Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
J Biomech ; 171: 112190, 2024 Jun 09.
Article En | MEDLINE | ID: mdl-38897049

Biological tissues decay over time after harvesting, which alters their biomechanical properties. This poses logistical challenges for studies investigating passive arterial biomechanics as tissues need to be characterized shortly after excision. Freezing and cryopreservation methods can help alleviate the need for biomechanical testing of fresh tissue in human ex vivo studies. However, these methods tend to eliminate or reduce arterial cell functionality and affect passive biomechanics. Furthermore, their impact on dynamic arterial biomechanics remains unknown despite arterial viscoelastic properties being an integral component contributing to arterial stiffness under in vivo loading conditions. The present study aims to investigate the impact of rapid cooling and subsequent storage at -80 °C on the passive viscoelastic properties of arterial tissue and aid in ascertaining whether this is a suitable method to delay tissue analysis for studies investigating passive arterial biomechanics. Control and frozen abdominal rat aorta segments were quasi-statically and dynamically tested using a biaxial testing set-up. The results were modeled using a constituent-based quasi-linear viscoelastic modeling framework, yielding directional stiffness parameters, individual constituent biomechanical contributions, and a quantification of viscoelastic stiffening under dynamic pressurization conditions. Frozen samples displayed significantly decreased wall thickness, viscoelastic dissipation, viscoelastic stiffening, and significantly decreased circumferential deformation with changes in luminal pressure. Furthermore, frozen samples displayed significantly increased circumferential stiffness, pulse wave velocity, and collagen load bearing. Consequently, these changes should be considered when utilizing this tissue preservation method to delay biomechanical characterization of rat aortic tissue.

2.
Mol Neurobiol ; 2023 Dec 26.
Article En | MEDLINE | ID: mdl-38147228

The blood-brain barrier consists of tightly connected endothelial cells protecting the brain's microenvironment from the periphery. These endothelial cells are characterized by specific tight junction proteins such as Claudin-5 and Occludin, forming the endothelial barrier. Disrupting these cells might lead to blood-brain barrier dysfunction. The Wnt/ß-catenin signaling pathway can regulate the expression of these tight junction proteins and subsequent barrier permeability. The aim of this study was to investigate the in vitro effects of Wnt7a mediated ß-catenin signaling on endothelial barrier integrity. Mouse brain endothelial cells, bEnd.3, were treated with recombinant Wnt7a protein or XAV939, a selective inhibitor of Wnt/ß-catenin mediated transcription to modulate the Wnt signaling pathway. The involvement of Wnt/HIF1α signaling was investigated by inhibiting Hif1α signaling with Hif1α siRNA. Wnt7a stimulation led to activation and nuclear translocation of ß-catenin, which was inhibited by XAV939. Wnt7a stimulation decreased Claudin-5 expression mediated by ß-catenin and decreased endothelial barrier formation. Wnt7a increased Hif1α and Vegfa expression mediated by ß-catenin. However, Hif1α signaling pathway did not regulate tight junction proteins Claudin-5 and Occludin. Our data suggest that Wnt7a stimulation leads to a decrease in tight junction proteins mediated by the nuclear translocation of ß-catenin, which hampers proper endothelial barrier formation. This process might be crucial in initiating endothelial cell proliferation and angiogenesis. Although HIF1α did not modulate the expression of tight junction proteins, it might play a role in brain angiogenesis and underlie pathogenic mechanisms in Wnt/HIF1α signaling in diseases such as cerebral small vessel disease.

3.
Fluids Barriers CNS ; 20(1): 75, 2023 Oct 24.
Article En | MEDLINE | ID: mdl-37875994

The brain is a highly metabolically active organ requiring a large amount of glucose. Methylglyoxal (MGO), a by-product of glucose metabolism, is known to be involved in microvascular dysfunction and is associated with reduced cognitive function. Maintenance of the blood-brain barrier (BBB) is essential to maintain optimal brain function and a large amount of evidence indicates negative effects of MGO on BBB integrity. In this review, we summarized the current literature on the effect of MGO on the different cell types forming the BBB. BBB damage by MGO most likely occurs in brain endothelial cells and mural cells, while astrocytes are most resistant to MGO. Microglia on the other hand appear to be not directly influenced by MGO but rather produce MGO upon activation. Although there is clear evidence that MGO affects components of the BBB, the impact of MGO on the BBB as a multicellular system warrants further investigation. Diminishing MGO stress can potentially form the basis for new treatment strategies for maintaining optimal brain function.


