14 research outputs found

    Heme oxygenase-1 is required for angiogenic function of bone marrow-derived progenitor cells : role in therapeutic revascularization

    Get PDF
    Aims: Heme oxygenase-1 (HO-1) is a cytoprotective enzyme that can be down-regulated in diabetes. Its importance for mature endothelium has been described, but its role in proangiogenic progenitors is not well known. We investigated the effect of HO-1 on the angiogenic potential of bone marrow-derived cells (BMDCs) and on blood flow recovery in ischemic muscle of diabetic mice. Results: Lack of HO-1 decreased the number of endothelial progenitor cells (Lin−CD45−cKit-Sca-1+VEGFR-2+) in murine bone marrow, and inhibited the angiogenic potential of cultured BMDCs, affecting their survival under oxidative stress, proliferation, migration, formation of capillaries, and paracrine proangiogenic potential. Transcriptome analysis of HO-1−/− BMDCs revealed the attenuated up-regulation of proangiogenic genes in response to hypoxia. Heterozygous HO-1+/− diabetic mice subjected to hind limb ischemia exhibited reduced local expression of vascular endothelial growth factor (VEGF), placental growth factor (PlGF), stromal cell-derived factor 1 (SDF-1), VEGFR-1, VEGFR-2, and CXCR-4. This was accompanied by impaired revascularization of ischemic muscle, despite a strong mobilization of bone marrow-derived proangiogenic progenitors (Sca-1+CXCR-4+) into peripheral blood. Blood flow recovery could be rescued by local injections of conditioned media harvested from BMDCs, but not by an injection of cultured BMDCs. Innovation: This is the first report showing that HO-1 haploinsufficiency impairs tissue revascularization in diabetes and that proangiogenic in situ response, not progenitor cell mobilization, is important for blood flow recovery. Conclusions: HO-1 is necessary for a proper proangiogenic function of BMDCs. A low level of HO-1 in hyperglycemic mice decreases restoration of perfusion in ischemic muscle, which can be rescued by a local injection of conditioned media from cultured BMDCs

    Physiopathologie et thérapie génique de la neuropathie optique associée au Syndrome de Wolfram

