Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Circulation ; 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38873770

ABSTRACT

BACKGROUND: Endothelial cell (EC) apoptosis and proliferation of apoptosis-resistant cells is a hallmark of pulmonary hypertension (PH). Yet, why some ECs die and others proliferate and how this contributes to vascular remodeling is unclear. We hypothesized that this differential response may: (1) relate to different EC subsets, namely pulmonary artery (PAECs) versus microvascular ECs (MVECs); (2) be attributable to autophagic activation in both EC subtypes; and (3) cause replacement of MVECs by PAECs with subsequent distal vessel muscularization. METHODS: EC subset responses to chronic hypoxia were assessed by single-cell RNA sequencing of murine lungs. Proliferative versus apoptotic responses, activation, and role of autophagy were assessed in human and rat PAECs and MVECs, and in precision-cut lung slices of wild-type mice or mice with endothelial deficiency in the autophagy gene Atg7 (Atg7EN-KO). Abundance of PAECs versus MVECs in precapillary microvessels was assessed in lung tissue from patients with PH and animal models on the basis of structural or surface markers. RESULTS: In vitro and in vivo, PAECs proliferated in response to hypoxia, whereas MVECs underwent apoptosis. Single-cell RNA sequencing analyses support these findings in that hypoxia induced an antiapoptotic, proliferative phenotype in arterial ECs, whereas capillary ECs showed a propensity for cell death. These distinct responses were prevented in hypoxic Atg7EN-KO mice or after ATG7 silencing, yet replicated by autophagy stimulation. In lung tissue from mice, rats, or patients with PH, the abundance of PAECs in precapillary arterioles was increased, and that of MVECs reduced relative to controls, indicating replacement of microvascular by macrovascular ECs. EC replacement was prevented by genetic or pharmacological inhibition of autophagy in vivo. Conditioned medium from hypoxic PAECs yet not MVECs promoted pulmonary artery smooth muscle cell proliferation and migration in a platelet-derived growth factor-dependent manner. Autophagy inhibition attenuated PH development and distal vessel muscularization in preclinical models. CONCLUSIONS: Autophagic activation by hypoxia induces in parallel PAEC proliferation and MVEC apoptosis. These differential responses cause a progressive replacement of MVECs by PAECs in precapillary pulmonary arterioles, thus providing a macrovascular context that in turn promotes pulmonary artery smooth muscle cell proliferation and migration, ultimately driving distal vessel muscularization and the development of PH.

2.
FASEB J ; 35(9): e21831, 2021 09.
Article in English | MEDLINE | ID: mdl-34383982

ABSTRACT

The nuclear factor of activated T-cells 5 (NFAT5) is a transcriptional regulator of macrophage activation and T-cell development, which controls stabilizing responses of cells to hypertonic and biomechanical stress. In this study, we detected NFAT5 in the media layer of arteries adjacent to human arteriosclerotic plaques and analyzed its role in vascular smooth muscle cells (VSMCs) known to contribute to arteriosclerosis through the uptake of lipids and transformation into foam cells. Exposure of both human and mouse VSMCs to cholesterol stimulated the nuclear translocation of NFAT5 and increased the expression of the ATP-binding cassette transporter Abca1, required to regulate cholesterol efflux from cells. Loss of Nfat5 promoted cholesterol accumulation in these cells and inhibited the expression of genes involved in the management of oxidative stress or lipid handling, such as Sod1, Plin2, Fabp3, and Ppard. The functional relevance of these observations was subsequently investigated in mice fed a high-fat diet upon induction of a smooth muscle cell-specific genetic ablation of Nfat5 (Nfat5(SMC)-/- ). Under these conditions, Nfat5(SMC)-/- but not Nfat5fl/fl mice developed small, focal lipid-rich lesions in the aorta after 14 and 25 weeks, which were formed by intracellular lipid droplets deposited in the sub-intimal VSMCs layer. While known for being activated by external stimuli, NFAT5 was found to mediate the expression of VSMC genes associated with the handling of lipids in response to a cholesterol-rich environment. Failure of this protective function may promote the formation of lipid-laden arterial VSMCs and pro-atherogenic vascular responses.


Subject(s)
Aorta/metabolism , Lipid Metabolism/physiology , Lipids/physiology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Transcription Factors/metabolism , ATP Binding Cassette Transporter 1/metabolism , Aged , Animals , Atherosclerosis/metabolism , Cells, Cultured , Cholesterol/metabolism , Female , Foam Cells/metabolism , Gene Expression Regulation/physiology , Humans , Hypercholesterolemia/metabolism , Male , Mice , Middle Aged , Oxidative Stress/physiology , Tunica Intima/metabolism
3.
Exp Cell Res ; 399(2): 112446, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33422461

