skip to main content


Title: Deciphering retinal diseases through the generation of three dimensional stem cell-derived organoids: Concise Review
Abstract

Three-dimensional (3D) retinal organoids, in vitro tissue structures derived from self-organizing cultures of differentiating human embryonic stem cells or induced pluripotent stem cells, could recapitulate some aspects of the cytoarchitectural structure and function of the retina in vivo. 3D retinal organoids display huge potential for the investigation of the pathogenesis of monogenic hereditary eye diseases that are related to the malfunction or degeneration of photoreceptors or retinal ganglion cells by providing an effective in vitro tool with multiple applications. In combination with recent genome editing tools, 3D retinal organoids could also represent a reliable and renewable source of transplantable cells for personalized therapies. In this review, we describe the recent advances in human pluripotent stem cells-derived retinal organoids, determination of their histoarchitecture, complexity, and maturity. We also discuss their application as a means to decipher the pathogenesis of retinal diseases, as well as the main drawbacks and challenges. Stem Cells  2019;37:1496–1504

</sec> </span> <a href='#' class='show open-abstract' style='margin-left:10px;'>more »</a> <a href='#' class='hide close-abstract' style='margin-left:10px;'>« less</a> <div style="clear:both;margin-bottom:20px;"></div> <dl class="dl-horizontal small"> <dt>NSF-PAR ID:</dt> <dd>10363099</dd> </dl> <dl class="dl-horizontal small"> <dt>Author(s) / Creator(s):</dt> <dd> <a target="_blank" rel="noopener noreferrer" href="https://par.nsf.gov/search/author:"Artero Castro, Ana""><span class="author" itemprop="author">Artero Castro, Ana</span> <sup class="text-muted"></sup></a><span class="sep">; </span><a target="_blank" rel="noopener noreferrer" href="https://par.nsf.gov/search/author:"Rodríguez Jimenez, Francisco Javier""><span class="author" itemprop="author">Rodríguez Jimenez, Francisco Javier</span> <sup class="text-muted"></sup></a><span class="sep">; </span><a target="_blank" rel="noopener noreferrer" href="https://par.nsf.gov/search/author:"Jendelova, Pavla""><span class="author" itemprop="author">Jendelova, Pavla</span> <sup class="text-muted"></sup></a><span class="sep">; </span><a target="_blank" rel="noopener noreferrer" href="https://par.nsf.gov/search/author:"Erceg, Slaven""><span class="author" itemprop="author">Erceg, Slaven</span> <sup class="text-muted"></sup></a></dd> </dl> <dl class="dl-horizontal small"> <dt>Publisher / Repository:</dt> <dd itemprop="publisher">Oxford University Press</dd> </dl> <dl class="dl-horizontal small"> <dt>Date Published:</dt> <dd> <time itemprop="datePublished" datetime="2019-10-31">2019-10-31</time> </dd> </dl> <dl class="dl-horizontal small"> <dt>Journal Name:</dt> <dd>Stem Cells</dd> </dl> <dl class="dl-horizontal small"> <dt>Volume:</dt> <dd>37</dd> </dl> <dl class="dl-horizontal small"> <dt>Issue:</dt> <dd>12</dd> </dl> <dl class="dl-horizontal small"> <dt>ISSN:</dt> <dd>1066-5099</dd> </dl> <dl class="dl-horizontal small"> <dt>Page Range / eLocation ID:</dt> <dd>p. 1496-1504</dd> </dl> <dl class="dl-horizontal small"> <dt>Format(s):</dt> <dd>Medium: X</dd> </dl> <dl class="dl-horizontal small"> <dt>Sponsoring Org:</dt> <dd itemprop="sourceOrganization">National Science Foundation</dd> </dl> <div class="clearfix"></div> </div> </div> <div id="citation-addl" class="hidden-print"> <h5 id='mlt-header'>More Like this</h5> <ol class="item-list documents" id="citation-mlt" style="min-height: 80px;"> <li> <div class="article item document" itemscope itemtype="http://schema.org/TechArticle"> <div class="item-info"> <div class="title"> <a href="https://par.nsf.gov/biblio/10383395-bioengineering-human-pluripotent-stem-cell-derived-retinal-organoids-optic-vesicle-containing-brain-organoids-ocular-diseases" itemprop="url"> <span