80 research outputs found

    The fate of heterotopically grafted neural precursor cells in the normal and dystrophic adult mouse retina

    Get PDF
    PURPOSE. To study the integration and differentiation of heterotopically transplanted neural precursor cells in the retina of adult mouse mutants displaying apoptotic degeneration of photoreceptor cells. METHODS. Neural precursor cells were isolated from the spinal cord of transgenic mouse embryos ubiquitously expressing enhanced green fluorescent protein. Cells were expanded in vitro and transplanted into the retina of adult wild-type and age-matched ␀2/␀1 knock-in mice. ␀2/␀1 knock-in mutants display apoptotic death of photoreceptor cells and were generated by placing the cDNA of the ␀1 subunit into the gene of the ␀2 subunit of Na,K-ATPase. The integration and differentiation of grafted cells in recipient retinas was studied 1 or 6 months after transplantation. RESULTS. Mutant retinas contained more donor-derived cells than wild-type hosts. Moreover, in mutants, donor cells integrated into deeper retinal layers. In both genotypes, grafted cells differentiated into astrocytes and oligodendrocytes. Only a few ganglion cell axons were myelinated by donor-derived oligodendrocytes 1 month after transplantation, whereas extensive myelination of the nerve fiber layer was observed 6 months after transplantation. Unequivocal evidence for differentiation of grafted cells into neurons was not obtained. CONCLUSIONS. Heterotopically transplanted neural precursor cells are capable of integrating, surviving, and differentiating into neural cell types in normal and dystrophic retinas of adult mice. The particular environment of a pathologically altered retina facilitates integration of transplanted precursor cells. In principle, neural precursors may thus be useful to substitute for or replace dysfunctional or degenerated cell types. Results of the present study also indicate that replacement of retinal cell types is likely to require more appropriate donor cells, such as retinal precursor cells. (Invest Ophthalmol Vis Sci. 2001;42:3311-3319) I nherited retinal dystrophies are a heterogeneous group of disorders characterized by progressive retinal degeneration. Effective therapeutic treatments of retinal dystrophies in humans are currently not available. However, animal experiments have shown beneficial effects of various therapeutic strategies, including gene therapy to substitute for the pathogenic gene, application of growth factors to minimize cell degeneration, or transplantation of committed cell types to replace dysfunctional or degenerated cells. 10 -12 Neural precursors have been isolated from the developing and adult brain and can be massively expanded in vitro, providing, in principle, unlimited amounts of cell material for transplantation (different from primary retinal cells). When transplanted into the developing or adult brain, they have been demonstrated to integrate extensively into the recipient tissue, to survive for extended periods, and to eventually differentiate into those cell types that are affected in the host. 22 These cells were expanded in vitro in the presence of mitogens and subsequently transplanted into the retina of adult wild-type mice and mouse mutants displaying apoptotic degeneration of photoreceptor cells. As a mutant host, we used ␀2/␀1 knock-in mice. 23 ␀1 and ␀2 are subunits of Na,K-ATPase, a heterodimeric ion pump additionally consisting of a catalytic ␣-subunit. 23,24 ␀-subunits play a pivotal role for the formation of functional Na,K-ATPases as exemplified, for instance, by the severe phenotype of ␀2-deficient mice. 25 Such mice display a variety of severe defects in the central nervous system (CNS), including massive apoptotic degeneration of photoreceptor cells, and die at the end of the third postnatal week. 23 To obtain information about the fate of heterotopically transplanted neural precursor cells in the normal and dystrophic mouse retina, we isolated such cells from the spinal cord of EGFP transgenic mice and transplanted them into the retinas of adult wild-type mice and ␀2/␀1 knock-in mutants. Heterotopically transplanted neural precursor cells integrated into the mutant retina without disrupting the histoarchitecture of the host tissue. Quantitative investigations revealed that donorderived cells were more numerous and more widely distributed in mutant retinas than in retinas of age-matched wild-type From th

    Stem Cell-Derived Photoreceptor Transplants Differentially Integrate Into Mouse Models of Cone-Rod Dystrophy