ABSTRACT

Communication of vascular cells is essential for the control of organotypic functions of blood vessels. In this context, vascular endothelial cells (EC) act as potent regulators of vascular smooth muscle cell (VSMC) functions such as contraction and relaxation. However, the impact of ECs on the gene expression pattern of VSMCs is largely unknown. Here, we investigated changes of the VSMC transcriptome by utilizing 3D human vascular organoids organized as a core of VSMCs enclosed by a monolayer of ECs. Microarray-based analyses indicated that interaction with ECs for 48 h down-regulates expression of genes in VSMCs controlling rate-limiting steps of the cholesterol biosynthesis such as HMGCR, HMGCS1, DHCR24 and DHCR7. Protein analyses revealed a decrease in the abundance of DHCR24 (24-dehydrocholesterol reductase) and lower cholesterol levels in VSMCs co-cultured with ECs. On the functional level, the blockade of the DHCR24 activity impaired adhesion, migration and proliferation of VSMCs. Collectively, these findings indicate that ECs have the capacity to instruct VSMCs to shut down the expression of DHCR24 thereby limiting their cholesterol biosynthesis, which may support their functional steady state.


Subject(s)
Cholesterol/metabolism , Endothelial Cells/physiology , Muscle, Smooth, Vascular/metabolism , Nerve Tissue Proteins/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , Cells, Cultured , Gene Expression Regulation, Enzymologic , Human Umbilical Vein Endothelial Cells , Humans , Lipid Metabolism/genetics , Myocytes, Smooth Muscle/metabolism , Nerve Tissue Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism
4.
Exp Cell Res ; 388(1): 111782, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31857114

ABSTRACT

Three-dimensional (3D) cell culture conditions are often used to promote the differentiation of human cells as a prerequisite for the study of organotypic functions and environment-specific cellular responses. Here, we assessed the molecular and functional phenotype of vascular smooth muscle cells (VSMCs) cultured as 3D multilayered aggregates. Microarray studies revealed that these conditions decrease the expression of genes associated with cell cycle control and DNA replication and cease proliferation of VSMCs. This was accompanied by a lower activity level of the mitogen-activated protein kinase ERK1/2 and an increase in autocrine TGFß/SMAD2/3-mediated signaling - a determinant of VSMC differentiation. However, inhibition of TGFß signaling did not affect markers of VSMC differentiation such as smooth muscle myosin heavy chain (MYH11) but stimulated pro-inflammatory NFκB-associated gene expression in the first place while decreasing the protein level of NFKB1/p105 and NFKB2/p100 - inhibitors of NFκB transcriptional activity. Moreover, loss of TGFß signaling also revived VSMC proliferation in 3D aggregates. In conclusion, assembly of VSMCs in multilayered aggregates alters their transcriptome to translate the cellular organization into a resting phenotype. In this context, TGFß signaling appears to attenuate cell growth and NFκB-controlled gene expression representing important aspects of VSMC quiescence.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Cell Aggregation , Cell Proliferation , Cells, Cultured , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Myosin Heavy Chains/metabolism , NF-kappa B/metabolism , Signal Transduction , Smad Proteins/metabolism , Transcriptome , Transforming Growth Factor beta/metabolism
5.
FASEB J ; 33(3): 3364-3377, 2019 03.
Article in English | MEDLINE | ID: mdl-30383452

ABSTRACT

The arterial wall adapts to alterations in blood flow and pressure by remodeling the cellular and extracellular architecture. Biomechanical stress of vascular smooth muscle cells (VSMCs) in the media is thought to precede this process and promote their activation and subsequent proliferation. However, molecular determinants orchestrating the transcriptional phenotype under these conditions have been insufficiently studied. We identified the transcription factor, nuclear factor of activated T cells 5 (NFAT5; or tonicity enhancer-binding protein) as a crucial regulatory element of mechanical stress responses of VSMCs. Here, the relevance of NFAT5 for arterial growth and thickening is investigated in mice upon inducible smooth muscle cell (SMC)-specific genetic ablation of Nfat5. In cultured mouse VSMCs, loss of Nfat5 inhibits the expression of gene sets involved in the control of the cell cycle and the interaction with the extracellular matrix and cytoskeletal dynamics. In vivo, SMC-specific knockout of Nfat5 did not affect the general vascular architecture and blood pressure levels under baseline conditions. However, proliferation of VSMCs and the thickening of the arterial wall were inhibited during both flow-induced collateral remodeling and hypertension-mediated arterial hypertrophy. Whereas originally described as a hypertonicity-responsive transcription factor, these findings identify NFAT5 as a novel molecular determinant of biomechanically induced phenotype changes of VSMCs and wall stress-induced arterial remodeling processes.-Arnold, C., Feldner, A., Zappe, M., Komljenovic, D., De La Torre, C., Ruzicka, P., Hecker, M., Neuhofer, W., Korff, T. Genetic ablation of NFAT5/TonEBP in smooth muscle cells impairs flow- and pressure-induced arterial remodeling in mice.


