Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38559112

ABSTRACT

Investigating the molecular, cellular, and tissue-level changes caused by disease, and the effects of pharmacological treatments across these biological scales, necessitates the use of multiscale computational modeling in combination with experimentation. Many diseases dynamically alter the tissue microenvironment in ways that trigger microvascular network remodeling, which leads to the expansion or regression of microvessel networks. When microvessels undergo remodeling in idiopathic pulmonary fibrosis (IPF), functional gas exchange is impaired due to loss of alveolar structures and lung function declines. Here, we integrated a multiscale computational model with independent experiments to investigate how combinations of biomechanical and biochemical cues in IPF alter cell fate decisions leading to microvascular remodeling. Our computational model predicted that extracellular matrix (ECM) stiffening reduced microvessel area, which was accompanied by physical uncoupling of endothelial cell (ECs) and pericytes, the cells that comprise microvessels. Nintedanib, an FDA-approved drug for treating IPF, was predicted to further potentiate microvessel regression by decreasing the percentage of quiescent pericytes while increasing the percentage of pericytes undergoing pericyte-myofibroblast transition (PMT) in high ECM stiffnesses. Importantly, the model suggested that YAP/TAZ inhibition may overcome the deleterious effects of nintedanib by promoting EC-pericyte coupling and maintaining microvessel homeostasis. Overall, our combination of computational and experimental modeling can explain how cell decisions affect tissue changes during disease and in response to treatments.

2.
Microcirculation ; 30(5-6): e12817, 2023 08.
Article in English | MEDLINE | ID: mdl-37248193

ABSTRACT

OBJECTIVE: Microvascular remodeling is governed by biomechanical and biochemical cues which are dysregulated in idiopathic pulmonary fibrosis. Understanding how these cues impact endothelial cell-pericyte interactions necessitates a model system in which both variables can be independently and reproducibly modulated. In this study we develop a tunable hydrogel-based angiogenesis assay to study how varying angiogenic growth factors and environmental stiffness affect sprouting and vessel organization. METHODS: Lungs harvested from mice were cut into 1 mm long segments then cultured on hydrogels having one of seven possible stiffness and growth factor combinations. Time course, brightfield, and immunofluorescence imaging were used to observe and quantify sprout formation. RESULTS: Our assay was able to support angiogenesis in a comparable manner to Matrigel in soft 2 kPa gels while enabling tunability to study the effects of stiffness on sprout formation. Matrigel and 2 kPa groups contained significantly more samples with sprouts when compared to the stiffer 10 and 20 kPa gels. Growth factor treatment did not have as obvious an effect, although the 20 kPa PDGF + FGF-treated group had significantly longer vessels than the vascular endothelial growth factor-treated group. CONCLUSIONS: We have developed a novel, tunable hydrogel assay for the creation of lung explant vessel organoids which can be modulated to study the impact of specific environmental cues on vessel formation and maturation.


Subject(s)
Idiopathic Pulmonary Fibrosis , Vascular Endothelial Growth Factor A , Mice , Animals , Vascular Endothelial Growth Factor A/pharmacology , Pericytes , Hydrogels/pharmacology , Neovascularization, Physiologic
3.
Biomed Opt Express ; 13(5): 2695-2706, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35774317

ABSTRACT

Cutaneous wounds affect millions of people every year. Vascularization and blood oxygen delivery are critical bottlenecks in wound healing, and understanding the spatiotemporal dynamics of these processes may lead to more effective therapeutic strategies to accelerate wound healing. In this work, we applied multi-parametric photoacoustic microscopy (PAM) to study vascular adaptation and the associated changes in blood oxygen delivery and tissue oxygen metabolism throughout the hemostasis, inflammatory, proliferation, and early remodeling phases of wound healing in mice with skin puncture wounds. Multifaceted changes in the vascular structure, function, and tissue oxygen metabolism were observed during the 14-day monitoring of wound healing. On the entire wound area, significant elevations of the arterial blood flow and tissue oxygen metabolism were observed right after wounding and remained well above the baseline over the 14-day period. On the healing front, biphasic changes in the vascular density and blood flow were observed, both of which peaked on day 1, remained elevated in the first week, and returned to the baselines by day 14. Along with the wound closure and thickening, tissue oxygen metabolism in the healing front remained elevated even after structural and functional changes in the vasculature were stabilized. On the newly formed tissue, significantly higher blood oxygenation, flow, and tissue metabolism were observed compared to those before wounding. Blood oxygenation and flow in the new tissue appeared to be independent of when it was formed, but instead showed noticeable dependence on the phase of wound healing. This PAM study provides new insights into the structural, functional, and metabolic changes associated with vascular adaptation during wound healing and suggests that the timing and target of vascular treatments for wound healing may affect the outcomes.