    Get PDF
    Citation: Santos-Ferreira T, Völkner M, Borsch O, et al. Stem cell-derived photoreceptor transplants differentially integrate into mouse models of cone-rod dystrophy. Invest Ophthalmol Vis Sci. 2016;57:3509-3520. DOI:10.1167/iovs.16-19087 PURPOSE. Preclinical studies on photoreceptor transplantation provided evidence for restoration of visual function with pluripotent stem cells considered as a potential source for sufficient amounts of donor material. Adequate preclinical models representing retinal disease conditions of potential future patients are needed for translation research. Here we compared transplant integration in mouse models with mild (prominin1-deficient; Prom1 METHODS. For photoreceptor transplant production, we combined the mouse embryonic stem cell retinal organoid system with rhodopsin-driven GFP cell labeling by recombinant adenoassociated virus (AAV). Organoid-derived photoreceptors were enriched by CD73-based magnetic-activated cell sorting (MACS) and transplanted subretinally into wild-type, Prom1 and Cpfl1/Rho À/À hosts. The survival, maturation, and synapse formation of donor cells was analyzed by immunohistochemistry. RESULTS. Retinal organoids yielded high photoreceptor numbers that were further MACSenriched to 85% purity. Grafted photoreceptors survived in the subretinal space of all mouse models. Some cells integrated into wild-type as well as Prom1 À/À mouse retinas and acquired a mature morphology, expressing rod and synaptic markers in close proximity to secondorder neurons. In contrast, in the novel Cpfl1/Rho À/À model with complete photoreceptor degeneration, transplants remained confined to the subretinal space, expressed rod-specific but only reduced synaptic markers, and did not acquire mature morphology. CONCLUSIONS. Comparison of photoreceptor grafts in preclinical models with incomplete or complete photoreceptor loss, showed differential transplant success with effective and impaired integration, respectively. Thus, Cpfl1/Rho À/À mice represent a potential benchmark model resembling patients with severe retinal degeneration to optimize photoreceptor replacement therapies

    Large-Area RPE Removal by Microsecond Laser followed by hiPS-RPE transplantation

    Get PDF
    Cell therapeutics for AMD were often implanted regardless of RPE status in the target zone. This may result in RPE multilayering. Here we study a novel laser to remove RPE without collateral damage prior to RPE implantation to encourage better subretinal integration. Pigment rabbits (n=24) were immunosuppressed with Sirolimus, Doxycyclin and Minocyclin. Using a SLO/ OCT (Heidelberg Engineering) extended with a prototype laser (Meridian Medical; wavelength: 532 nm; pulse duration, 8 ”s), a large area of RPE was selectively removed in 19 rabbits. Animals without laser lesions served as controls (n=5). A 25 gauge vitrectomy (Geuder) with removal of posterior hyaloid membrane was performed thereafter. Human iPS-RPE (1000 cells/ ”l) were manually injected using a 100 ”l syringe (Hamilton) connected to a 38G cannula (MedOne) into the RPE laser lesion, or over healthy RPE in controls, monitored by intraoperative OCT imaging (RESCAN 700, Zeiss). In vivo follow up/ retinal imaging was up to 12 weeks including fluorescein and indocyanine angiography, as well as SD-OCT (Spectralis Ÿ, Heidelberg Engineering). Representative RPE laser wounds exhibited mild late phase FA& ICGA leakage, without abnormal outer retinal or choroidal hyperreflectivity on OCT. By contrast, lesions with earlier leakage on FA/ ICGA showed beam-sized outer retinal hyperreflectivity on OCT, suggesting coagulation. The size of the RPE wounds was typically 10-12mm2.iOCT demonstrated in an immediate and directed spread of the bleb retinal detachment (bRD) within the lasered zone. By contrast, bRDs performed over non-lasered RPE raised slower with a circular spread. Subretinal injection ranged from 5-70”l, with lesser volumes/ larger bRDs areas over lasered regions.At 6 and 12 weeks, none of implanted regions showed FA/ICGA leakage, some lesions had blockage due to hyperpigmentation; on OCT, representative areas showed preserved ellipsoid bands, with some RPE undulations. Lasered/implanted areas with a peripheral hyperpigmentation showed central outer retinal atrophy along with irregular RPE. Control implantation sites showed retinal atrophy and a variably thickened RPE band. Large-area RPE removal with laser disruption is feasible in healthy rabbits and appears to facilitate superior integration of RPE suspension grafts, compared to subretinal injection alone. Future work aims to correlate histology with in vivo imaging. This abstract was presented at the 2022 ARVO Annual Meeting, held in Denver, CO, May 1-4, 2022, and virtually