Subject(s)
Blood Pressure/genetics , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Transcription Factors/genetics , Vascular Remodeling/genetics , Animals , Cell Cycle/genetics , Cell Proliferation/genetics , Cells, Cultured , Extracellular Matrix/genetics , Hypertension/genetics , Mice , Regional Blood Flow/genetics
6.
Dev Med Child Neurol ; 62(11): 1283-1293, 2020 11.
Article in English | MEDLINE | ID: mdl-32588919

ABSTRACT

AIM: To evaluate the effect of progressive resistance training of the ankle plantarflexors on gait efficiency, activity, and participation in adolescents with cerebral palsy (CP). METHOD: Sixty-four adolescents (10-19y; 27 females, 37 males; Gross Motor Function Classification System [GMFCS] levels I-III) were randomized to 30 sessions of resistance training (10 supervised and 20 unsupervised home sessions) over 10 weeks or usual care. The primary outcome was gait efficiency indicated by net nondimensional oxygen cost (NNcost). Secondary outcomes included physical activity, gross motor function, participation, muscle strength, muscle and tendon size, and muscle and tendon stiffness. Analysis was intention-to-treat. RESULTS: Median attendance at the 10 supervised sessions was 80% (range 40-100%). There was no between-group difference in NNcost at 10 (mean difference: 0.02, 95% confidence interval [CI] -0.07 to 0.11, p=0.696) or 22 weeks (mean difference: -0.08, 95% CI -0.18 to 0.03, p=0.158). There was also no evidence of between-group differences in secondary outcomes at 10 or 22 weeks. There were 123 adverse events reported by 27 participants in the resistance training group. INTERPRETATION: We found that 10 supervised sessions and 20 home sessions of progressive resistance training of the ankle plantarflexors did not improve gait efficiency, muscle strength, activity, participation, or any biomechanical outcome among adolescents with CP. WHAT THIS PAPER ADDS: Thirty sessions of progressive resistance training of the ankle plantarflexors over 10 weeks did not improve gait efficiency among ambulatory adolescents with cerebral palsy. Resistance training did not improve muscle strength, activity, or participation. Ninety percent of participants experienced an adverse event. Most adverse events were expected and no serious adverse events were reported.


Subject(s)
Ankle , Cerebral Palsy/rehabilitation , Gait Disorders, Neurologic/rehabilitation , Muscle, Skeletal , Outcome Assessment, Health Care , Resistance Training/methods , Adolescent , Adult , Ankle/physiopathology , Biomechanical Phenomena/physiology , Cerebral Palsy/complications , Child , Exercise/physiology , Gait Disorders, Neurologic/etiology , Humans , Muscle Strength/physiology , Muscle, Skeletal/physiopathology , Resistance Training/adverse effects , Treatment Failure , Young Adult
7.
FASEB J ; 32(4): 2021-2035, 2018 04.
Article in English | MEDLINE | ID: mdl-29208700

ABSTRACT

G protein-mediated signaling plays a decisive role in blood pressure regulation and the phenotype of vascular smooth muscle cells (VSMCs); however, the relevance of proteins that restrict G protein activity is not well characterized in this context. Here, we investigated the influence of regulator of G protein signaling 5 (RGS5), an inhibitor of Gαq/11 and Gαi/o activity, on blood pressure and the VSMC phenotype during experimental hypertension. In mice, loss of RGS5 did not affect baseline blood pressure, but prevented hypertension-induced structural remodeling. RGS5-deficient arterial VSMCs did not acquire a synthetic phenotype as evidenced by their inability to decrease the abundance of contractile markers-α-smooth muscle actin and smooth muscle-myosin heavy chain-or to proliferate under these conditions. Mechanistically, hypertensive pressure levels or biomechanical stretch are sufficient to increase the expression of RGS5. Loss of RGS5 severely impairs the activation of RhoA and stress fiber formation. In stretch-exposed VSMCs, RhoA activity was amplified upon inhibition of PKC, which mimics the downstream effects evoked by RGS5-mediated inhibition of Gαq/11 signaling. Collectively, our findings underline that RhoA activation may depend on the restriction of G protein activity and identify RGS5 as a mechanosensitive regulatory protein that is required to promote the synthetic VSMC phenotype as a prerequisite for structural renovation of the arterial wall during hypertension.-Arnold, C., Demirel, E., Feldner, A., Genové, G., Zhang, H., Sticht, C., Wieland, T., Hecker, M., Heximer, S., Korff, T. Hypertension-evoked RhoA activity in vascular smooth muscle cells requires RGS5.