4.
Ann Plast Surg ; 88(5 Suppl 5): S466-S472, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35502953

ABSTRACT

BACKGROUND: Acellular dermal matrix (ADM) supported implant-based reconstruction remains the most commonly performed mode of reconstruction after breast cancer. Acellular dermal matrix clinical usage has reported benefits but requires rapid and efficient vascular and cellular incorporation into the recipient to have the best outcomes. Orderly transition from M1 to M2 macrophage phenotypic profile, coordinated in part by interleukin 4 (IL-4), is an important component of vascular stabilization and remodeling. Using the ADM substrate as a delivery device for immunomodulation of macrophage phenotype holds the potential to improve integration. METHODS: Interleukin 4 was adsorbed onto ADM samples and drug elution curves were measured. Next, experimental groups of 8 C57BL/6 mice had 5-mm ADM discs surgically placed in a dorsal window chamber with a vascularized skin flap on one side and a plastic cover slip on the other in a model of implant-based breast reconstruction. Group 1 consisted of IL-4 (5 µg) adsorbed into the ADM preoperatively and group 2 consisted of an untreated ADM control. Serial gross examinations were performed with histology at day 21 for markers of vascularization, mesenchymal cell infiltration, and macrophage lineage. RESULTS: Drug elution curves showed sustained IL-4 release for 10 days after adsorption. Serial gross examination showed similar rates of superficial vascular investment of the ADM beginning at the periphery by day 14 and increasing through day 21. Interleukin-4 treatment led to significantly increased CD31 staining of vascular endothelial cells within the ADM over the control group (P < 0.05) at 21 days. Although vimentin staining did not indicate a significant increase in fibroblasts overall, IL-4 did result in a significant increase in expression of α-smooth muscle actin. The expression of macrophage phenotype markers Arginase1 and iNOS present within the ADM were not significantly affected by IL-4 treatment at the day 21 time point. CONCLUSIONS: Acellular dermal matrix has the potential to be used for immunomodulatory cytokine delivery during the timeframe of healing. Using implanted ADM as a delivery vehicle to drive IL-4 mediated angiogenesis and vascular remodeling significantly enhanced vascularity within the ADM substrate.


Subject(s)
Acellular Dermis , Interleukin-4 , Acellular Dermis/drug effects , Animals , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Immunomodulation , Interleukin-4/immunology , Interleukin-4/pharmacokinetics , Interleukin-4/pharmacology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Vascular Remodeling
5.
Cell Mol Bioeng ; 14(4): 321-338, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34290839

ABSTRACT

INTRODUCTION: Pharmacologic approaches for promoting angiogenesis have been utilized to accelerate healing of chronic wounds in diabetic patients with varying degrees of success. We hypothesize that the distribution of proangiogenic drugs in the wound area critically impacts the rate of closure of diabetic wounds. To evaluate this hypothesis, we developed a mathematical model that predicts how spatial distribution of VEGF-A produced by delivery of a modified mRNA (AZD8601) accelerates diabetic wound healing. METHODS: We modified a previously published model of cutaneous wound healing based on coupled partial differential equations that describe the density of sprouting capillary tips, chemoattractant concentration, and density of blood vessels in a circular wound. Key model parameters identified by a sensitivity analysis were fit to data obtained from an in vivo wound healing study performed in the dorsum of diabetic mice, and a pharmacokinetic model was used to simulate mRNA and VEGF-A distribution following injections with AZD8601. Due to the limited availability of data regarding the spatial distribution of AZD8601 in the wound bed, we performed simulations with perturbations to the location of injections and diffusion coefficient of mRNA to understand the impact of these spatial parameters on wound healing. RESULTS: When simulating injections delivered at the wound border, the model predicted that injections delivered on day 0 were more effective in accelerating wound healing than injections delivered at later time points. When the location of the injection was varied throughout the wound space, the model predicted that healing could be accelerated by delivering injections a distance of 1-2 mm inside the wound bed when compared to injections delivered on the same day at the wound border. Perturbations to the diffusivity of mRNA predicted that restricting diffusion of mRNA delayed wound healing by creating an accumulation of VEGF-A at the wound border. Alternatively, a high mRNA diffusivity had no effect on wound healing compared to a simulation with vehicle injection due to the rapid loss of mRNA at the wound border to surrounding tissue. CONCLUSIONS: These findings highlight the critical need to consider the location of drug delivery and diffusivity of the drug, parameters not typically explored in pre-clinical experiments, when designing and testing drugs for treating diabetic wounds. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s12195-021-00678-9.