    No full text
    Wolfram Syndrome (WS; OMIM #222300, prevalence 1-9 / 1 000 000) has a juvenile onset and incorporates diabetes insipidus, diabetes mellitus, optic atrophy (OA), and deafness; leading to death in middle age. OA is its first neurological symptom, starting in adolescence and ending with blindness within 8 years. Unfortunately, a suitable WS mouse model comprising ophthalmologic symptoms has not yet been found, therefore the search for its treatment is delayed. In this thesis, I studied visual impairment in two WS mouse models along with a success of a gene therapy (GT) approach with the human WFS1 gene.Firstly, 3 and 6 months old Wfs1exon8del mice were examined for the visual acuity (VA) and contrast sensitivity via changes in the opto-motor reflex (OMR), the neural retinal function via electroretinogram (ERG), as well as the eye physiology via fundoscopy and optic coher-ence tomography (OCT). Also, the proportion of retinal ganglion cells (RGC) and the axonal loss at the age of 7 months were determined with anti-Brn3a immuno-labeling of retinal sections and electron microscopy of optic nerve (ON) sections, respectively. There was a progressive loss of VA and contrast sensitivity in Wfs1exon8del-/- mice, starting already at 1 month of age. It was accompanied by optic disc pallor, retinal thinning as well as axonal damage. However, there was no RGC loss and the endoplasmic reticulum (ER) stress in the retina was at a normal level. It suggested a presence of another cause for the reported degeneration in KO mice; in opposition to what was proposed in the literature. I brief, KO mice exhibit significant WS ophthalmic phenotype.Secondly, in search for another model, visual functions of Wfs1E864K mouse line were investigated. This line was originally a model of Wolfram-like Syndrome, characterized by dominant mutations in WFS1 leading to congenital progressive hearing impairment, diabetes mellitus and OA. Only homozygous mutants, however, showed expected visual impairment. Already at 1 month of age, Wfs1E864K/E864K mice had drastic loss of RGC function, albeit keeping the cell number at a normal level. This was accompanied by retinal thinning and a severe ON damage, as shown with OCT and fundoscopy, respectively. In contrast, the RGC function in Wfs1E864K/+ mice dropped slightly only at the age of 7 and 12 months, showing that the pathology of the E864K mutation-driven disease in mice is different than in humans. Therefore, Wfs1E864K/E864K mice, with their strong ophthalmic phenotype, could potentially serve as a model of the classical WS.Finally, to investigate future treatment options, 1 month old Wfs1exon8del+/+ (WT) and Wfs1exon8del-/- (KO) mice underwent a uni- and bi-lateral intravitreal gene therapy (GT) with AAV-2/2-CMV-WFS1. Exams at 3 and 6 months of age showed improved VA, as well as optic pallor and axonal damage rescue in KO mice. Also, no adverse effects related to either GT or sham injections were noted. Following this idea, the Wfs1E864K/E864K mice were also subjected to intravitreal GT, delivered at P14, but without success.In conclusion, Wfs1exon8del mouse line is a reliable model of WS, including the visual aspects. I propose the Wfs1E864K/E864K model as an alternative, especially to investigate Wfs1 function in the eye. Finally, the intravitreal AAV-driven GT with WFS1 has a potential to partially rescue the ophthalmic phenotype, paving the wave towards the treatment for WS patients.Le Syndrome de Wolfram (SW; OMIM #222300, prĂ©valence 1-9 / 1000 000) est une maladie neurodĂ©gĂ©nĂ©rative, qui se prĂ©sente avec un dĂ©but juvĂ©nile, intĂ©grant le diabĂšte insipide, diabĂšte sucrĂ©, l’atrophie optique (AO), et la surditĂ©. AO est gĂ©nĂ©ralement son premier symptĂŽme neurologique, commençant Ă  l’ñge de 11 ans et se terminant par la cĂ©citĂ© 8 ans plus tard. Malheureusement, un modĂšle murin du SW appropriĂ© aux symptĂŽmes ophtalmologiques n'a pas encore Ă©tĂ© trouvĂ©, donc la recherche de la thĂ©rapie pour sauver la vision en est Ă  ces dĂ©buts. Dans cette thĂšse, j’ai Ă©tudiĂ© l’atteinte visuelle de deux modĂšles de souris mutantes pour le SW et succĂšs d’une approche de thĂ©rapie gĂ©nique (TG) avec le gĂšne humain WFS1.PremiĂšrement, les souris Wfs1exon8del sont Ă©tĂ© examinĂ©es Ă  3 et 6 mois pour l’acuitĂ© visuelle (AV) et la sensibilitĂ© aux contrastes (SC) via changements dans le reflexe optomoteur (ROM), la fonction rĂ©tinienne neurale par Ă©lectrorĂ©tinogramme (ERG), ainsi que la physiologie de l’Ɠil par la fondoscopie et tomographie par cohĂ©rence optique (TCO). De plus, la proportion des cellules ganglionnaire de la rĂ©tine (CGRs) et la perte axonale dans le nerf optique (NO) Ă  7 mois ont Ă©tĂ© examinĂ©s avec marquage anti-Brn3a et microscopie Ă©lectronique, respectivement. Il y avait une perte progressive de l’AV et la SC chez les souris KO Ă  partir du 1 mois. Elle Ă©tait accompagnĂ©e d'une pĂąleur du disque optique (DO), d'amincissement de la rĂ©tine ainsi que des lĂ©sions axonales. Par contre, il n’avait pas de perte des CGRs ni stress du rĂ©ticulum endoplasmique dans la rĂ©tine. BriĂšvement, les souris KO prĂ©sentent un phĂ©notype ophtalmique du SW significatif et peuvent servir comme modĂšle.DeuxiĂšmement, Ă  la recherche d'un autre modĂšle du SW, les fonctions visuelles de la lignĂ©e Wfs1E864K de la souris ont Ă©tĂ© Ă©tudiĂ©es. DĂ©jĂ  Ă  1 mois, les souris Wfs1E864K/E864K avait une perte drastique de la fonction des CGRs, mais en gardant le nombre de cellules Ă  un niveau normal. Ceci a Ă©tĂ© accompagnĂ© par un amincissement de la rĂ©tine et d’un sĂ©vĂšre dommage du NO, comme montrĂ© par le TCO et la fondoscopie, respectivement. En consĂ©quence, les souris Wfs1E864K/E864K, avec leur fort phĂ©notype ophtalmique, pourraient servir comme modĂšle du SW classique.Enfin, pour enquĂȘter sur les futures options de traitement contre le SW, les souris de la lignĂ©e Wfs1exon8del Ă  1 mois ont subi une TG intravitrĂ©enne avec AAV-2/2-CMV-WFS1. Les examens Ă  3 et 6 mois ont montrĂ© une amĂ©lioration de l’AV, ainsi que le sauvetage de la pĂąleur du DO et rĂ©duction des lĂ©sions axonales chez les souris KO. En outre, aucun effet indĂ©sirable liĂ© Ă  des injections TG n’ont Ă©tĂ© notĂ©. Suivant cette idĂ©e, les souris Wfs1E864K/E864K ont Ă©galement Ă©tĂ© soumis Ă  la TG intravitrĂ©enne, dĂ©livrĂ©e Ă  P14, mais sans succĂšs.En conclusion, la lignĂ©e Wfs1exon8del de la souris est un modĂšle fiable du SW, y compris les aspects visuels. Je propose le modĂšle Wfs1E864K/E864K comme une alternative, en particulier pour enquĂȘter sur la fonction de Wfs1 dans l'Ɠil. Enfin, la GT intravitrĂ©enne avec WFS1 a un potentiel pour sauver partiellement le phĂ©notype ophtalmique, ouvrant la voie vers le traitement pour les patients du S