Subject(s)
Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , RGS Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Male , Mechanotransduction, Cellular , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Myosins/metabolism , Protein Kinase C/metabolism , RGS Proteins/genetics , Stress Fibers/metabolism , rhoA GTP-Binding Protein
8.
Mol Cell Proteomics ; 16(9): 1563-1577, 2017 09.
Article in English | MEDLINE | ID: mdl-28637836

ABSTRACT

Preserving the native phenotype of primary cells in vitro is a complex challenge. Recently, hydrogel-based cellular matrices have evolved as alternatives to conventional cell culture techniques. We developed a bacterial cellulose-based aqueous gel-like biomaterial, dubbed Xellulin, which mimics a cellular microenvironment and seems to maintain the native phenotype of cultured and primary cells. When applied to human umbilical vein endothelial cells (HUVEC), it allowed the continuous cultivation of cell monolayers for more than one year without degradation or dedifferentiation. To investigate the impact of Xellulin on the endothelial cell phenotype in detail, we applied quantitative transcriptomics and proteomics and compared the molecular makeup of native HUVEC, HUVEC on collagen-coated Xellulin and collagen-coated cell culture plastic (polystyrene).Statistical analysis of 12,475 transcripts and 7831 proteins unveiled massive quantitative differences of the compared transcriptomes and proteomes. K-means clustering followed by network analysis showed that HUVEC on plastic upregulate transcripts and proteins controlling proliferation, cell cycle and protein biosynthesis. In contrast, HUVEC on Xellulin maintained, by and large, the expression levels of genes supporting their native biological functions and signaling networks such as integrin, receptor tyrosine kinase MAP/ERK and PI3K signaling pathways, while decreasing the expression of proliferation associated proteins. Moreover, CD34-an endothelial cell differentiation marker usually lost early during cell culture - was re-expressed within 2 weeks on Xellulin but not on plastic. And HUVEC on Xellulin showed a significantly stronger functional responsiveness to a prototypic pro-inflammatory stimulus than HUVEC on plastic.Taken together, this is one of the most comprehensive transcriptomic and proteomic studies of native and propagated HUVEC, which underscores the importance of the morphology of the cellular microenvironment to regulate cellular differentiation, and demonstrates, for the first time, the potential of Xellulin as versatile tool promoting an in vivo-like phenotype in primary and propagated cell culture.


Subject(s)
Cell Differentiation/drug effects , Cellulose/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Proteome/metabolism , Transcriptome/genetics , Cell Separation , Cells, Cultured , Cluster Analysis , Collagen/pharmacology , Gene Expression Profiling , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Stroke ; 49(6): 1479-1487, 2018 06.
Article in English | MEDLINE | ID: mdl-29760276

ABSTRACT

BACKGROUND AND PURPOSE: Detection and localization of the early phase of blood-brain barrier disruption (BBBD) in vivo during cerebral ischemia/reperfusion injury remain a major challenge but may be a relevant outcome parameter in stroke. METHODS: We studied early BBBD in mice after transient middle cerebral artery occlusion by multimodal, high-field (9.4T) in vivo magnetic resonance imaging, including the contrast agent gadofluorineM as an albumin-binding tracer. GadofluorineM contrast-enhanced magnetic resonance imaging was performed to determine BBBD at 2, 6, and 24 hours after reperfusion. BBBD was confirmed and localized along the microvascular tree by using fluorescent gadofluorineM and immunofluorescence stainings (cluster of differentiation 31, ephrin type-B receptor 4, alpha smooth muscle actin, ionized calcium binding adaptor molecule 1). RESULTS: GadofluorineM contrast-enhanced magnetic resonance imaging revealed a multifocal spatial distribution of early BBBD and its close association with the microvasculature at a resolution of 40 µm. GadofluorineM leakage was closely associated with ephrin type-B receptor 4-positive but not alpha smooth muscle actin-positive vessels. The multifocal pattern of early BBBD (already at 2 hours after reperfusion) thus occurred in the distal capillary and venular microvascular bed. These multifocal zones showed distinct imaging signs indicative of early vasogenic edema. The total volume of multifocal early BBBD accurately predicted infarct size at 24 hours after reperfusion. CONCLUSIONS: Early BBBD in focal cerebral ischemia initiates multifocally in the distal capillary and venular bed of the cerebral microvasculature. It is closely associated with perimicrovascular vasogenic edema and microglial activation and predicts the extent of final infarction.


Subject(s)
Blood-Brain Barrier/pathology , Brain Ischemia/pathology , Capillaries/pathology , Stroke/pathology , Animals , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/pathology , Brain Edema/pathology , Cerebrovascular Circulation/physiology , Infarction, Middle Cerebral Artery/pathology , Magnetic Resonance Imaging/methods , Male , Mice, Inbred C57BL , Reperfusion Injury/pathology
10.
FASEB J ; 30(2): 923-32, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26546130

ABSTRACT

Subcutaneously growing tumors are widely utilized to study tumor angiogenesis and the efficacy of antiangiogenic therapies in mice. To additionally assess functional and morphologic alterations of the vasculature in the periphery of a growing tumor, we exploited the easily accessible and hierarchically organized vasculature of the mouse auricle. By site-specific subcutaneous implantation of a defined preformed mouse B16/F0 melanoma aggregate, a solid tumor nodule developed within 14 d. Growth of the tumor nodule was accompanied by a 4-fold increase in its perfusion as well as a 2- to 4-fold elevated diameter and perfusion of peripheral blood vessels that had connected to the tumor capillary microvasculature. By transdermal application of the anticancer drug bortezomib, tumor growth was significantly diminished by about 50% without provoking side effects. Moreover, perfusion and tumor microvessel diameter as well as growth and perfusion of arterial or venous blood vessels supplying or draining the tumor microvasculature were decreased under these conditions by up to 80%. Collectively, we observed that the progressive tumor growth is accompanied by the enlargement of supplying and draining extratumoral blood vessels. This process was effectively suppressed by bortezomib, thereby restricting the perfusion capacity of both extra and intratumoral blood vessels.