6.
Sci Rep ; 10(1): 15808, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32978500

ABSTRACT

Retinal diseases are frequently characterized by the accumulation of excessive scar tissue found throughout the neural retina. However, the pathophysiology of retinal fibrosis remains poorly understood, and the cell types that contribute to the fibrotic response are incompletely defined. Here, we show that myofibroblast differentiation of mural cells contributes directly to retinal fibrosis. Using lineage tracing technology, we demonstrate that after chemical ocular injury, Myh11+ mural cells detach from the retinal microvasculature and differentiate into myofibroblasts to form an epiretinal membrane. Inhibition of TGFßR attenuates Myh11+ retinal mural cell myofibroblast differentiation, and diminishes the subsequent formation of scar tissue on the surface of the retina. We demonstrate retinal fibrosis within a murine model of oxygen-induced retinopathy resulting from the intravitreal injection of adipose Myh11-derived mesenchymal stem cells, with ensuing myofibroblast differentiation. In this model, inhibiting TGFßR signaling does not significantly alter myofibroblast differentiation and collagen secretion within the retina. This work shows the complexity of retinal fibrosis, where scar formation is regulated both by TGFßR and non-TGFßR dependent processes involving mural cells and derived mesenchymal stem cells. It also offers a cautionary note on the potential deleterious, pro-fibrotic effects of exogenous MSCs once intravitreally injected into clinical patients.


Subject(s)
Cell Differentiation , Cicatrix/pathology , Fibrosis/pathology , Mesenchymal Stem Cells/pathology , Myofibroblasts/pathology , Myosin Heavy Chains/metabolism , Retinal Diseases/pathology , Animals , Cells, Cultured , Cicatrix/metabolism , Female , Fibrosis/metabolism , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Myofibroblasts/metabolism , Retinal Diseases/metabolism , Signal Transduction
7.
CPT Pharmacometrics Syst Pharmacol ; 9(7): 384-394, 2020 07.
Article in English | MEDLINE | ID: mdl-32438492

ABSTRACT

Intradermal delivery of AZD8601, an mRNA designed to produce vascular endothelial growth factor A (VEGF-A), has previously been shown to accelerate cutaneous wound healing in a murine diabetic model. Here, we develop population pharmacokinetic and pharmacodynamic models aiming to quantify the effect of AZD8601 injections on the dynamics of wound healing. A dataset of 584 open wound area measurements from 131 mice was integrated from 3 independent studies encompassing different doses, dosing timepoints, and number of doses. Evaluation of several candidate models showed that wound healing acceleration is not likely driven directly by time-dependent VEGF-A concentration. Instead, we found that administration of AZD8601 induced a sustained acceleration of wound healing depending on the accumulated dose, with a dose producing 50% of the maximal effect of 92 µg. Simulations with this model showed that a single dose of 200 µg AZD8601 can reduce the time to reach 50% wound healing by up to 5 days.


Subject(s)
Diabetes Mellitus, Experimental/therapy , RNA, Messenger/administration & dosage , Vascular Endothelial Growth Factor A/genetics , Wound Healing/genetics , Animals , Diabetes Mellitus, Experimental/complications , Mice , Models, Biological , RNA, Messenger/genetics , Time Factors
8.
Microcirculation ; 27(3): e12598, 2020 04.
Article in English | MEDLINE | ID: mdl-31660674

ABSTRACT

OBJECTIVE: Arteriogenesis is an important mechanism that contributes to restoration of oxygen supply in chronically ischemic tissues, but remains incompletely understood due to technical limitations. This study presents a novel approach for comprehensive assessment of the remodeling pattern in a complex microvascular network containing multiple collateral microvessels. METHODS: We have developed a hardware-software integrated platform for quantitative, longitudinal, and label-free imaging of network-wide hemodynamic changes and arteriogenesis at the single-vessel level. By ligating feeding arteries in the mouse ear, we induced network-wide hemodynamic redistribution and localized arteriogenesis. The utility of this technology was demonstrated by studying the influence of obesity on microvascular arteriogenesis. RESULTS: Simultaneously monitoring the remodeling of competing collateral arterioles revealed a new, inverse relationship between initial vascular resistance and extent of arteriogenesis. Obese mice exhibited similar remodeling responses to lean mice through the first week, including diameter increase and flow upregulation in collateral arterioles. However, these gains were subsequently lost in obese mice. CONCLUSIONS: Capable of label-free, comprehensive, and dynamic quantification of structural and functional changes in the microvascular network in vivo, this platform opens up new opportunities to study the mechanisms of microvascular arteriogenesis, its implications in diseases, and approaches to pharmacologically rectify microvascular dysfunction.