    Short-term follow up after Large-Area RPE Removal by Microsecond Laser followed by hiPS-RPE suspension transplantation in rabbits

    Get PDF
    Cell therapy is a promising treatment for retinal pigment epithelium (RPE)-associated eye diseases. Herein, microsecond laser irradiation targeting RPE cells was used for large-area RPE removal followed by subretinal injection of human induced pluripotent stem cell derived RPE (hiPS-RPE). 19 immunosuppressed pigmented rabbits (Chinchilla bastard hybrid) underwent a large area RPE removal using an infrared reflectance (IR) confocal scanning laser ophthalmoscope (cSLO) with spectral-domain optical coherence tomography (SD-OCT) system (Heidelberg Engineering ) extended with a prototype laser (modified Merilas 532 shortpulse ophthalmic laser photocoagulator, Meridian Medical) (wavelength, 532 nm; pulse duration, 8 ”s), followed by a 25G vitrectomy. Subsequently, a suspension of hiPS-RPE (1000 cells/ ”l) was grafted subretinally into the RPE laser lesion under real-time intraoperative OCT imaging (RESCAN 700, Zeiss) by manual injection via a 25/38G cannula connected to a 100”l Hamilton syringe. 5 rabbits served as a control with hiPS-RPE injected subretinally over healthy RPE. The rabbits were followed with in vivo multimodal retinal imaging at baseline after laser and then for 7 days including fluorescein (FA) and indocyanine angiography (ICGA), aw well as SD-OCT (Spectralis Ÿ, Heidelberg Engineering). Baseline imaging of RPE laser wounds showed mild late phase FA/ICGA leakage, with normal outer retinal and choroidal reflectivity on OCT, without signs of coagulation. The size of the RPE wounds was typically 10-12mm2. Real time iOCT showed a directed spread of the bleb retinal detachment (bRD) within the lasered zone, in contrast to a circular spread in controls. Subretinal injection ranged from 5-20”l, with lesser volumes/ larger bRD areas over lasered regions. At 7 days, implanted regions showed FA/ICGA leakage, blockage due to hyperpigmentation was observed mostly at the edges of the lasered zone; OCT showed hyperreflectivity of the outer retina with RPE irregularities. Control implantation sites showed hyperreflectivity in all retinal layers and a variably thickened RPE band suggesting clumping. Microsecond laser irradiation to the RPE seems to accelerate the subretinal integration of hiPS-RPE, when compared to subretinal injection over intact RPE. Future work will address correlation of multimodal imaging and histology. This abstract was presented at the 2022 ARVO Annual Meeting, held in Denver, CO, May 1-4, 2022, and virtually

    In Vivo Analysis of Disease-Associated Point Mutations Unveils Profound Differences in mRNA Splicing of Peripherin-2 in Rod and Cone Photoreceptors

    Get PDF
    Point mutations in peripherin-2 (PRPH2) are associated with severe retinal degenerative disorders affecting rod and/or cone photoreceptors. Various disease-causing mutations have been identified, but the exact contribution of a given mutation to the clinical phenotype remains unclear. Exonic point mutations are usually assumed to alter single amino acids, thereby influencing specific protein characteristics;however, they can also affect mRNA splicing. To examine the effects of distinct PRPH2 point mutations on mRNA splicing and protein expression in vivo, we designed PRPH2 minigenes containing the three coding exons and relevant intronic regions of human PRPH2. Minigenes carrying wild type PRPH2 or PRPH2 exon 2 mutations associated with rod or cone disorders were expressed in murine photoreceptors using recombinant adeno-associated virus (rAAV) vectors. We detect three PRPH2 splice isoforms in rods and cones: correctly spliced, intron 1 retention, and unspliced. In addition, we show that only the correctly spliced isoform results in detectable protein expression. Surprisingly, compared to rods, differential splicing leads to lower expression of correctly spliced and higher expression of unspliced PRPH2 in cones. These results were confirmed in qRT-PCR experiments from FAC-sorted murine rods and cones. Strikingly, three out of five cone disease-causing PRPH2 mutations profoundly enhanced correct splicing of PRPH2, which correlated with strong upregulation of mutant PRPH2 protein expression in cones. By contrast, four out of six PRPH2 mutants associated with rod disorders gave rise to a reduced PRPH2 protein expression via different mechanisms. These mechanisms include aberrant mRNA splicing, protein mislocalization, and protein degradation. Our data suggest that upregulation of PRPH2 levels in combination with defects in the PRPH2 function caused by the mutation might be an important mechanism leading to cone degeneration. By contrast, the pathology of rod-specific PRPH2 mutations is rather characterized by PRPH2 downregulation and impaired protein localization