Subject(s)
Bortezomib/pharmacology , Drug Delivery Systems/methods , Ear Neoplasms , Melanoma , Neovascularization, Pathologic , Administration, Cutaneous , Animals , Bortezomib/adverse effects , Cell Line, Tumor , Ear Neoplasms/blood supply , Ear Neoplasms/drug therapy , Ear Neoplasms/pathology , Melanoma/blood supply , Melanoma/drug therapy , Melanoma/pathology , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
11.
J Exp Biol ; 220(Pt 7): 1287-1294, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28108669

ABSTRACT

Children perform cyclic motor tasks less efficiently than adults; however, the mechanisms underlying such differences are not fully understood. One mechanism that may contribute to these age-related differences is a differential contribution of muscles and tendons to a given muscle-tendon unit (MTU) excursion. The aims of this study were to (i) compare muscle and tendon excursion between children and adults performing vertical hopping, and (ii) determine whether children and adults choose a hopping frequency that maximizes movement efficiency, based on the utilization of energy-saving mechanisms. Twelve children (8.8±0.3 years) and 12 adults (26.0±2.1 years) performed 20 s of two-legged hopping at a self-selected frequency and at 1.33, 2.00, 2.67 and 3.33 Hz. Gastrocnemius medialis MTU excursion was estimated from kinematic data and muscle and tendon excursions were derived using a combination of 3D-motion capture and ultrasonography. Optimum hopping frequency was determined as the frequency that maximized surrogate measures of elastic energy storage potential of the tendon and minimized muscle excursion. Adults presented a significantly greater potential for elastic energy storage in combination with lower muscle excursion than children at their self-selected frequency, suggesting that children do not utilize these energy-saving mechanisms as effectively as adults. However, tendon elastic energy storage was maximized and muscle excursion minimized at the preferred frequency in both children and adults, indicating that children may select their preferred hopping frequency based on the same criteria as adults. These findings increase our understanding of the mechanisms contributing to the higher energy cost of movement performance in children, and have implications for the interpretation of age-related differences in complex task performance.


Subject(s)
Muscle, Skeletal/physiology , Tendons/physiology , Adult , Biomechanical Phenomena , Child , Elasticity , Female , Humans , Locomotion , Male , Muscle Contraction , Young Adult
12.
FASEB J ; 29(7): 3076-84, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25857554

ABSTRACT

Given the need for robust and cost-efficient in vitro models to study angiogenesis and reproducibly analyze potential pro- and antiangiogenic compounds in preclinical studies, we developed a 3-dimensional in vitro angiogenesis assay that is based on collagen gel-embedded, size-defined spheroids generated from cultured human umbilical vein endothelial cells (HUVECs). Despite its wide distribution, limitations, sensitivity, robustness, and improvements, the capacity of this assay for functional screening purposes has not been elucidated thus far. By using time-lapse video microscopy, we show that tip cells lead the formation of capillary-like and partially lumenized sprouts originating from the spheroids. Angiogenic sprouting from spheroids generated from 5 different primary cultured human endothelial cell types was induced by physiologic concentrations of vascular endothelial cell growth factor 165. Based on this assay system, we determined the capacity of 880 approved drugs to interfere with or boost angiogenic sprouting, thereby assessing their putative angiogenesis-related side effects or novel applications. However, although this assay allowed for a rapid and reproducible determination of functional IC50 values of individual compounds, the sprouting results were partially affected by the HUVEC passage number and donor variability. To overcome this limitation, immortalized HUVECs (iHUVECs) showing a more homogenous response in terms of proliferation and sprouting over multiple population doublings were used in the course of this study. Collectively, the spheroid-based angiogenesis assay provides a sensitive and versatile tool to study the impact of pro- and antiangiogenic determinants on multiple steps of the angiogenic cascade. It is compatible with different endothelial cell types and allows use of iHUVECs to improve its overall robustness.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/physiology , Neovascularization, Physiologic , Spheroids, Cellular/cytology , Spheroids, Cellular/physiology , Angiogenesis Inducing Agents/pharmacology , Angiogenesis Inhibitors/pharmacology , Cells, Cultured , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Fibroblast Growth Factor 2/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Indoles/pharmacology , Microscopy, Video , Neovascularization, Physiologic/drug effects , Pyrroles/pharmacology , Recombinant Proteins/pharmacology , Spheroids, Cellular/drug effects , Vascular Endothelial Growth Factor A/pharmacology
13.
FASEB J ; 28(8): 3518-27, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24769668