Subject(s)
Angiography , Collateral Circulation , Hemodynamics , Ischemia , Neovascularization, Physiologic , Animals , Arterioles/diagnostic imaging , Arterioles/physiopathology , Female , Ischemia/diagnostic imaging , Ischemia/physiopathology , Mice , Mice, Transgenic
9.
Sci Rep ; 8(1): 17509, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504800

ABSTRACT

Capable of mediating efficient transfection and protein production without eliciting innate immune responses, chemically modified mRNA holds great potential to produce paracrine factors at a physiologically beneficial level, in a spatiotemporally controlled manner, and with low toxicity. Although highly promising in cardiovascular medicine and wound healing, effects of this emerging therapeutic on the microvasculature and its bioactivity in disease settings remain poorly understood. Here, we longitudinally and comprehensively characterize microvascular responses to AZD8601, a modified mRNA encoding vascular endothelial growth factor A (VEGF-A), in vivo. Using multi-parametric photoacoustic microscopy, we show that intradermal injection of AZD8601 formulated in a biocompatible vehicle results in pronounced, sustained and dose-dependent vasodilation, blood flow upregulation, and neovessel formation, in striking contrast to those induced by recombinant human VEGF-A protein, a non-translatable variant of AZD8601, and citrate/saline vehicle. Moreover, we evaluate the bioactivity of AZD8601 in a mouse model of diabetic wound healing in vivo. Using a boron nanoparticle-based tissue oxygen sensor, we show that sequential dosing of AZD8601 improves vascularization and tissue oxygenation of the wound bed, leading to accelerated re-epithelialization during the early phase of diabetic wound healing.


Subject(s)
Diabetic Angiopathies/etiology , Diabetic Angiopathies/pathology , Microvessels/metabolism , RNA, Messenger/genetics , Vascular Endothelial Growth Factor A/genetics , Wound Healing/genetics , Animals , Diabetic Angiopathies/diagnostic imaging , Disease Models, Animal , Humans , Mice , Microvessels/drug effects , Myocytes, Smooth Muscle/metabolism , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/genetics , Oxygen Consumption , Time-Lapse Imaging , Wound Healing/drug effects
10.
Sci Rep ; 7(1): 9049, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28831080

ABSTRACT

Microvascular endothelial cell heterogeneity and its relationship to hemodynamics remains poorly understood due to a lack of sufficient methods to examine these parameters in vivo at high resolution throughout an angiogenic network. The availability of surrogate markers for functional vascular proteins, such as green fluorescent protein, enables expression in individual cells to be followed over time using confocal microscopy, while photoacoustic microscopy enables dynamic measurement of blood flow across the network with capillary-level resolution. We combined these two non-invasive imaging modalities in order to spatially and temporally analyze biochemical and biomechanical drivers of angiogenesis in murine corneal neovessels. By stimulating corneal angiogenesis with an alkali burn in Tie2-GFP fluorescent-reporter mice, we evaluated how onset of blood flow and surgically-altered blood flow affects Tie2-GFP expression. Our study establishes a novel platform for analyzing heterogeneous blood flow and fluorescent reporter protein expression across a dynamic microvascular network in an adult mammal.


Subject(s)
Capillaries/physiology , Endothelium, Vascular/metabolism , Gene Expression , Microcirculation , Receptor, TIE-2/genetics , Regional Blood Flow/genetics , Vascular Remodeling/genetics , Animals , Biomarkers , Corneal Neovascularization/genetics , Corneal Neovascularization/metabolism , Endothelial Cells/metabolism , Genes, Reporter , Hemodynamics , Mice , Microscopy, Fluorescence , Molecular Imaging
11.
Sci Rep ; 7(1): 447, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28348370

ABSTRACT

Successful tissue repair requires the activities of myeloid cells such as monocytes and macrophages that guide the progression of inflammation and healing outcome. Immunoregenerative materials leverage the function of endogenous immune cells to orchestrate complex mechanisms of repair; however, a deeper understanding of innate immune cell function in inflamed tissues and their subsequent interactions with implanted materials is necessary to guide the design of these materials. Blood monocytes exist in two primary subpopulations, characterized as classical inflammatory or non-classical. While classical monocytes extravasate into inflamed tissue and give rise to macrophages or dendritic cells, the recruitment kinetics and functional role of non-classical monocytes remains unclear. Here, we demonstrate that circulating non-classical monocytes are directly recruited to polymer films within skin injuries, where they home to a perivascular niche and generate alternatively activated, wound healing macrophages. Selective labeling of blood monocyte subsets indicates that non-classical monocytes are biased progenitors of alternatively activated macrophages. On-site delivery of the immunomodulatory small molecule FTY720 recruits S1PR3-expressing non-classical monocytes that support vascular remodeling after injury. These results elucidate a previously unknown role for blood-derived non-classical monocytes as contributors to alternatively activated macrophages, highlighting them as key regulators of inflammatory response and regenerative outcome.