    Imaging of nanoparticle-labeled stem cells using magnetomotive optical coherence tomography, laser speckle reflectometry, and light microscopy

    Get PDF
    Cell transplantation and stem cell therapy are promising approaches for regenerative medicine and are of interest to researchers and clinicians worldwide. However, currently, no imaging technique that allows three-dimensional in vivo inspection of therapeutically administered cells in host tissues is available. Therefore, we investigate magnetomotive optical coherence tomography (MM-OCT) of cells labeled with magnetic particles as a potential noninvasive cell tracking method. We develop magnetomotive imaging of mesenchymal stem cells for future cell therapy monitoring. Cells were labeled with fluorescent iron oxide nanoparticles, embedded in tissue-mimicking agar scaffolds, and imaged using a microscope setup with an integrated MM-OCT probe. Magnetic particle-induced motion in response to a pulsed magnetic field of 0.2 T was successfully detected by OCT speckle variance analysis, and cross-sectional and volumetric OCT scans with highlighted labeled cells were obtained. In parallel, fluorescence microscopy and laser speckle reflectometry were applied as two-dimensional reference modalities to image particle distribution and magnetically induced motion inside the sample, respectively. All three optical imaging modalities were in good agreement with each other. Thus, magnetomotive imaging using iron oxide nanoparticles as cellular contrast agents is a potential technique for enhanced visualization of selected cells in OCT

    Reliability of human retina organoid generation from hiPSC-derived neuroepithelial cysts

    Get PDF
    The possible applications for human retinal organoids (HROs) derived from human induced pluripotent stem cells (hiPSC) rely on the robustness and transferability of the methodology for their generation. Standardized strategies and parameters to effectively assess, compare, and optimize organoid protocols are starting to be established, but are not yet complete. To advance this, we explored the efficiency and reliability of a differentiation method, called CYST protocol, that facilitates retina generation by forming neuroepithelial cysts from hiPSC clusters. Here, we tested seven different hiPSC lines which reproducibly generated HROs. Histological and ultrastructural analyses indicate that HRO differentiation and maturation are regulated. The different hiPSC lines appeared to be a larger source of variance than experimental rounds. Although previous reports have shown that HROs in several other protocols contain a rather low number of cones, HROs from the CYST protocol are consistently richer in cones and with a comparable ratio of cones, rods, and MĂŒller glia. To provide further insight into HRO cell composition, we studied single cell RNA sequencing data and applied CaSTLe, a transfer learning approach. Additionally, we devised a potential strategy to systematically evaluate different organoid protocols side-by-side through parallel differentiation from the same hiPSC batches: In an explorative study, the CYST protocol was compared to a conceptually different protocol based on the formation of cell aggregates from single hiPSCs. Comparing four hiPSC lines showed that both protocols reproduced key characteristics of retinal epithelial structure and cell composition, but the CYST protocol provided a higher HRO yield. So far, our data suggest that CYST-derived HROs remained stable up to at least day 200, while single hiPSC-derived HROs showed spontaneous pathologic changes by day 200. Overall, our data provide insights into the efficiency, reproducibility, and stability of the CYST protocol for generating HROs, which will be useful for further optimizing organoid systems, as well as for basic and translational research applications

    Efficacy and safety of baricitinib in hospitalized adults with severe or critical COVID‑19 (Bari‑SolidAct): a randomised, double‑blind, placebo‑controlled phase 3 trial