ABSTRACT

Despite the high prevalence of venous diseases that are associated with and based on the structural reorganization of the venous vessel wall, not much is known about their mechanistic causes. In this context, we demonstrated that the quantity of myocardin, a transcriptional regulator of the contractile and quiescent smooth muscle cell phenotype, was diminished in proliferating synthetic venous smooth muscle cells (VSMCs) of human and mouse varicose veins by 51 and 60%, respectively. On the basis of the relevance of proteasomal activity for such phenotypic changes, we hypothesized that the observed VSMC activation is attenuated by the proteasome inhibitor bortezomib. This drug fully abolished VSMC proliferation and loss of myocardin in perfused mouse veins and blocked VSMC invasion in collagen gels by almost 80%. In line with this, topical transdermal treatment with bortezomib diminished VSMC proliferation by 80%, rescued 90% of VSMC myocardin abundance, and inhibited varicose-like venous remodeling by 67 to 72% in a mouse model. Collectively, our data indicate that the proteasome plays a pivotal role in VSMC phenotype changes during venous remodeling processes. Its inhibition protects from varicose-like vein remodeling in mice and may thus serve as a putative therapeutic strategy to treat human varicose veins.


Subject(s)
Boronic Acids/therapeutic use , Myocytes, Smooth Muscle/drug effects , Protease Inhibitors/therapeutic use , Pyrazines/therapeutic use , Varicose Veins/drug therapy , Animals , Animals, Outbred Strains , Boronic Acids/pharmacology , Bortezomib , Cell Division/drug effects , Cell Movement , Cells, Cultured , Collagen , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Mice , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/metabolism , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/physiology , Proteolysis , Pyrazines/pharmacology , Spheroids, Cellular , Trans-Activators/metabolism , Varicose Veins/enzymology , Varicose Veins/pathology
14.
Vasa ; 43(2): 88-99, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24627315

ABSTRACT

The development of varicose veins or chronic venous insufficiency is preceded by and associated with the pathophysiological remodelling of the venous wall. Recent work suggests that an increase in venous filling pressure is sufficient to promote varicose remodelling of veins by augmenting wall stress and activating venous endothelial and smooth muscle cells. In line with this, known risk factors such as prolonged standing or an obesity-induced increase in venous filling pressure may contribute to varicosis. This review focuses on biomechanically mediated mechanisms such as an increase in wall stress caused by venous hypertension or alterations in blood flow, which may be involved in the onset of varicose vein development. Finally, possible therapeutic options to counteract or delay the progress of this venous disease are discussed.


Subject(s)
Hemodynamics , Mechanotransduction, Cellular , Varicose Veins/physiopathology , Veins/physiopathology , Venous Insufficiency/physiopathology , Animals , Biomechanical Phenomena , Chronic Disease , Disease Progression , Humans , Risk Factors , Stress, Mechanical , Varicose Veins/etiology , Varicose Veins/pathology , Veins/pathology , Venous Insufficiency/etiology , Venous Insufficiency/pathology , Venous Pressure
15.
J Appl Biomech ; 30(1): 12-20, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23549340

ABSTRACT

The purpose of this study was to examine the interactions between aging, activity levels and maximal power production during cycling. Participants were divided into younger adults (YA), older active adults (OA,) and older sedentary adults (OS). Absolute maximum power was significantly greater in YA compared with OS and OA; no differences were found between OA and OS. The age-related difference in maximum power was accompanied by greater absolute peak knee extension and knee flexion powers. Relative joint power contributions revealed both age- and activity-related differences. YA produced less relative hip extension power than older adults, regardless of activity level. The OS participants produced less relative knee flexion power than active adults, regardless of age. The results show the age-related decline in muscular power production is joint specific and that activity level can be a modifier of intersegmental coordination, which has implications for designing interventions for the aging population.


Subject(s)
Aging/physiology , Energy Metabolism/physiology , Hip Joint/physiology , Knee Joint/physiology , Motor Activity/physiology , Muscle Contraction/physiology , Physical Endurance/physiology , Adult , Female , Humans , Male , Middle Aged , Young Adult
16.
Theranostics ; 14(7): 2856-2880, 2024.
Article in English | MEDLINE | ID: mdl-38773968

