90 research outputs found

    Response to 'The evolving mystery of why skeletal muscle is spared in seropositive neuromyelitis optica'

    Get PDF
    Dear Editor, We would like to thank you for the opportunity to respond to the questions raised in Dr Verkman's letter and to elucidate related aspects. We also thank Dr Verkman and colleagues for their attention to our study. The use of g‐STED super‐resolution microscopy versus freeze‐fracture electron microscopy (FFEM) to analyse skeletal muscle and brain AQP4 supramolecular assemblies (OAPs) used in our study 1 has been disputed by Verkman et al. While we agree that FFEM is the gold standard to visualise OAPs and also measure their size, we also are aware that the very small amount of the plasma membrane that can be suitable for analysis represents a major limit to obtaining statistically significant data (such as the OAP dimension) representative of the entire tissue. In contrast, STED microscopy has the enormous advantage of analysing, in real time, very large portions of the plasma membrane, with a resolution that in our setup can reach approximately 30 nm, providing the possibility to have a more complete vision of the entire tissue and handle a large amount of data. Considering the ‘contradiction of available data’, Verkman et al. refer to an FFEM study on OAP structure and organisation performed before the identification of AQP4 (or MIWC) as the molecular determinant of OAPs 2. Moreover, the same study did not directly compare muscle and brain OAP size 2. It was Verkman's group that later performed the first study 3 in which the role of AQP4 in OAP formation and in different tissues was directly analysed by FFEM in AQP4‐WT and null mice. This study literally reported: ‘The density of OAPs in brain was similar to that of OAPs in muscle, however, the patch sizes were somewhat bigger than in muscle
’ 3. Therefore, our interpretation is that our results are rather ‘in line with available data’ with a step forward in which g‐STED has helped to quantify the Verkman group's observation that OAPs in brain are ‘somewhat bigger that in muscle’. Anyhow, it is not of secondary importance that, independently of the size of skeletal muscle OAPs, super‐resolution microscopy revealed that AQP4 sarcolemma organisation in fast‐twitch skeletal muscle fibres is different compared to brain perivascular astrocyte endfeet. A second issue raised by Verkman et al. refers to their own studies in which they have demonstrated that small changes in isoform ratio should not substantially affect NMO‐IgG binding 4. In this case, we have to take into account that those studies have two major limits: (1) they were obtained in heterologous systems in which only two isoforms were over‐expressed; and (2) they were obtained mainly using a recombinant monoclonal antibody, very far from the complexity of real human polyclonal autoantibodies, as demonstrated by epitope mapping studies 5. We believe that the unquestionable advantage of our study is that it has been performed on tissues expressing endogenous AQP4 with all the players (known and unknown) for AQP4 clustering. One of these players is the recently identified AQP4ex isoform 6, which is strongly expressed in skeletal muscle. As AQP4ex modulates AQP4 cluster size, 6 it may, for example, have a role in the different supramolecular organisation. With regard to the concern about the use of non‐fixed frozen tissues in our study, it is well‐established that the use of unfixed tissue for immunofluorescence is crucial to preserve the conformational epitopes necessary for AQP4‐IgG binding 5. While the fascinating mystery of why skeletal muscle is spared in seropositive neuromyelitis optica will certainly benefit from further studies, we believe that a very small piece has been added in this direction here. We would like to thank all involved for the opportunity to continue this fascinating discussion and look forward to hearing from you. Yours sincerely

    Potential role of the methylation of VEGF gene promoter in response to hypoxia in oxygen-induced retinopathy: beneïŹcial effect of the absence of AQP4