Blood-Brain Barrier , Endothelial Cells , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Pyruvaldehyde/pharmacology , Pyruvaldehyde/metabolism , Magnesium Oxide/pharmacology , Brain/metabolism
4.
Pharmacol Res ; 197: 106970, 2023 Nov.
Article En | MEDLINE | ID: mdl-37884069

Vascular cognitive impairment (VCI) describes neurodegenerative disorders characterized by a vascular component. Pathologically, it involves decreased cerebral blood flow (CBF), white matter lesions, endothelial dysfunction, and blood-brain barrier (BBB) impairments. Molecularly, oxidative stress and inflammation are two of the major underlying mechanisms. Nitric oxide (NO) physiologically stimulates soluble guanylate cyclase (sGC) to induce cGMP production. However, under pathological conditions, NO seems to be at the basis of oxidative stress and inflammation, leading to a decrease in sGC activity and expression. The native form of sGC needs a ferrous heme group bound in order to be sensitive to NO (Fe(II)sGC). Oxidation of sGC leads to the conversion of ferrous to ferric heme (Fe(III)sGC) and even heme-loss (apo-sGC). Both Fe(III)sGC and apo-sGC are insensitive to NO, and the enzyme is therefore inactive. sGC activity can be enhanced either by targeting the NO-sensitive native sGC (Fe(II)sGC), or the inactive, oxidized sGC (Fe(III)sGC) and the heme-free apo-sGC. For this purpose, sGC stimulators acting on Fe(II)sGC and sGC activators acting on Fe(III)sGC/apo-sGC have been developed. These sGC agonists have shown their efficacy in cardiovascular diseases by restoring the physiological and protective functions of the NO-sGC-cGMP pathway, including the reduction of oxidative stress and inflammation, and improvement of vascular functioning. Yet, only very little research has been performed within the cerebrovascular system and VCI pathology when focusing on sGC modulation and its potential protective mechanisms on vascular and neural function. Therefore, within this review, the potential of sGC as a target for treating VCI is highlighted.


Cognitive Dysfunction , Vascular Diseases , Humans , Soluble Guanylyl Cyclase , Cognitive Dysfunction/drug therapy , Cyclic GMP , Heme , Inflammation
5.
Acta Neuropathol Commun ; 11(1): 128, 2023 08 07.
Article En | MEDLINE | ID: mdl-37550790

Cerebral small vessel disease is characterised by decreased cerebral blood flow and blood-brain barrier impairments which play a key role in the development of white matter lesions. We hypothesised that cerebral hypoperfusion causes local hypoxia, affecting oligodendrocyte precursor cell-endothelial cell signalling leading to blood-brain barrier dysfunction as an early mechanism for the development of white matter lesions. Bilateral carotid artery stenosis was used as a mouse model for cerebral hypoperfusion. Pimonidazole, a hypoxic cell marker, was injected prior to humane sacrifice at day 7. Myelin content, vascular density, blood-brain barrier leakages, and hypoxic cell density were quantified. Primary mouse oligodendrocyte precursor cells were exposed to hypoxia and RNA sequencing was performed. Vegfa gene expression and protein secretion was examined in an oligodendrocyte precursor cell line exposed to hypoxia. Additionally, human blood plasma VEGFA levels were measured and correlated to blood-brain barrier permeability in normal-appearing white matter and white matter lesions of cerebral small vessel disease patients and controls. Cerebral blood flow was reduced in the stenosis mice, with an increase in hypoxic cell number and blood-brain barrier leakages in the cortical areas but no changes in myelin content or vascular density. Vegfa upregulation was identified in hypoxic oligodendrocyte precursor cells, which was mediated via Hif1α and Epas1. In humans, VEGFA plasma levels were increased in patients versus controls. VEGFA plasma levels were associated with increased blood-brain barrier permeability in normal appearing white matter of patients. Cerebral hypoperfusion mediates hypoxia induced VEGFA expression in oligodendrocyte precursor cells through Hif1α/Epas1 signalling. VEGFA could in turn increase BBB permeability. In humans, increased VEGFA plasma levels in cerebral small vessel disease patients were associated with increased blood-brain barrier permeability in the normal appearing white matter. Our results support a role of VEGFA expression in cerebral hypoperfusion as seen in cerebral small vessel disease.