    Optical Coherence Tomography: Imaging Mouse Retinal Ganglion Cells <em>In Vivo</em>

    No full text
    International audienceStructural changes in the retina are common manifestations of ophthalmic diseases. Optical coherence tomography (OCT) enables their identification in vivo-rapidly, repetitively, and at a high resolution. This protocol describes OCT imaging in the mouse retina as a powerful tool to study optic neuropathies (OPN). The OCT system is an interferometry-based, non-invasive alternative to common post mortem histological assays. It provides a fast and accurate assessment of retinal thickness, allowing the possibility to track changes, such as retinal thinning or thickening. We present the imaging process and analysis with the example of the Opa1delTTAG mouse line. Three types of scans are proposed, with two quantification methods: standard and homemade calipers. The latter is best for use on the peripapillary retina during radial scans; being more precise, is preferable for analyzing thinner structures. All approaches described here are designed for retinal ganglion cells (RGC) but are easily adaptable to other cell populations. In conclusion, OCT is efficient in mouse model phenotyping and has the potential to be used for the reliable evaluation of therapeutic interventions

    Pre-emptive hypoxia-regulated HO-1 gene therapy improves post-ischaemic limb perfusion and tissue regeneration in mice

    Get PDF
    Aims Haem oxygenase-1 (HO-1) is a haem-degrading enzyme that generates carbon monoxide, bilirubin, and iron ions. Through these compounds, HO-1 mitigates cellular injury by exerting antioxidant, anti-apoptotic, and anti-inflammatory effects. Here, we examined the influence of HO-1 deficiency and transient hypoxia/ischaemia-induced HO-1 overexpression on post-injury hindlimb recovery. Methods and results Mice lacking functional HO-1 (HO-1−/−) showed reduced reparative neovascularization in ischaemic skeletal muscles, impaired blood flow (BF) recovery, and increased muscle cell death compared with their wild-type littermates. Human microvascular endothelial cells (HMEC-1) transfected with plasmid vector (pHRE-HO-1) carrying human HO-1 driven by three hypoxia response elements (HREs) and cultured in 0.5% oxygen demonstrated markedly increased expression of HO-1. Such upregulated HO-1 levels were effective in conferring protection against H2O2-induced cell death and in promoting the proangiogenic phenotype of HMEC-1 cells. More importantly, when delivered in vivo, pHRE-HO-1 significantly improved the post-ischaemic foot BF in mice subjected to femoral artery ligation. These effects were associated with reduced levels of pro-inflammatory cytokines (IL-6 and CXCL1) and lower numbers of transferase-mediated dUTP nick-end labelling-positive cells. Moreover, HO-1 delivered into mouse skeletal muscles seems to influence the regenerative potential of myocytes as it significantly changed the expression of transcriptional (Pax7, MyoD, myogenin) and post-transcriptional (miR-146a, miR-206) regulators of skeletal muscle regeneration. Conclusion Our results suggest the therapeutic potential of HO-1 for prevention of adverse effects in critical limb ischaemia.</p

    Impairment of visual function and retinal ER stress activation in Wfs1-deficient mice