Subject(s)
Macrophages/pathology , Monocytes/pathology , Soft Tissue Injuries/pathology , Stem Cells/pathology , Wound Healing , Adoptive Transfer , Animals , Antigens, CD/metabolism , Arterioles/drug effects , Arterioles/metabolism , Biocompatible Materials/pharmacology , Cell Differentiation/drug effects , Fingolimod Hydrochloride/pharmacology , Implants, Experimental , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Monocytes/drug effects , Monocytes/metabolism , Skin/blood supply , Skin/pathology , Wound Healing/drug effects
12.
J Forensic Sci ; 61(6): 1493-1497, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27500887

ABSTRACT

Objective documentation of tears and abrasions to the external genitalia after sexual assault is an important part of the forensic examination. A 1% aqueous solution of toluidine blue (TB) dye is often used to highlight injury, but the dye can be difficult to see on dark skin. We evaluated a fluorescent dye for detecting injuries on all skin colors in a relevant preclinical murine model. We compared the ability of trained, blinded observers to detect unstained wounds and wounds stained with 1% fluorescein sodium (FL) and with TB. We also evaluated the cutaneous wound healing response after application of both dyes. The no-dye group had significantly fewer (p < 0.05) true-positive identifications compared to all the dye groups. No differences in effectiveness were detected between dye treatments. All groups exhibited statistically similar healing rates. These findings support the evaluation of fluorescein sodium in a future clinical study with human subjects.


Subject(s)
Fluorescent Dyes , Rape , Sex Offenses , Animals , Disease Models, Animal , Humans , Mice , Tolonium Chloride
13.
Stem Cells Transl Med ; 4(5): 459-67, 2015 May.
Article in English | MEDLINE | ID: mdl-25769654

ABSTRACT

Diabetic retinopathy is characterized by progressive vascular dropout with subsequent vision loss. We have recently shown that an intravitreal injection of adipose-derived stem cells (ASCs) can stabilize the retinal microvasculature, enabling repair and regeneration of damaged capillary beds in vivo. Because an understanding of ASC status from healthy versus diseased donors will be important as autologous cellular therapies are developed for unmet clinical needs, we took advantage of the hyperglycemic Akimba mouse as a preclinical in vivo model of diabetic retinopathy in an effort aimed at evaluating therapeutic efficacy of adipose-derived stem cells (mASCs) derived either from healthy, nondiabetic or from diabetic mice. To these ends, Akimba mice received intravitreal injections of media conditioned by mASCs or mASCs themselves, subsequent to development of substantial retinal capillary dropout. mASCs from healthy mice were more effective than diabetic mASCs in protecting the diabetic retina from further vascular dropout. Engrafted ASCs were found to preferentially associate with the retinal vasculature. Conditioned medium was unable to recapitulate the vasoprotection seen with injected ASCs. In vitro diabetic ASCs showed decreased proliferation and increased apoptosis compared with healthy mASCs. Diabetic ASCs also secreted less vasoprotective factors than healthy mASCs, as determined by high-throughput enzyme-linked immunosorbent assay. Our findings suggest that diabetic ASCs are functionally impaired compared with healthy ASCs and support the utility of an allogeneic injection of ASCs versus autologous or conditioned media approaches in the treatment of diabetic retinopathy.


Subject(s)
Cell- and Tissue-Based Therapy , Diabetes Mellitus, Experimental/therapy , Diabetic Retinopathy/therapy , Stem Cell Transplantation , Adipocytes/cytology , Animals , Culture Media, Conditioned , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/pathology , Disease Models, Animal , Mice , Stem Cells/cytology
14.
Arterioscler Thromb Vasc Biol ; 34(9): 2012-22, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24969773