Cerebral Small Vessel Diseases , Oligodendrocyte Precursor Cells , White Matter , Humans , Mice , Animals , Blood-Brain Barrier/metabolism , Oligodendrocyte Precursor Cells/metabolism , White Matter/pathology , Hypoxia/metabolism , Cerebral Small Vessel Diseases/pathology , Vascular Endothelial Growth Factor A/metabolism
6.
Molecules ; 28(14)2023 Jul 18.
Article En | MEDLINE | ID: mdl-37513355

The AT1 receptor has mainly been associated with the pathological effects of the renin-angiotensin system (RAS) (e.g., hypertension, heart and kidney diseases), and constitutes a major therapeutic target. In contrast, the AT2 receptor is presented as the protective arm of this RAS, and its targeting via specific agonists is mainly used to counteract the effects of the AT1 receptor. The discovery of a local RAS has highlighted the importance of the balance between AT1/AT2 receptors at the tissue level. Disruption of this balance is suggested to be detrimental. The fine tuning of this balance is not limited to the regulation of the level of expression of these two receptors. Other mechanisms still largely unexplored, such as S-nitrosation of the AT1 receptor, homo- and heterodimerization, and the use of AT1 receptor-biased agonists, may significantly contribute to and/or interfere with the settings of this AT1/AT2 equilibrium. This review will detail, through several examples (the brain, wound healing, and the cellular cycle), the importance of the functional balance between AT1 and AT2 receptors, and how new molecular pharmacological approaches may act on its regulation to open up new therapeutic perspectives.


Cardiovascular System , Hypertension , Humans , Renin-Angiotensin System/physiology , Cardiovascular System/metabolism , Hypertension/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Angiotensin II/metabolism
7.
Biomech Model Mechanobiol ; 22(5): 1607-1623, 2023 Oct.
Article En | MEDLINE | ID: mdl-37129690

Arteries exhibit fully nonlinear viscoelastic behaviours (i.e. both elastically and viscously nonlinear). While elastically nonlinear arterial models are well established, effective mathematical descriptions of nonlinear viscoelasticity are lacking. Quasi-linear viscoelasticity (QLV) offers a convenient way to mathematically describe viscoelasticity, but its viscous linearity assumption is unsuitable for whole-wall vascular applications. Conversely, application of fully nonlinear viscoelastic models, involving deformation-dependent viscous parameters, to experimental data is impractical and often reduces to identifying specific solutions for each tested loading condition. The present study aims to address this limitation: By applying QLV theory at the wall constituent rather than at the whole-wall level, the deformation-dependent relative contribution of the constituents allows to capture nonlinear viscoelasticity with a unique set of deformation-independent model parameters. Five murine common carotid arteries were subjected to a protocol of quasi-static and harmonic, pseudo-physiological biaxial loading conditions to characterise their viscoelastic behaviour. The arterial wall was modelled as a constrained mixture of an isotropic elastin matrix and four families of collagen fibres. Constituent-based QLV was implemented by assigning different relaxation functions to collagen- and elastin-borne parts of the wall stress. Nonlinearity in viscoelasticity was assessed via the pressure dependency of the dynamic-to-quasi-static stiffness ratio. The experimentally measured ratio increased with pressure, from 1.03 [Formula: see text] 0.03 (mean [Formula: see text] standard deviation) at 80-40 mmHg to 1.58 [Formula: see text] 0.22 at 160-120 mmHg. Constituent-based QLV captured well this trend by attributing the wall viscosity predominantly to collagen fibres, whose recruitment starts at physiological pressures. In conclusion, constituent-based QLV offers a practical and effective solution to model arterial viscoelasticity.


Elastin , Nonlinear Dynamics , Animals , Mice , Viscosity , Collagen , Carotid Artery, Common , Elasticity , Stress, Mechanical , Models, Biological
8.
J Hypertens ; 41(7): 1159-1167, 2023 07 01.
Article En | MEDLINE | ID: mdl-37071429