    Get PDF
    Hypoxia-dependent accumulation of vascular endothelial growth factor (VEGF) plays a major role in retinal diseases characterized by neovessel formation. In this study, we investigated whether the glial water channel Aquaporin-4 (AQP4) is involved in the hypoxia-dependent VEGF upregulation in the retina of a mouse model of oxygen-induced retinopathy (OIR). The expression levels of VEGF, the hypoxia-inducible factor-1a (HIF-1a) and the inducible form of nitric oxide synthase (iNOS), the production of nitric oxide (NO), the methylation status of the HIF-1 binding site (HBS) in the VEGF gene promoter, the binding of HIF-1a to the HBS, the retinal vascularization and function have been determined in the retina of wild-type (WT) and AQP4 knock out (KO) mice under hypoxic (OIR) or normoxic conditions. In response to 5 days of hypoxia, WT mice were characterized by (i) AQP4 upregulation, (ii) increased levels of VEGF, HIF-1a, iNOS and NO, (iii) pathological angiogenesis as determined by engorged retinal tufts and (iv) dysfunctional electroretinogram (ERG). AQP4 deletion prevents VEGF, iNOS and NO upregulation in response to hypoxia thus leading to reduced retinal damage although in the presence of high levels of HIF-1a. In AQP4 KO mice, HBS demethylation in response to the beginning of hypoxia is lower than in WT mice reducing the binding of HIF-1a to the VEGF gene promoter. We conclude that in the absence of AQP4, an impaired HBS demethylation prevents HIF-1 binding to the VEGF gene promoter and the relative VEGF transactivation, reducing the VEGF-induced retinal damage in response to hypoxia

    Glio-vascular modifications caused by Aquaporin-4 deletion in the mouse retina

    Get PDF
    Aquaporin-4 (AQP4) is the Central Nervous System water channel highly expressed at the perivascular glial domain. In the retina, two types of AQP4 expressing glial cells take part in the blood-retinal barrier (BRB), astrocytes and MĂŒller cells. The aim of the present study is to investigate the effect of AQP4 deletion on the retinal vasculature by looking at typical pathological hallmark such as BRB dysfunction and gliotic condition.AQP4 dependent BRB properties were evaluated by measuring the number of extravasations in WT and AQP4 KO retinas by Evans blue injection assay. AQP4 deletion did not affect the retinal vasculature, as assessed by Isolectin B4 staining, but caused BRB impairment to the deep plexus capillaries while the superficial and intermediate capillaries were not compromised. To investigate for gliotic responses caused by AQP4 deletion, MĂŒller cells and astrocytes were analysed by immunofluorescence and western blot, using the MĂŒller cell marker Glutamine Synthetase (GS) and the astrocyte marker GFAP. While GS expression was not altered in AQP4 KO retinas, a strong GFAP upregulation was found at the level of AQP4 KO astrocytes at the superficial plexus and not at MĂŒller cells at the intermediate and deep plexi. These data, together with the upregulation of inflammatory markers (TNF-α, IL-6, IL-1ÎČ and ICAM-1) in AQP4 KO retinas indicated AQP4 deletion as responsible for a gliotic phenotype. Interestingly, no GFAP altered expression was found in AQP4 siRNA treated astrocyte primary cultures. All together these results indicate that AQP4 deletion is directly responsible for BRB dysfunction and gliotic condition in the mouse retina. The selective activation of glial cells at the primary plexus suggests that different regulatory elements control the reaction of astrocytes and MĂŒller cells. Finally, GFAP upregulation is strictly linked to gliovascular crosstalk, as it is absent in astrocytes in culture. This study is useful to understand the role of AQP4 in the perivascular domain in the retina and its possible implications in the pathogenesis of retinal vascular diseases and of Neuromyelitis Optica, a human disease characterized by anti-AQP4 auto-antibodies

    Direct 3D imaging through spatial coherence of light

    Full text link
    Wide-field imaging is widely adopted due to its fast acquisition, cost-effectiveness and ease of use. Its extension to direct volumetric applications, however, is burdened by the trade-off between resolution and depth of field (DOF), dictated by the numerical aperture of the system. We demonstrate that such trade-off is not intrinsic to wide-field imaging, but stems from the spatial incoherence of light: images obtained through spatially coherent illumination are shown to have resolution and DOF independent of the numerical aperture. This fundamental discovery enabled us to demonstrate an optimal combination of coherent resolution-DOF enhancement and incoherent tomographic sectioning for scanning-free, wide-field 3D microscopy on a multicolor histological section.Comment: 17 pages, 6 figures. Supplemental document available upon request to the authors. Submitted to Lasers and Photonics Review