    Get PDF
    Wolfram syndrome is an early onset genetic disease (1/180,000) featuring diabetes mellitus and optic neuropathy, associated to mutations in the WFS1 gene. Wfs1-/- mouse model shows pancreatic beta cell atrophy, but its visual performance has not been investigated, prompting us to study its visual function and histopathology of the retina and optic nerve. Electroretinogram and visual evoked potentials (VEPs) were performed in Wfs1-/- and Wfs1+/+ mice at 3, 6, 9 and 12 months of age. Fundi were pictured with Micron III apparatus. Retinal ganglion cell (RGC) abundance was determined from Brn3a immunolabeling of retinal sections. RGC axonal loss was quantified by electron microscopy in transversal optic nerve sections. Endoplasmic reticulum stress was assessed using immunoglobulin binding protein (BiP), protein disulfide isomerase (PDI) and inositol-requiring enzyme 1 alpha (Ire1α) markers. Electroretinograms amplitudes were slightly reduced and latencies increased with time in Wfs1-/- mice. Similarly, VEPs showed decreased N+P amplitudes and increased N-wave latency. Analysis of unfolded protein response signaling revealed an activation of endoplasmic reticulum stress in Wfs1-/- mutant mouse retinas. Altogether, progressive VEPs alterations with minimal neuronal cell loss suggest functional alteration of the action potential in the Wfs1-/- optic pathways

    Retinal ER stress and UPR evaluation in <i>Wfs1<sup>−/−</sup></i> mice.

    No full text
    <p>Immunoblots (A) detected GRP78/BiP, PDI, IRE1, and beta actin in protein lysates of 12 month old <i>Wfs1<sup>+/+</sup></i> (n = 3) and <i>Wfs1<sup>−/−</sup></i> (n = 3) mouse retinas and in mouse NIH3T3 fibroblasts treated with thapsigargin. Mean relative quantities for each protein according to <i>Wfs1</i> genotype were obtained after normalization with beta actin values. Significance (*) is indicated when p<0.05.</p

    RGC cell density in <i>Wfs1</i><sup>−/−</sup> mice.

    No full text
    <p>Brn3a and total nuclei were counted in nerve fiber layer of 12 month retinal sections (A). RGC cell density is given as average Brn3a cell nuclei in 1000 pixel segments of retina (B). <i>Thy1</i> and <i>Nrn1</i> relative gene expressions were quantified in retinal tissue at 3 (n = 4 <i>Wfs1</i><sup>+/+</sup>, n = 4 <i>Wfs1</i><sup>−/−</sup>), 7 (n = 5 <i>Wfs1</i><sup>+/+</sup>, n = 7 <i>Wfs1</i><sup>−/−</sup>) and 12 months (n = 8 <i>Wfs1</i><sup>+/+</sup>, n = 8 <i>Wfs1</i><sup>−/−</sup>) (C). Average values are presented +/− SEM, with statistical significance (*) indicated if p<0.05.</p

    Photoreceptor and inner retinal functions in <i>Wfs1</i><sup>−/−</sup> mice.

    No full text
    <p>Photoreceptor and inner retina electroretinogram data are presented in panels A and B, from ERG a- and b-wave respectively, on <i>Wfs1</i><sup>+/+</sup> and <sup>−/−</sup> mice. Both waves are represented by mean values of amplitudes and latencies at the ages of 3 (n = 10 <i>Wfs1</i><sup>+/+</sup>, n = 8 <i>Wfs1</i><sup>−/−</sup>), 6 (n = 8 <i>Wfs1</i><sup>+/+</sup>, n = 7 <i>Wfs1</i><sup>−/−</sup>), 9 (n = 12 <i>Wfs1</i><sup>+/+</sup>, n = 14 <i>Wfs1</i><sup>−/−</sup>) and 12 months (n = 10 <i>Wfs1</i><sup>+/+</sup>, n = 10 <i>Wfs1</i><sup>−/−</sup>) in scotopic and mesopic conditions. Black and red traces correspond to <i>Wfs1<sup>+/+</sup></i> and <i>Wfs1<sup>−/−</sup></i> animals respectively. Statistical significance is indicated when p<0.05 (*) and 0.01 (**).</p

    Light-induced electric conduction from retina to visual cortex.

    No full text
    <p>Visual evoked potentials measured from 3 to 12 months are depicted as average +/− SEM of N+P amplitudes (A) and N- and P-wave latencies (B). Black circles and red squares correspond respectively to <i>Wfs1</i><sup>+/+</sup> and <i>Wfs1</i><sup>−/−</sup> animals. Statistical significance is indicated when p<0.05 (*) and 0.01 (**). For 3 and 6 months n = 9 for <i>Wfs1</i><sup>+/+</sup> and <i>Wfs1</i><sup>−/−</sup>, for 9 months n = 13 <i>Wfs1</i><sup>+/+</sup> and n = 20 <i>Wfs1</i><sup>−/−</sup>, for 12 months n = 14 <i>Wfs1</i><sup>+/+</sup> and n = 9 <i>Wfs1</i><sup>−/−</sup>.</p
    corecore