BACKGROUND: SBP and blood pressure variability are independent risk factors for cerebral small vessel disease, a leading cause for stroke and dementia. Calcium-channel blockers are known to reduce blood pressure variability and may thus offer benefit against dementia. Beyond this effect, the impact of calcium-channel blockers on hypertension-induced neuroinflammation, and especially, microglial phenotype remains unknown. We aimed to study the ability of amlopidine to alleviate microglia inflammation, and slow down cognitive dysfunction in aged hypertensive mice. METHODS: Hypertensive BPH/2J and normotensive BPN/3J mice were studied until 12 months of age. Hypertensive mice were untreated or received amlodipine (10 mg/kg per day). Blood pressure parameters were measured by telemetry and tail cuff plethysmography. Mice underwent repeated series of cognitive tasks. Brain immunohistochemistry was performed to study blood-brain barrier dysfunction and microglial pro-inflammatory phenotype (CD68 + Iba1 + cells; morphological analysis). RESULTS: Amlodipine normalized SBP over the entire life span and decreased blood pressure variability. BPH/2J mice exhibited impaired short-term memory that was prevented by amlodipine at 12 months (discrimination index 0.41 ±â€Š0.25 in amlodipine-treated vs. 0.14 ±â€Š0.15 in untreated BPH/2J mice, P  = 0.02). Amlopidine treatment of BPH/2J did not prevent blood-brain barrier leakage, a measure of cerebral small vessel disease, but limited its size. Microglia's inflammatory phenotype in BPH/2J, characterized by an increased number of Iba1 + CD68 + cells, increased soma size and shortened processes, was partly reduced by amlodipine. CONCLUSION: Amlodipine attenuated the short-term memory impairment in aged hypertensive mice. Beyond its blood pressure lowering capacity, amlodipine may be cerebroprotective by modulating neuroinflammation.


Dementia , Hypertension , Animals , Mice , Amlodipine/pharmacology , Amlodipine/therapeutic use , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Blood Pressure/physiology , Calcium , Calcium Channel Blockers/therapeutic use , Hypertension/complications , Hypertension/drug therapy , Hypertension/genetics , Microglia , Neuroinflammatory Diseases
9.
J Cereb Blood Flow Metab ; 42(6): 979-996, 2022 06.
Article En | MEDLINE | ID: mdl-35209740

Extremely low frequency electromagnetic stimulation (ELF-EMS) has been considered as a neuroprotective therapy for ischemic stroke based on its capacity to induce nitric oxide (NO) signaling. Here, we examined whether ELF-EMS reduces ischemic stroke volume by stimulating cerebral collateral perfusion. Moreover, the pathway responsible for ELF-EMS-induced NO production was investigated. ELF-EMS diminished infarct growth following experimental stroke in collateral-rich C57BL/6 mice, but not in collateral-scarce BALB/c mice, suggesting that decreased lesion sizes after ELF-EMS results from improved collateral blood flow. In vitro analysis demonstrated that ELF-EMS increased endothelial NO levels by stimulating the Akt-/eNOS pathway. Furthermore, ELF-EMS augmented perfusion in the hind limb of healthy mice, which was mediated by enhanced Akt-/eNOS signaling. In healthy C57BL/6 mouse brains, ELF-EMS treatment increased cerebral blood flow in a NOS-dependent manner, whereas no improvement in cerebrovascular perfusion was observed in collateral-sparse BALB/c mice. In addition, ELF-EMS enhanced cerebral blood flow in both the contra- and ipsilateral hemispheres of C57BL/6 mice subjected to experimental ischemic stroke. In conclusion, we showed that ELF-EMS enhances (cerebro)vascular perfusion by stimulating NO production, indicating that ELF-EMS could be an attractive therapeutic strategy for acute ischemic stroke by improving cerebral collateral blood flow.


Brain Ischemia , Ischemic Stroke , Animals , Brain Ischemia/therapy , Cerebrovascular Circulation , Collateral Circulation/physiology , Electromagnetic Phenomena , Ischemia , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide , Proto-Oncogene Proteins c-akt
10.
Pharmaceuticals (Basel) ; 14(8)2021 Aug 06.
Article En | MEDLINE | ID: mdl-34451870

Blockers of angiotensin II type 1 receptor (AT1R) exert antidepressant-like effects by indirectly facilitating the activation of the angiotensin II type 2 receptor (AT2R), which leads to increased surface expression and transactivation of tropomyosin-related kinase B receptors (TRKB). Compound 21 (C21) is a non-peptide AT2R agonist that produces neuroprotective effects. However, the behavioral effects of C21 and its involvement with the brain-derived neurotrophic factor (BDNF)-TRKB system still need further investigation. The aim of the present study was to assess the effect of C21 on the activation of TRKB and its consequences on conditioned fear. The administration of C21 (0.1-10 µM/15 min) increased the surface levels of TRKB but was not sufficient to increase the levels of phosphorylated TRKB (pTRKB) in cultured cortical neurons from rat embryos. Consistent with increased TRKB surface expression, C21 (10 µM/15 min or 3 days) facilitated the effect of BDNF (0.1 ng/mL/15 min) on pTRKB in these cells. In contextual fear conditioning, the freezing time of C21-treated (administered intranasally) wild-type mice was decreased compared to the vehicle-treated group, but no effect of C21 was observed in BDNF.het animals. We observed no effect of C21 in the elevated plus-maze test for anxiety. Taken together, our results indicate that C21 facilitated BDNF effect by increasing the levels of TRKB on the cell surface and reduced the freezing time of mice in a BDNF-dependent manner, but not through a general anxiolytic-like effect.