    The ST2/IL-33 Pathway in Adult and Paediatric Heart Disease and Transplantation

    Get PDF
    ST2 is a member of interleukin 1 receptor family with soluble sST2 and transmembrane ST2L isoforms. The ligand of ST2 is IL-33, which determines the activation of numerous intracytoplasmic mediators following the binding with ST2L and IL-1RAcP, leading to nuclear signal and cardiovascular effect. Differently, sST2 is released in the blood and works as a decoy receptor, binding IL-33 and blocking IL-33/ST2L interaction. sST2 is mainly involved in maintaining homeostasis and/or alterations of different tissues, as counterbalance/activation of IL-33/ST2L axis is typically involved in the development of fibrosis, tissue damage, inflammation and remodeling. sST2 has been described in different clinical reports as a fundamental prognostic marker in patients with cardiovascular disease, as well as marker for the treatment monitoring of patients with heart failure; however, further studies are needed to better elucidate its role. In this review we reported the current knowledge about its role in coronary artery disease, heart failure, heart transplantation, heart valve disease, pulmonary arterial hypertension, and cardiovascular interventions

    Cellular and Molecular Mechanisms Activated by a Left Ventricular Assist Device

    Get PDF
    Left ventricular assist devices (LVADs) represent the final treatment for patients with end-stage heart failure (HF) not eligible for transplantation. Although LVAD design has been further improved in the last decade, their use is associated with different complications. Specifically, inflammation, fibrosis, bleeding events, right ventricular failure, and aortic valve regurgitation may occur. In addition, reverse remodeling is associated with substantial cellular and molecular changes of the failing myocardium during LVAD support with positive effects on patients’ health. All these processes also lead to the identification of biomarkers identifying LVAD patients as having an augmented risk of developing associated adverse events, thus highlighting the possibility of identifying new therapeutic targets. Additionally, it has been reported that LVAD complications could cause or exacerbate a state of malnutrition, suggesting that, with an adjustment in nutrition, the general health of these patients could be improved

    Histamine treatment induces rearrangements of orthogonal arrays of particles (OAPs) in human AQP4-expressing gastric cells

    Get PDF
    To test the involvement of the water channel aquaporin (AQP)-4 in gastric acid physiology, the human gastric cell line (HGT)-1 was stably transfected with rat AQP4. AQP4 was immunolocalized to the basolateral membrane of transfected HGT-1 cells, like in native parietal cells. Expression of AQP4 in transfected cells increased the osmotic water permeability coefficient (Pf) from 2.02 ± 0.3 × 10−4 to 16.37 ± 0.5 × 10−4 cm/s at 20°C. Freeze-fracture EM showed distinct orthogonal arrays of particles (OAPs), the morphological signature of AQP4, on the plasma membrane of AQP4-expressing cells. Quantitative morphometry showed that the density of OAPs was 2.5 ± 0.3% under basal condition and decreased by 50% to 1.2 ± 0.3% after 20 min of histamine stimulation, mainly due to a significant decrease of the OAPs number. Concomitantly, Pf decreased by ∌35% in 20-min histamine-stimulated cells. Both Pf and OAPs density were not modified after 10 min of histamine exposure, time at which the maximal hormonal response is observed. Cell surface biotinylation experiments confirmed that AQP4 is internalized after 20 min of histamine exposure, which may account for the downregulation of water transport. This is the first evidence for short term rearrangement of OAPs in an established AQP4-expressing cell line

    LRRC8A is essential for swelling-activated chloride current and for regulatory volume decrease in astrocytes

    Get PDF
    Consolidated evidence indicates that astroglial cells are critical in the homeostatic regulation of cellular volume by means of ion channels and aquaporin-4. Volume-regulated anion channel (VRAC) is the chloride channel that is activated upon cell swelling and critically contributes to cell volume regulation in astrocytes. The molecular identity of VRAC has been recently defined, revealing that it belongs to the leucine-rich repeat-containing 8 (LRRC8) protein family. However, there is a lack of evidence demonstrating that LRRC8A underpins VRAC currents in astrocyte. Nonetheless, direct evidence of the role of LRRC8A in astrocytic regulatory volume decrease remains to be proved. Here, we aim to bridge this gap in knowledge by combining RNA interference specific for LRRC8A with patch-clamp analyses and a water-permeability assay. We demonstrated that LRRC8A molecular expression is essential for swelling-activated chloride current via VRAC in primary-cultured cortical astrocytes. The knockdown of LRRC8A with a specific short interference RNA abolished the recovery of the cell volume after swelling induced by hypotonic challenge. In addition, immunoblotting, immunofluorescence, confocal imaging, and immunogold electron microscopy demonstrated that LRRC8A is expressed in the plasma membrane of primary cortical astrocytes and in situ in astrocytes at the perivascular interface with endothelial cells. Collectively, our results suggest that LRRC8A is an essential subunit of VRAC and a key factor for astroglial volume homeostasis.-Formaggio, F., Saracino, E., Mola, M. G., Rao, S. B., Amiry-Moghaddam, M., Muccini, M., Zamboni, R., Nicchia, G. P., Caprini, M., Benfenati, V. LRRC8A is essential for swelling-activated chloride current and for regulatory volume decrease in astrocytes