ABSTRACT

Cell metabolism reprogramming to sustain energy production, while reducing oxygen and energy consuming processes is crucially important for the adaptation to hypoxia/ischemia. Adaptive metabolic rewiring is controlled by hypoxia-inducible factors (HIFs). Accumulating experimental evidence indicates that timely activation of HIF in brain-resident cells improves the outcome from acute ischemic stroke. However, the underlying molecular mechanisms are still incompletely understood. Thus, we investigated whether HIF-dependent metabolic reprogramming affects the vulnerability of brain-resident cells towards ischemic stress. Methods: We used genetic and pharmacological approaches to activate HIF in the murine brain in vivo and in primary neurons and astrocytes in vitro. Numerous metabolomic approaches and molecular biological techniques were applied to elucidate potential HIF-dependent effects on the central carbon metabolism of brain cells. In animal and cell models of ischemic stroke, we analysed whether HIF-dependent metabolic reprogramming influences the susceptibility to ischemic injury. Results: Neuron-specific gene ablation of prolyl-4-hydroxylase domain 2 (PHD2) protein, negatively regulating the protein stability of HIF-α in an oxygen dependent manner, reduced brain injury and functional impairment of mice after acute stroke in a HIF-dependent manner. Accordingly, PHD2 deficient neurons showed an improved tolerance towards ischemic stress in vitro, which was accompanied by enhanced HIF-1-mediated glycolytic lactate production through pyruvate dehydrogenase kinase-mediated inhibition of the pyruvate dehydrogenase. Systemic treatment of mice with roxadustat, a low-molecular weight pan-PHD inhibitor, not only increased the abundance of numerous metabolites of the central carbon and amino acid metabolism in murine brain, but also ameliorated cerebral tissue damage and sensorimotor dysfunction after acute ischemic stroke. In neurons and astrocytes roxadustat provoked a HIF-1-dependent glucose metabolism reprogramming including elevation of glucose uptake, glycogen synthesis, glycolytic capacity, lactate production and lactate release, which enhanced the ischemic tolerance of astrocytes, but not neurons. We found that strong activation of HIF-1 in neurons by non-selective inhibition of all PHD isoenzymes caused a HIF-1-dependent upregulation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 redirecting glucose-6-phosphate from pentose phosphate pathway (PPP) to the glycolysis pathway. This was accompanied by a reduction of NADPH production in the PPP, which further decreased the low intrinsic antioxidant reserve of neurons, making them more susceptible to ischemic stress. Nonetheless, in organotypic hippocampal cultures with preserved neuronal-glial interactions roxadustat decreased the neuronal susceptibility to ischemic stress, which was largely prevented by restricting glycolytic energy production through lactate transport blockade. Conclusion: Collectively, our results indicate that HIF-1-mediated metabolic reprogramming alleviates the intrinsic vulnerability of brain-resident cells to ischemic stress.


Subject(s)
Astrocytes , Carbon , Hypoxia-Inducible Factor 1, alpha Subunit , Hypoxia-Inducible Factor-Proline Dioxygenases , Ischemic Stroke , Neurons , Animals , Female , Male , Mice , Astrocytes/metabolism , Astrocytes/drug effects , Brain/metabolism , Brain Ischemia/metabolism , Carbon/metabolism , Cellular Reprogramming/drug effects , Disease Models, Animal , Glycolysis/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Ischemic Stroke/metabolism , Mice, Inbred C57BL , Neurons/metabolism , Procollagen-Proline Dioxygenase/metabolism , Procollagen-Proline Dioxygenase/genetics
17.
Sci Transl Med ; 16(736): eabq4581, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38416842

ABSTRACT

Fibrosis is a hallmark of chronic disease. Although fibroblasts are involved, it is unclear to what extent endothelial cells also might contribute. We detected increased expression of the transcription factor Sox9 in endothelial cells in several different mouse fibrosis models. These models included systolic heart failure induced by pressure overload, diastolic heart failure induced by high-fat diet and nitric oxide synthase inhibition, pulmonary fibrosis induced by bleomycin treatment, and liver fibrosis due to a choline-deficient diet. We also observed up-regulation of endothelial SOX9 in cardiac tissue from patients with heart failure. To test whether SOX9 induction was sufficient to cause disease, we generated mice with endothelial cell-specific overexpression of Sox9, which promoted fibrosis in multiple organs and resulted in signs of heart failure. Endothelial Sox9 deletion prevented fibrosis and organ dysfunction in the two mouse models of heart failure as well as in the lung and liver fibrosis mouse models. Bulk and single-cell RNA sequencing of mouse endothelial cells across multiple vascular beds revealed that SOX9 induced extracellular matrix, growth factor, and inflammatory gene expression, leading to matrix deposition by endothelial cells. Moreover, mouse endothelial cells activated neighboring fibroblasts that then migrated and deposited matrix in response to SOX9, a process partly mediated by the secreted growth factor CCN2, a direct SOX9 target; endothelial cell-specific Sox9 deletion reversed these changes. These findings suggest a role for endothelial SOX9 as a fibrosis-promoting factor in different mouse organs during disease and imply that endothelial cells are an important regulator of fibrosis.