11.
Cells ; 9(12)2020 12 10.
Article En | MEDLINE | ID: mdl-33322009

Vascular calcification (VC), the ectopic deposition of calcium phosphate crystals in the vessel wall, is one of the primary contributors to cardiovascular death. The pathology of VC is determined by vascular topography, pre-existing diseases, and our genetic heritage. VC evolves from inflammation, mediated by macrophages, and from the osteochondrogenic transition of vascular smooth muscle cells (VSMC) in the atherosclerotic plaque. This pathologic transition partly resembles endochondral ossification, involving the chronologically ordered activation of the ß-catenin-independent and -dependent Wingless and Int-1 (WNT) pathways and the termination of peroxisome proliferator-activated receptor γ (PPARγ) signal transduction. Several atherosclerotic plaque studies confirmed the differential activity of PPARγ and the WNT signaling pathways in VC. Notably, the actively regulated ß-catenin-dependent and -independent WNT signals increase the osteochondrogenic transformation of VSMC through the up-regulation of the osteochondrogenic transcription factors SRY-box transcription factor 9 (SOX9) and runt-related transcription factor 2 (RUNX2). In addition, we have reported studies showing that WNT signaling pathways may be antagonized by PPARγ activation via the expression of different families of WNT inhibitors and through its direct interaction with ß-catenin. In this review, we summarize the existing knowledge on WNT and PPARγ signaling and their interplay during the osteochondrogenic differentiation of VSMC in VC. Finally, we discuss knowledge gaps on this interplay and its possible clinical impact.


PPAR gamma/metabolism , Vascular Calcification/metabolism , Wnt Proteins/metabolism , Chondrogenesis , Core Binding Factor Alpha 1 Subunit/genetics , Humans , Osteogenesis , SOX9 Transcription Factor/genetics , Signal Transduction , Up-Regulation , Vascular Calcification/genetics , beta Catenin/metabolism
12.
Theranostics ; 10(21): 9512-9527, 2020.
Article En | MEDLINE | ID: mdl-32863942

Rationale: Hypertension is a major risk factor for cerebral small vessel disease, the most prevalent cause of vascular cognitive impairment. As we have shown, hypertension induced by a prolonged Angiotensin II infusion is associated with increased permeability of the blood-brain barrier (BBB), chronic activation of microglia and myelin loss. In this study we therefore aim to determine the contribution of microglia to hypertension-induced cognitive impairment in an experimental hypertension model by a pharmacological depletion approach. Methods: For this study, adult Cx3Cr1 gfp/wtxThy1 yfp/0 reporter mice were infused for 12 weeks with Angiotensin II or saline and subgroups were treated with PLX5622, a highly selective CSF1R tyrosine kinase inhibitor. Systolic blood pressure (SBP) was measured via tail-cuff. Short- and long-term spatial memory was assessed during an Object Location task and a Morris Water Maze task (MWM). Microglia depletion efficacy was assessed by flow cytometry and immunohistochemistry. BBB leakages, microglia phenotype and myelin integrity were assessed by immunohistochemistry. Results: SBP, heart weight and carotid pulsatility were increased by Ang II and were not affected by PLX5622. Short-term memory was significantly impaired in Ang II hypertensive mice, and partly prevented in Ang II mice treated with PLX5622. Histological and flow cytometry analysis revealed almost complete ablation of microglia and a 60% depletion of brain resident perivascular macrophages upon CSF1R inhibition. Number and size of BBB leakages were increased in Ang II hypertensive mice, but not altered by PLX5622 treatment. Microglia acquired a pro-inflammatory phenotype at the site of BBB leakages in both Saline and Ang II mice and were successfully depleted by PLX5622. There was however no significant change in myelin integrity at the site of leakages. Conclusion: Our results show that depletion of microglia and PVMs, by CSF1R inhibition prevents short-term memory impairment in Ang II induced hypertensive mice. We suggest this beneficial effect is mediated by the major decrease of pro-inflammatory microglia within BBB leakages. This novel finding supports the critical role of brain immune cells in the pathogenesis of hypertension-related cognitive impairment. An adequate modulation of microglia /PVM density and phenotype may constitute a relevant approach to prevent and/or limit the progression of vascular cognitive impairment.