    Get PDF
    Background Baricitinib has shown efcacy in hospitalized patients with COVID-19, but no placebo-controlled trials have focused specifcally on severe/critical COVID, including vaccinated participants. Methods Bari-SolidAct is a phase-3, multicentre, randomised, double-blind, placebo-controlled trial, enrolling participants from June 3, 2021 to March 7, 2022, stopped prematurely for external evidence. Patients with severe/ critical COVID-19 were randomised to Baricitinib 4 mg once daily or placebo, added to standard of care. The primary endpoint was all-cause mortality within 60 days. Participants were remotely followed to day 90 for safety and patient related outcome measures. Results Two hundred ninety-nine patients were screened, 284 randomised, and 275 received study drug or placebo and were included in the modifed intent-to-treat analyses (139 receiving baricitinib and 136 placebo). Median age was 60 (IQR 49–69) years, 77% were male and 35% had received at least one dose of SARS-CoV2 vaccine. There were 21 deaths at day 60 in each group, 15.1% in the baricitinib group and 15.4% in the placebo group (adjusted absolute diference and 95% CI −0.1% [−8·3 to 8·0]). In sensitivity analysis censoring observations after drug discontinuation or rescue therapy (tocilizumab/increased steroid dose), proportions of death were 5.8% versus 8.8% (−3.2% [−9.0 to 2.7]), respectively. There were 148 serious adverse events in 46 participants (33.1%) receiving baricitinib and 155 in 51 participants (37.5%) receiving placebo. In subgroup analyses, there was a potential interaction between vaccination status and treatment allocation on 60-day mortality. In a subsequent post hoc analysis there was a signifcant interac‑ tion between vaccination status and treatment allocation on the occurrence of serious adverse events, with more respiratory complications and severe infections in vaccinated participants treated with baricitinib. Vaccinated partici‑ pants were on average 11 years older, with more comorbidities. Conclusion This clinical trial was prematurely stopped for external evidence and therefore underpowered to con‑ clude on a potential survival beneft of baricitinib in severe/critical COVID-19. We observed a possible safety signal in vaccinated participants, who were older with more comorbidities. Although based on a post-hoc analysis, these fnd‑ ings warrant further investigation in other trials and real-world studies

    Efficacy and safety of baricitinib in hospitalized adults with severe or critical COVID-19 (Bari-SolidAct): a randomised, double-blind, placebo-controlled phase 3 trial

    Get PDF
    © The Author(s) 2023. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.[Background] Baricitinib has shown efficacy in hospitalized patients with COVID-19, but no placebo-controlled trials have focused specifically on severe/critical COVID, including vaccinated participants.[Methods] Bari-SolidAct is a phase-3, multicentre, randomised, double-blind, placebo-controlled trial, enrolling participants from June 3, 2021 to March 7, 2022, stopped prematurely for external evidence. Patients with severe/critical COVID-19 were randomised to Baricitinib 4 mg once daily or placebo, added to standard of care. The primary endpoint was all-cause mortality within 60 days. Participants were remotely followed to day 90 for safety and patient related outcome measures.[Results] Two hundred ninety-nine patients were screened, 284 randomised, and 275 received study drug or placebo and were included in the modified intent-to-treat analyses (139 receiving baricitinib and 136 placebo). Median age was 60 (IQR 49–69) years, 77% were male and 35% had received at least one dose of SARS-CoV2 vaccine. There were 21 deaths at day 60 in each group, 15.1% in the baricitinib group and 15.4% in the placebo group (adjusted absolute difference and 95% CI − 0.1% [− 8·3 to 8·0]). In sensitivity analysis censoring observations after drug discontinuation or rescue therapy (tocilizumab/increased steroid dose), proportions of death were 5.8% versus 8.8% (− 3.2% [− 9.0 to 2.7]), respectively. There were 148 serious adverse events in 46 participants (33.1%) receiving baricitinib and 155 in 51 participants (37.5%) receiving placebo. In subgroup analyses, there was a potential interaction between vaccination status and treatment allocation on 60-day mortality. In a subsequent post hoc analysis there was a significant interaction between vaccination status and treatment allocation on the occurrence of serious adverse events, with more respiratory complications and severe infections in vaccinated participants treated with baricitinib. Vaccinated participants were on average 11 years older, with more comorbidities.[Conclusion] This clinical trial was prematurely stopped for external evidence and therefore underpowered to conclude on a potential survival benefit of baricitinib in severe/critical COVID-19. We observed a possible safety signal in vaccinated participants, who were older with more comorbidities. Although based on a post-hoc analysis, these findings warrant further investigation in other trials and real-world studies. Trial registration Bari-SolidAct is registered at NCT04891133 (registered May 18, 2021) and EUClinicalTrials.eu (2022-500385-99-00).EU-SolidAct is part of the European pandemic preparedness network EU RESPONSE, funded by the EU Horizon 2020 Research and Innovation programme, under grant number 101015736. EU-SolidAct has also received funding from CAPNET (France) and Klinbeforsk (Norway).Peer reviewe
    • 

    corecore