ABSTRACT

OBJECTIVE: Chronic arterial occlusion results in arteriogenesis of collateral blood vessels. This process has been shown to be dependent on the recruitment of growth-promoting macrophages to remodeling collaterals. However, the potential role of venules in monocyte recruitment during microvascular arteriogenesis is not well demonstrated. First, we aim to document that arteriogenesis occurs in the mouse spinotrapezius ligation model. Then, we investigate the temporal and spatial distribution, as well as proliferation, of monocytes/macrophages recruited to collateral arterioles in response to elevated fluid shear stress. APPROACH AND RESULTS: Laser speckle flowmetry confirmed a postligation increase in blood velocity within collateral arterioles but not within venules. After 72 hours post ligation, collateral arteriole diameters were increased, proliferating cells were identified in vessel walls of shear-activated collaterals, and perivascular CD206(+) macrophages demonstrated proliferation. A 5-ethynyl-2'-deoxyuridine assay identified proliferation. CD68(+)CD206(+) cells around collaterals were increased 96%, whereas CX3CR1((+/GFP)) cells were increased 126% in ligated versus sham groups after 72 hours. CX3CR1((+/GFP)) cells were predominately venule associated at 6 hours after ligation; and CX3CR1((+/GFP hi)) cells shifted from venule to arteriole associated between 6 and 72 hours after surgery exclusively in ligated muscle. We report accumulation and extravasation of adhered CX3CR1((+/GFP)) cells in and from venules, but not from arterioles, after ligation. CONCLUSIONS: Our results demonstrate that arteriogenesis occurs in the murine spinotrapezius ligation model and implicate postcapillary venules as the site of tissue entry for circulating monocytes. Local proliferation of macrophages is also documented. These data open up questions about the role of arteriole-venule communication during monocyte recruitment.


Subject(s)
Ischemia/physiopathology , Monocytes/physiology , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/physiology , Venules/pathology , Animals , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Arterioles , CX3C Chemokine Receptor 1 , Cell Division , Endothelium, Vascular/pathology , Female , Genes, Reporter , Hemorheology , Laser-Doppler Flowmetry , Lectins, C-Type/analysis , Ligation , Male , Mannose Receptor , Mannose-Binding Lectins/analysis , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Receptors, Cell Surface/analysis , Receptors, Chemokine/analysis , Receptors, Chemokine/genetics
15.
Microcirculation ; 19(2): 143-54, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21954923

ABSTRACT

OBJECTIVE: We examined the effects of exogenously delivered thrombin on cell recruitment in skeletal muscle and the formation of new collateral arterioles in the microvasculature in response to ligation-induced ischemia. METHODS: Thrombin or vehicle was locally applied to both ligated and nonoperated Balb/c spinotrapezius muscles, which were harvested after three or seven days, imaged using confocal microscopy, and analyzed. RESULTS: Thrombin treatment resulted in accelerated arterialization of collateral capillaries and accelerated tissue reperfusion in ischemic muscles. Uninjured muscle treated with thrombin displayed increased vascular cell adhesion molecule 1 expression on arteriole and venule endothelium, increased expression of smooth muscle α-actin on capillary-sized vessels, increased infiltration by CD11b(+) leukocytes, and mast cell infiltration and degranulation. CONCLUSIONS: Exogenous delivery of thrombin enhances microvascular collateral development in response to ischemic insult, and accelerates tissue reperfusion. Elicited responses from multiple cell types probably contribute to these effects.


Subject(s)
Hemostatics/pharmacology , Ischemia/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Thrombin/pharmacology , Actins/metabolism , Animals , Arterioles/metabolism , Arterioles/pathology , Arterioles/physiopathology , Capillaries/metabolism , Capillaries/pathology , Capillaries/physiopathology , Cattle , Cell Degranulation , Ischemia/pathology , Ischemia/physiopathology , Leukocytes/metabolism , Leukocytes/pathology , Male , Mast Cells/metabolism , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Reperfusion
16.
Microcirculation ; 18(3): 163-71, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21418375

ABSTRACT

OBJECTIVES: Vascular obstructive events can be partially compensated for by remodeling processes that increase vessel diameter and collateral tortuosity. However, methods for visualizing remodeling events in vivo and with temporal comparisons from the same animal remain elusive. METHODS: Using a novel infrared conjugated polyethylene glycol dye, we investigated the possibility of intravital vascular imaging of the mouse ear before and after ligation of the primary feeder artery. For comparison, we used two different mouse models known to have impaired vascular remodeling after ligation (i.e., aged and PAI-1(-/-) mice). The results obtained with the infrared dye were confirmed using immunofluorescence labeling of the ear microvasculature with confocal microscopy. RESULTS: After ligation, increases in vessel diameter (between 10% and 60%) and tortuosity (approximately 15%) were observed in C57Bl/6 mice using both the infrared dye and the immunofluorescence technique. However, aged C57Bl/6 and PAI-1(-/-) mice did not show vascular remodeling following ligation. CONCLUSIONS: Vascular remodeling can be visualized and accurately quantified using a new infrared dye in vivo. This analysis technique could be generally employed for quantitative investigations of changes in vascular remodeling.