Angiotensin II/pharmacology , Cognitive Dysfunction/prevention & control , Enzyme Inhibitors/pharmacology , Hypertension/chemically induced , Macrophages/drug effects , Microglia/drug effects , Animals , Blood Pressure/drug effects , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/metabolism , Cognitive Dysfunction/metabolism , Disease Models, Animal , Hypertension/metabolism , Macrophages/metabolism , Male , Mice , Microglia/metabolism , Organic Chemicals/pharmacology
13.
Cells ; 9(6)2020 06 25.
Article En | MEDLINE | ID: mdl-32630426

Key pathological features of cerebral small vessel disease (cSVD) include impairment of the blood brain barrier (BBB) and the progression of white matter lesions (WMLs) amongst other structural lesions, leading to the clinical manifestations of cSVD. The function of endothelial cells (ECs) is of major importance to maintain a proper BBB. ECs interact with several cell types to provide structural and functional support to the brain. Oligodendrocytes (OLs) myelinate axons in the central nervous system and are crucial in sustaining the integrity of white matter. The interplay between ECs and OLs and their precursor cells (OPCs) has received limited attention yet seems of relevance for the study of BBB dysfunction and white matter injury in cSVD. Emerging evidence shows a crosstalk between ECs and OPCs/OLs, mediated by signaling through the Wingless and Int-1 (WNT)/ß-catenin pathway. As the latter is involved in EC function (e.g., angiogenesis) and oligodendrogenesis, we reviewed the role of WNT/ß-catenin signaling for both cell types and performed a systematic search to identify studies describing a WNT-mediated interplay between ECs and OPCs/OLs. Dysregulation of this interaction may limit remyelination of WMLs and render the BBB leaky, thereby initiating a vicious neuroinflammatory cycle. A better understanding of the role of this signaling pathway in EC-OL crosstalk is essential in understanding cSVD development.


Cerebral Small Vessel Diseases/genetics , Endothelial Cells/metabolism , Oligodendroglia/metabolism , Wnt Signaling Pathway/genetics , Humans
15.
Front Neurosci ; 13: 1291, 2019.
Article En | MEDLINE | ID: mdl-31866808

Cerebral small vessels feed and protect the brain parenchyma thanks to the unique features of the blood-brain barrier. Cerebrovascular dysfunction is therefore seen as a detrimental factor for the initiation of several central nervous system (CNS) disorders, such as stroke, cerebral small vessel disease (cSVD), and Alzheimer's disease. The main working hypothesis linking cerebrovascular dysfunction to brain disorders includes the contribution of neuroinflammation. While our knowledge on microglia cells - the brain-resident immune cells - has been increasing in the last decades, the specific populations of microglia and macrophages surrounding brain vessels, vessel-associated microglia (VAM), and perivascular macrophages (PVMs), respectively, have been overlooked. This review aims to summarize the knowledge gathered on VAM and PVMs, to discuss existing knowledge gaps of importance for later studies and to summarize evidences for their contribution to cerebrovascular dysfunction.

16.
J Neuroinflammation ; 16(1): 79, 2019 Apr 10.
Article En | MEDLINE | ID: mdl-30971251

BACKGROUND: Microglia play crucial roles in the maintenance of brain homeostasis. Activated microglia show a biphasic influence, promoting beneficial repair and causing harmful damage via M2 and M1 microglia, respectively. It is well-known that microglia are initially activated to the M2 state and subsequently switch to the M1 state, called M2-to-M1 class switching in acute ischemic models. However, the activation process of microglia in chronic and sporadic hypertension remains poorly understood. We aimed to clarify the process using a chronic hypertension model, the deoxycorticosterone acetate (DOCA)-salt-treated Wistar rats. METHODS: After unilateral nephrectomy, the rats were randomly divided into DOCA-salt, placebo, and control groups. DOCA-salt rats received a weekly subcutaneous injection of DOCA (40 mg/kg) and were continuously provided with 1% NaCl in drinking water. Placebo rats received a weekly subcutaneous injection of vehicle and were provided with tap water. Control rats received no administration of DOCA or NaCl. To investigate the temporal expression profiles of M1- and M2-specific markers for microglia, the animals were subjected to the immunohistochemical and biochemical studies after 2, 3, or 4 weeks DOCA-salt treatment. RESULTS: Hypertension occurred after 2 weeks of DOCA and salt administration, when round-shaped microglia with slightly shortened processes were observed juxtaposed to the vessels, although the histopathological findings were normal. After 3 weeks of DOCA and salt administration, M1-state perivascular and parenchyma microglia significantly increased, when local histopathological findings began to be observed but cerebrovascular destruction did not occur. On the other hand, M2-state microglia were never observed around the vessels at this period. Interestingly, prior to M1 activation, about 55% of perivascular microglia transiently expressed Ki-67, one of the cell proliferation markers. CONCLUSIONS: We concluded that the resting perivascular microglia directly switched to the pro-inflammatory M1 state via a transient proliferative state in DOCA-salt rats. Our results suggest that the activation machinery of microglia in chronic hypertension differs from acute ischemic models. Proliferative microglia are possible initial key players in the development of hypertension-induced cerebral vessel damage. Fine-tuning of microglia proliferation and activation could constitute an innovative therapeutic strategy to prevent its development.