Subject(s)
Heart Failure , Transcription Factors , Animals , Humans , Mice , Disease Models, Animal , Endothelial Cells , Fibrosis , Intercellular Signaling Peptides and Proteins , Liver Cirrhosis/complications , SOX9 Transcription Factor/genetics
18.
J Appl Biomech ; 29(2): 241-4, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23645495

ABSTRACT

The muscle-tendon moment arm is an important input parameter for musculoskeletal models. Moment arms change as a function of joint angle and contraction state and depend on the method being employed. The overall purpose was to gain insights into the interactive effects of joint angle, contraction state and method on the Achilles tendon moment arm using the center of rotation (COR) and the tendon excursion method (TE). Achilles tendon moment arms were obtained at rest (TErest, CORrest) and during a maximum voluntary contraction (CORMVC) at four angles. We found strong correlations between TErest and CORMVC for all angles (.72 ≤ r ≤ .93) with Achilles tendon moment arms using CORMVC being 33-36% greater than those obtained from TErest. The relationship between Achilles tendon moment arms and angle was similar across both methods and both levels of muscular contraction. Finally, Achilles tendon moment arms for CORMVC were 1-8% greater than for COR(rest).


Subject(s)
Achilles Tendon/physiology , Ankle Joint/physiology , Movement/physiology , Muscle Contraction/physiology , Biomechanical Phenomena , Female , Humans , Male , Muscle, Skeletal/physiology , Range of Motion, Articular/physiology , Rotation
19.
Nat Cancer ; 4(11): 1544-1560, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37749321

ABSTRACT

Cachexia is a major cause of morbidity and mortality in individuals with cancer and is characterized by weight loss due to adipose and muscle tissue wasting. Hallmarks of white adipose tissue (WAT) remodeling, which often precedes weight loss, are impaired lipid storage, inflammation and eventually fibrosis. Tissue wasting occurs in response to tumor-secreted factors. Considering that the continuous endothelium in WAT is the first line of contact with circulating factors, we postulated whether the endothelium itself may orchestrate tissue remodeling. Here, we show using human and mouse cancer models that during precachexia, tumors overactivate Notch1 signaling in distant WAT endothelium. Sustained endothelial Notch1 signaling induces a WAT wasting phenotype in male mice through excessive retinoic acid production. Pharmacological blockade of retinoic acid signaling was sufficient to inhibit WAT wasting in a mouse cancer cachexia model. This demonstrates that cancer manipulates the endothelium at distant sites to mediate WAT wasting by altering angiocrine signals.


Subject(s)
Adipose Tissue, White , Cachexia , Neoplasms , Receptor, Notch1 , Animals , Humans , Male , Mice , Adipose Tissue, White/pathology , Cachexia/pathology , Neoplasms/complications , Signal Transduction , Tretinoin , Receptor, Notch1/metabolism
20.
Exp Dermatol ; 21(2): 147-53, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22142364

ABSTRACT

In human skin, ultraviolet radiation (UVR)-induced erythema is characterized by the inflammatory and angiogenic activation of dermal endothelial cells. Recently, it has been shown that the release of angiopoietin-2 (Ang-2) from cytoplasmic storages of activated endothelial cells is crucial for the induction of inflammation and angiogenesis. Therefore, we hypothesized that UVR exposure induces the release of Ang-2 from endothelial cells controlling the early steps of erythema formation. In an in vivo study, suction blister fluids generated from UV-irradiated skin showed significantly increased concentrations of Ang-2, vascular endothelial growth factor (VEGF) and tumor necrosis factor-α (TNFα). Likewise, in vitro UVR exposure of human dermal microvascular endothelial cells (HDMECs) triggered the release of Ang-2 that enhanced the pro-inflammatory response of these cells and facilitated their detachment from smooth muscle cells as evidenced by employing a three-dimensional co-culture spheroid model. These effects were inhibited by angiopoietin-1 (Ang-1), which competes with Ang-2 for binding the endothelial cell Tie2 receptor. Collectively, these observations suggest that UVR triggers the release of endothelial Ang-2 which may promote the destabilization and pro-inflammatory phenotype of the microvascular endothelium. As Ang-1 counteracts UVR-induced effects, stimulating the Ang-1 activity may represent a strategy to stabilize the dermal microcirculatory system, thus protecting against UVR-induced skin damages.


Subject(s)
Angiopoietin-2/metabolism , Dermis/cytology , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , Ultraviolet Rays , Adult , Aged , Angiopoietin-1/pharmacology , Angiopoietin-2/pharmacology , Blister/metabolism , Cell Adhesion/drug effects , Cell Adhesion/radiation effects , Coculture Techniques , Dermis/blood supply , E-Selectin/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Gene Expression/drug effects , Gene Expression/genetics , Humans , Middle Aged , Myocytes, Smooth Muscle/cytology , Skin/metabolism , Skin/radiation effects , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Endothelial Growth Factor A/metabolism , Weibel-Palade Bodies/metabolism , Weibel-Palade Bodies/radiation effects , Young Adult , von Willebrand Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL