/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Choque Hemorrágico/etiologia , Choque Hemorrágico/metabolismo , Esfingosina/análogos & derivados , Animais , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Humanos , Hipotensão/etiologia , Hipotensão/metabolismo , Masculino , Ratos , Ressuscitação , Choque Hemorrágico/diagnóstico , Choque Hemorrágico/terapia , Esfingosina/farmacologia
11.
Sci Rep ; 7(1): 2195, 2017 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-28526859

RESUMO

An emerging challenge in tissue engineering biomimetic models is recapitulating the physiological complexity associated with real tissues. Recently, our laboratory introduced the rat mesentery culture model as an ex vivo experimental platform for investigating the multi-cellular dynamics involved in angiogenesis within an intact microvascular network using time-lapse imaging. A critical question remains whether the vessels maintain their functionality. The objective of this study was to determine whether vascular smooth muscle cells in cultured microvascular networks maintain the ability to constrict. Adult rat mesenteric tissues were harvested and cultured for three days in either MEM or MEM plus 10% serum. On Day 0 and Day 3 live microvascular networks were visualized with FITC conjugated BSI-lectin labeling and arteriole diameters were compared before and five minutes after topical exposure to vasoconstrictors (50 mM KCl and 20 nM Endothelin-1). Arterioles displayed a vasoconstriction response to KCl and endothelin for each experimental group. However, the Day 3 serum cultured networks were angiogenic, characterized by increased vessel density, and displayed a decreased vasoconstriction response compared to Day 0 networks. The results support the physiological relevance of the rat mesentery culture model as a biomimetic tool for investigating microvascular growth and function ex vivo.


Assuntos
Arteríolas/citologia , Microvasos , Modelos Biológicos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Animais , Masculino , Mesentério , Neovascularização Fisiológica , Ratos , Imagem com Lapso de Tempo , Vasoconstrição
12.
Lymphat Res Biol ; 14(2): 62-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27267167

RESUMO

BACKGROUND: Lymphatic function is critical for maintaining interstitial fluid balance and is linked to multiple pathological conditions. While smooth muscle contractile mechanisms responsible for fluid flow through collecting lymphatic vessels are well studied, how fluid flows into and through initial lymphatic networks remains poorly understood. The objective of this study was to estimate the pressure difference needed for flow through an intact initial lymphatic network. METHODS AND RESULTS: Pressure drops were computed for real and theoretical networks with varying branch orders using a segmental Poiseuille flow model. Vessel geometries per branch order were based on measurements from adult Wistar rat mesenteric initial lymphatic networks. For computational predications based on real network geometries and combinations of low or high output velocities (2 mm/s, 4 mm/s) and viscosities (1 cp, 1.5 cp), pressure drops were estimated to range 0.31-2.57 mmHg. The anatomical data for the real networks were also used to create a set of theoretical networks in order to identify possible minimum and maximum pressure drops. The pressure difference range for the theoretical networks was 0.16-3.16 mmHg. CONCLUSIONS: The results support the possibility for suction pressures generated from cyclic smooth muscle contractions of upstream collecting lymphatics being sufficient for fluid flow through an initial lymphatic network.


Assuntos
Linfa , Sistema Linfático/fisiopatologia , Vasos Linfáticos/fisiopatologia , Pressão , Algoritmos , Animais , Feminino , Sistema Linfático/metabolismo , Vasos Linfáticos/metabolismo , Modelos Biológicos , Ratos , Reologia , Viscosidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...