Brain/pathology , Cell Proliferation/physiology , Hypertension/complications , Hypertension/pathology , Microglia/classification , Microglia/pathology , Animals , Antigens, CD/metabolism , Blood Pressure/drug effects , Brain/drug effects , Calcium-Binding Proteins/metabolism , Carboxymethylcellulose Sodium/pharmacology , Cell Proliferation/drug effects , Desoxycorticosterone Acetate/toxicity , Disease Models, Animal , Functional Laterality , Hypertension/diagnostic imaging , Hypertension/etiology , Ki-67 Antigen/metabolism , Magnetic Resonance Imaging , Male , Microfilament Proteins/metabolism , Microglia/drug effects , Mineralocorticoids/toxicity , Nephrectomy/adverse effects , Rats , Rats, Wistar , Sodium Chloride/toxicity , Time Factors
17.
Eur J Med Chem ; 158: 334-352, 2018 Oct 05.
Article En | MEDLINE | ID: mdl-30223121

Because of the complex biological networks, many pathologic disorders fail to be treated with a molecule directed towards a single target. Thus, combination therapies are often necessary, but they have many drawbacks. An alternative consists in building molecules intended to interact with multiple targets, called designed multiple ligands. We followed such a strategy in order to treat metabolic syndrome, by setting up molecules directed towards both type 1 angiotensin II (AT1) receptor and peroxisome proliferator-activated receptor-γ (PPAR-γ). For this purpose, many molecules were prepared by merging both pharmacophores following three different strategies. Their ability to activate PPAR-γ and to block AT1 receptors were evaluated in vitro. This strategy led to the preparation of many new PPAR-γ activating and AT1 blocking molecules. Among them, some exhibited both activities, highlighting the convenience of this approach.


Angiotensin II Type 1 Receptor Blockers/chemistry , Angiotensin II Type 1 Receptor Blockers/pharmacology , Drug Design , PPAR gamma/agonists , Angiotensin II Type 1 Receptor Blockers/chemical synthesis , Animals , Chromans/chemical synthesis , Chromans/chemistry , Chromans/pharmacology , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Imidazoles/pharmacology , Ligands , MCF-7 Cells , Male , Molecular Docking Simulation , PPAR gamma/metabolism , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Triazoles/chemical synthesis , Triazoles/chemistry , Triazoles/pharmacology
18.
Hypertens Res ; 41(10): 817-827, 2018 Oct.
Article En | MEDLINE | ID: mdl-30120397

The causal relation between hypertension and cerebral small vessel disease (cSVD) remains elusive, and appropriate animal models are scarce. We aimed to assess the relevance of prolonged angiotensin II-induced hypertension in mice for the study of cSVD.Adult male C57BL/6 mice were continuously infused for 3 months with Angiotensin II (Ang II; 2 µg/kg/min, sc) or saline (control) via osmotic minipumps. Blood pressure, neurological function, locomotor activity, and working memory (Y-maze alternation task) were assessed throughout the study. Short-term memory performance (object location task) was measured after 3 months of infusion. Blood-brain barrier (BBB) function was assessed by the presence of IgG leakage and quantified in each brain area of interest. Microglial activation and myelin loss were studied in the areas of leakage.Systolic blood pressure increased and remained elevated over the 3 months of Ang II infusion, while neurological scores and locomotor activity did not change. Working memory performance was also not changed, yet short-term memory performance was impaired in Ang II-treated mice compared to controls. While BBB leakages were present in both groups, mainly in the neocortex, hippocampus, and cerebral nuclei, Ang II-treated mice showed greater leakage than control mice, along with greater microglial density and soma size. Myelin loss was observed for the largest leaks.Prolonged Ang II-induced hypertension is associated with large BBB leaks, microglial activation, myelin loss, and memory dysfunction in the absence of stroke.