Subject(s)
Arteries/pathology , Coloring Agents , Dilatation, Pathologic/pathology , Molecular Probes , Animals , Dilatation, Pathologic/diagnosis , Disease Models, Animal , Ligation , Methods , Mice , Molecular Probes/chemistry
17.
Arterioscler Thromb Vasc Biol ; 30(4): 809-17, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20110576

ABSTRACT

OBJECTIVE: Activated endothelium and increased monocyte-endothelial interactions in the vessel wall are key early events in atherogenesis. ATP binding cassette (ABC) transporters play important roles in regulating sterol homeostasis in many cell types. Endothelial cells (EC) have a high capacity to efflux sterols and express the ABC transporter, ABCG1. Here, we define the role of ABCG1 in the regulation of lipid homeostasis and inflammation in aortic EC. METHODS AND RESULTS: Using EC isolated from ABCG1-deficient mice (ABCG1 KO), we observed reduced cholesterol efflux to high-density lipoprotein compared to C57BL/6 (B6) EC. However, total cholesteryl ester levels were not changed in ABCG1 KO EC. Secretions of KC, MCP-1, and IL-6 by ABCG1 KO EC were significantly increased, and surface expressions of intercellular adhesion molecule-1 and E-selectin were increased several-fold on ABCG1 KO EC. Concomitant with these findings, we observed a 4-fold increase in monocyte adhesion to the intact aortic endothelium of ABCG1 KO mice ex vivo and to isolated aortic EC from these mice in vitro. In a gain-of-function study in vitro, restoration of ABCG1 expression in ABCG1 KO EC reduced monocyte-endothelial interactions. Utilizing pharmacological inhibitors for STAT3 and the IL-6 receptor, we found that blockade of STAT3 and IL-6 receptor signaling in ABCG1 KO EC completely abrogated monocyte adhesion to ABCG1 KO endothelium. CONCLUSIONS: ABCG1 deficiency in aortic endothelial cells activates endothelial IL-6-IL-6 receptor-STAT3 signaling, thereby increasing monocyte-endothelial interactions and vascular inflammation.


Subject(s)
Cell Adhesion , Endothelial Cells/metabolism , Inflammation/metabolism , Lipoproteins/deficiency , Monocytes/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , Animals , Cell Adhesion/drug effects , Cells, Cultured , Chemokine CCL2/metabolism , Chemokine CXCL1/metabolism , Chemotaxis , Cholesterol/metabolism , Cholesterol, HDL/metabolism , E-Selectin/metabolism , Endothelial Cells/drug effects , Endothelial Cells/immunology , Inflammation/immunology , Inflammation Mediators/metabolism , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Lipoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Monocytes/immunology , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction , Transfection , Tumor Necrosis Factor-alpha/metabolism
18.
Arterioscler Thromb Vasc Biol ; 28(11): 2003-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18688018

ABSTRACT

OBJECTIVE: Platelet endothelial cell adhesion molecule-1 (PECAM-1, CD31) has recently been shown to form an essential element of a mechanosensory complex that mediates endothelial responses to fluid shear stress. The aim of this study was to determine the in vivo role of PECAM-1 in atherosclerosis. METHODS AND RESULTS: We crossed C57BL/6 Pecam1(-/-) mice with apolipoprotein E-deficient (Apoe(-/-)) mice. On a Western diet, Pecam1(-/-)Apoe(-/-) mice showed reduced atherosclerotic lesion size compared to Apoe(-/-) mice. Striking differences were observed in the lesser curvature of the aortic arch, an area of disturbed flow, but not in the descending thoracic or abdominal aorta. Vascular cell adhesion molecule-1 (VCAM-1) expression, macrophage infiltration, and endothelial nuclear NF-kappaB were all reduced in Pecam1(-/-)Apoe(-/-) mice. Bone marrow transplantation suggested that endothelial PECAM-1 is the main determinant of atherosclerosis in the aortic arch, but that hematopoietic PECAM-1 promotes lesions in the abdominal aorta. In vitro data show that siRNA-based knockdown of PECAM-1 attenuates endothelial NF-kappaB activity and VCAM-1 expression under conditions of atheroprone flow. CONCLUSIONS: These results indicate that endothelial PECAM-1 contributes to atherosclerotic lesion formation in regions of disturbed flow by regulating NF-kappaB-mediated gene expression.