    Role of the H-bond between L53 and T56 for Aquaporin-4 epitope in Neuromyelitis Optica

    Get PDF
    Aquaporin-4 (AQP4) is the CNS water channel organized into well-ordered protein aggregates called Orthogonal Arrays of Particles (OAPs). Neuromyelitis Optica (NMO) is an autoimmune disease caused by anti-OAP autoantibodies (AQP4-IgG). Molecular Dynamics (MD) simulations have identified an H-bond between L53 and T56 as the key for AQP4 epitope and therefore of potential interest for drug design in NMO field. In the present study, we have experimentally tested this MD-prediction using the classic mutagenesis approach. We substituted T56 with V56 and tested this mutant for AQP4 aggregates and AQP4-IgG binding. gSTED super-resolution microscopy showed that the mutation does not affect AQP4 aggregate dimension; immunofluorescence and cytofluorimetric analysis demonstrated its unaltered AQP4-IgG binding, therefore invalidating the MD-prediction. We later investigated whether AQP4, expressed in Sf9 insect and HEK-293F cells, is able to correctly aggregate before and after the purification steps usually applied to obtain AQP4 crystal. The results demonstrated that AQP4-IgG recognizes AQP4 expressed in Sf9 and HEK-293F cells by immunofluorescence even though BN-PAGE analysis showed that AQP4 forms smaller aggregates when expressed in insect cells compared to mammalian cell lines. Notably, after AQP4 purification, from both insect and HEK-293F cells, no aggregates are detectable by BN-PAGE and AQP4-IgG binding is impaired in sandwich ELISA assays. All together these results indicate that 1) the MD prediction under analysis is not supported by experimental data and 2) the procedure to obtain AQP4 crystals might affect its native architecture and, as a consequence, MD simulations. In conclusion, given the complex nature of the AQP4 epitope, MD might not be the suitable for molecular medicine advances in NMO

    AQP4-independent TRPV4 modulation of plasma membrane water permeability

    Get PDF
    : Despite of the major role of aquaporin (AQP) water channels in controlling transmembrane water fluxes, alternative ways for modulating water permeation have been proposed. In the Central Nervous System (CNS), Aquaporin-4 (AQP4) is reported to be functionally coupled with the calcium-channel Transient-Receptor Potential Vanilloid member-4 (TRPV4), which is controversially involved in cell volume regulation mechanisms and water transport dynamics. The present work aims to investigate the selective role of TRPV4 in regulating plasma membrane water permeability in an AQP4-independent way. Fluorescence-quenching water transport experiments in Aqp4-/- astrocytes revealed that cell swelling rate is significantly increased upon TRPV4 activation and in the absence of AQP4. The biophysical properties of TRPV4-dependent water transport were therefore assessed using the HEK-293 cell model. Calcein quenching experiments showed that chemical and thermal activation of TRPV4 overexpressed in HEK-293 cells leads to faster swelling kinetics. Stopped-flow light scattering water transport assay was used to measure the osmotic permeability coefficient (Pf, cm/s) and activation energy (Ea, kcal/mol) conferred by TRPV4. Results provided evidence that although the Pf measured upon TRPV4 activation is lower than the one obtained in AQP4-overexpressing cells (Pf of AQP4 = 0.01667 ± 0.0007; Pf of TRPV4 = 0.002261 ± 0.0004; Pf of TRPV4 + 4αPDD = 0.007985 ± 0.0006; Pf of WT = 0.002249 ± 0.0002), along with activation energy values (Ea of AQP4 = 0.86 ± 0.0006; Ea of TRPV4 + 4αPDD = 2.73 ± 1.9; Ea of WT = 8.532 ± 0.4), these parameters were compatible with a facilitated pathway for water movement rather than simple diffusion. The possibility to tune plasma membrane water permeability more finely through TRPV4 might represent a protective mechanism in cells constantly facing severe osmotic challenges to avoid the potential deleterious effects of the rapid cell swelling occurring via AQP channels
    • 

    corecore