Angiotensin II , Brain/physiopathology , Cognitive Dysfunction/etiology , Hypertension/complications , Memory, Short-Term/physiology , Animals , Blood Pressure/physiology , Blood-Brain Barrier/physiopathology , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Hypertension/chemically induced , Hypertension/physiopathology , Male , Mice , Motor Activity/physiology
19.
Pharmacol Rev ; 70(1): 68-141, 2018 01.
Article En | MEDLINE | ID: mdl-29247129

WNT signaling is an elaborate and complex collection of signal transduction pathways mediated by multiple signaling molecules. WNT signaling is critically important for developmental processes, including cell proliferation, differentiation and tissue patterning. Little WNT signaling activity is present in the cardiovascular system of healthy adults, but reactivation of the pathway is observed in many pathologies of heart and blood vessels. The high prevalence of these pathologies and their significant contribution to human disease burden has raised interest in WNT signaling as a potential target for therapeutic intervention. In this review, we first will focus on the constituents of the pathway and their regulation and the different signaling routes. Subsequently, the role of WNT signaling in cardiovascular development is addressed, followed by a detailed discussion of its involvement in vascular and cardiac disease. After highlighting the crosstalk between WNT, transforming growth factor-ß and angiotensin II signaling, and the emerging role of WNT signaling in the regulation of stem cells, we provide an overview of drugs targeting the pathway at different levels. From the combined studies we conclude that, despite the sometimes conflicting experimental data, a general picture is emerging that excessive stimulation of WNT signaling adversely affects cardiovascular pathology. The rapidly increasing collection of drugs interfering at different levels of WNT signaling will allow the evaluation of therapeutic interventions in the pathway in relevant animal models of cardiovascular diseases and eventually in patients in the near future, translating the outcomes of the many preclinical studies into a clinically relevant context.


Cardiovascular Diseases/metabolism , Wnt Signaling Pathway/physiology , Animals , Cardiovascular Diseases/drug therapy , Humans , Molecular Targeted Therapy , Wnt Signaling Pathway/drug effects
20.
Drugs ; 76(1): 1-12, 2016 Jan.
Article En | MEDLINE | ID: mdl-26631237

Cardiovascular and metabolic disorders, such as hypertension, insulin resistance, dyslipidemia or obesity are linked with chronic low-grade inflammation and dysregulation of the renin-angiotensin system (RAS). Consequently, RAS inhibition by ACE inhibitors or angiotensin AT1 receptor (AT1R) blockers is the evidence-based standard for cardiovascular risk reduction in high-risk patients, including diabetics with albuminuria. In addition, RAS inhibition reduces the new onset of diabetes mellitus. Yet, the high and increasing prevalence of metabolic disorders, and the high residual risk even in properly treated patients, calls for additional means of pharmacological intervention. In the past decade, the stimulation of the angiotensin AT2 receptor (AT2R) has been shown to reduce inflammation, improve cardiac and vascular remodeling, enhance insulin sensitivity and increase adiponectin production. Therefore, a concept of dual AT1R/AT2R modulation emerges as a putative means for risk reduction in cardio-metabolic diseases. The approach employing simultaneous RAS blockade (AT1R) and RAS stimulation (AT2R) is distinct from previous attempts of double intervention in the RAS by dual blockade. Dual blockade abolishes the AT1R-linked RAS almost completely with subsequent risk of hypotension and hypotension-related events, i.e. syncope or renal dysfunction. Such complications might be especially prominent in patients with renal impairment or patients with isolated systolic hypertension and normal-to-low diastolic blood pressure values. In contrast to dual RAS blockade, the add-on of AT2R stimulation does not exert significant blood pressure effects, but it may complement and enhance the anti-inflammatory and antifibrotic/de-stiffening effects of the AT1R blockade and improve the metabolic profile. Further studies will have to investigate these putative effects in particular for settings in which blood pressure reduction is not primarily desired.


Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Cardiovascular Diseases/drug therapy , Metabolic Diseases/drug therapy , Receptor, Angiotensin, Type 2/agonists , Renin-Angiotensin System/drug effects , Cardiovascular Diseases/complications , Cardiovascular Diseases/physiopathology , Drug Therapy, Combination , Humans , Inflammation/complications , Inflammation/drug therapy , Metabolic Diseases/complications , Metabolic Diseases/physiopathology
...