Subject(s)
Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Endothelial Cells/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cells, Cultured , Dietary Fats , Disease Models, Animal , Disease Progression , Endothelial Cells/pathology , Humans , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , RNA Interference , RNA, Small Interfering/metabolism , Regional Blood Flow , Stress, Mechanical , Vascular Cell Adhesion Molecule-1/metabolism
19.
J Immunol ; 181(2): 1357-64, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606690

ABSTRACT

IL-23 and IL-17A regulate granulopoiesis through G-CSF, the main granulopoietic cytokine. IL-23 is secreted by activated macrophages and dendritic cells and promotes the expansion of three subsets of IL-17A-expressing neutrophil-regulatory T (Tn) cells; CD4(-)CD8(-)alphabeta(low), CD4(+)CD8(-)alphabeta(+) (Th17), and gammadelta(+) T cells. In this study, we investigate the effects of IL-17A on circulating neutrophil levels using IL-17R-deficient (Il17ra(-/-)) mice and Il17ra(-/-)Itgb2(-/-) mice that lack both IL-17R and all four beta(2) integrins. IL-17R deficiency conferred a reduction in neutrophil numbers and G-CSF levels, as did Ab blockade against IL-17A in wild-type mice. Bone marrow transplantation revealed that IL-17R expression on nonhemopoietic cells had the greatest effects on regulating blood neutrophil counts. Although circulating neutrophil numbers were reduced, IL-17A expression, secretion, and the number of IL-17A-producing Tn cells were elevated in Il17ra(-/-) and Il17ra(-/-)Itgb2(-/-) mice, suggesting a negative feedback effect through IL-17R. The negative regulation of IL-17A-producing T cells and IL-17A and IL-17F gene expression through the interactions of IL-17A or IL-17F with IL-17R was confirmed in splenocyte cultures in vitro. We conclude that IL-17A regulates blood neutrophil counts by inducing G-CSF production mainly in nonhemopoietic cells. IL-17A controls the expansion of IL-17A-producing Tn cell populations through IL-17R.


Subject(s)
Granulocyte Colony-Stimulating Factor/metabolism , Interleukin-17/metabolism , Interleukin-23/metabolism , Neutrophils/physiology , Receptors, Interleukin-17/metabolism , T-Lymphocyte Subsets/immunology , Animals , Cell Polarity , Dendritic Cells/immunology , Dendritic Cells/metabolism , Granulocyte Colony-Stimulating Factor/immunology , Interleukin-17/immunology , Interleukin-23/immunology , Leukocyte Count , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Receptors, Interleukin-17/deficiency , Receptors, Interleukin-17/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism
20.
J Immunol ; 179(12): 8274-9, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18056371

ABSTRACT

IL-23 is secreted by macrophages and dendritic cells in response to microbial products and inflammatory cytokines. IL-23 is a heterodimer composed of the unique IL-23p19 subunit linked to the common p40 subunit that it shares with IL-12. IL-23 is implicated in autoimmune diseases, where it supports the expansion of IL-17A-producing CD4+ Th17 cells. IL-23 also regulates granulopoiesis in a neutrostat regulatory feedback loop through IL-17A-producing neutrophil regulatory (Tn) cells, most of which express gammadelta TCR. This homeostatic system is disrupted in mice lacking adhesion molecules like beta2-integrins (Itgb2-/-) which have defective neutrophil trafficking and neutrophilia. To test the role of IL-23 in the homeostatic regulation of circulating neutrophil numbers, we measured blood neutrophil numbers in p40-deficient (IL12b-/-) mice and found them reduced compared with wild-type mice. IL12b-/-Itgb2-/- mice, lacking beta2-integrins, IL-12, and IL-23 showed significantly blunted neutrophilia compared with Itgb2-/- mice. Treatment of both IL12b-/- and IL12b-/-Itgb2-/- mice with IL-23, but not IL-12, restored circulating neutrophil counts. Serum levels of IL-17A were readily detectable in Itgb2-/- mice, but not in IL12b-/-Itgb2-/- mice, suggesting that IL-17A production is reduced when IL-23 is absent. Similarly, tissue mRNA expression of IL-17A was reduced in IL12b-/-Itgb2-/-mice compared with Itgb2-/- controls. The total number of CD3+ IL-17A-producing Tn cells were significantly reduced in the spleen and lamina propria of IL12b-/-Itgb2-/- mice, with the largest reduction found in gammadelta+ T cells. Our results suggest a prominent role of IL-23 in the regulation of granulopoiesis and the prevalence of IL-17A-producing Tn cells.


Subject(s)
Interleukin-12 Subunit p40/physiology , Interleukin-23/physiology , Neutropenia/genetics , Neutrophils/immunology , Animals , CD18 Antigens/genetics , Homeostasis/immunology , Interleukin-12 Subunit p40/genetics , Interleukin-17/blood , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-23/genetics , Mice , Mice, Mutant Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...