class='span-link' itemprop="name">Bioengineering Human Pluripotent Stem Cell-Derived Retinal Organoids and Optic Vesicle-Containing Brain Organoids for Ocular Diseases</span> </a> </div> <div> <strong> <a class="misc external-link" href="https://doi.org/10.3390/cells11213429" target="_blank" title="Link to document DOI">https://doi.org/10.3390/cells11213429  <span class="fas fa-external-link-alt"></span></a> </strong> </div> <div class="metadata"> <span class="authors"> <span class="author" itemprop="author">Arthur, Peggy</span> <span class="sep">; </span><span class="author" itemprop="author">Muok, Laureana</span> <span class="sep">; </span><span class="author" itemprop="author">Nathani, Aakash</span> <span class="sep">; </span><span class="author" itemprop="author">Zeng, Eric Z.</span> <span class="sep">; </span><span class="author" itemprop="author">Sun, Li</span> <span class="sep">; </span><span class="author" itemprop="author">Li, Yan</span> <span class="sep">; </span><span class="author" itemprop="author">Singh, Mandip</span> </span> <span class="year">( <time itemprop="datePublished" datetime="2022-11-01">November 2022</time> , Cells) </span> </div> <div style="cursor: pointer;-webkit-line-clamp: 5;" class="abstract" itemprop="description"> Retinal organoids are three-dimensional (3D) structures derived from human pluripotent stem cells (hPSCs) that mimic the retina’s spatial and temporal differentiation, making them useful as in vitro retinal development models. Retinal organoids can be assembled with brain organoids, the 3D self-assembled aggregates derived from hPSCs containing different cell types and cytoarchitectures that resemble the human embryonic brain. Recent studies have shown the development of optic cups in brain organoids. The cellular components of a developing optic vesicle-containing organoids include primitive corneal epithelial and lens-like cells, retinal pigment epithelia, retinal progenitor cells, axon-like projections, and electrically active neuronal networks. The importance of retinal organoids in ocular diseases such as age-related macular degeneration, Stargardt disease, retinitis pigmentosa, and diabetic retinopathy are described in this review. This review highlights current developments in retinal organoid techniques, and their applications in ocular conditions such as disease modeling, gene therapy, drug screening and development. In addition, recent advancements in utilizing extracellular vesicles secreted by retinal organoids for ocular disease treatments are summarized. </div> <a href='#' class='show open-abstract' style='margin-left:10px;'>more »</a> <a href='#' class='hide close-abstract' style='margin-left:10px;'>« less</a> </div><div class="clearfix"></div> </div> </li> <li> <div class="article item document" itemscope itemtype="http://schema.org/TechArticle"> <div class="item-info"> <div class="title"> <a href="https://par.nsf.gov/biblio/10381941-wnt-signaling-directs-human-pluripotent-stem-cells-vascularized-cardiac-organoids-chamber-like-structures" itemprop="url"> <span class='span-link' itemprop="name">Wnt signaling directs human pluripotent stem cells into vascularized cardiac organoids with chamber-like structures</span> </a> </div> <div> <strong> <a class="misc external-link" href="https://doi.org/10.3389/fbioe.2022.1059243" target="_blank" title="Link to document DOI">https://doi.org/10.3389/fbioe.2022.1059243  <span class="fas fa-external-link-alt"></span></a> </strong> </div> <div class="metadata"> <span class="authors"> <span class="author" itemprop="author">Liang, Po-Yu</span> <span class="sep">; </span><span class="author" itemprop="author">Chang, Yun</span> <span class="sep">; </span><span class="author" itemprop="author">Jin, Gyuhyung</span> <span class="sep">; </span><span class="author" itemprop="author">Lian, Xiaojun</span> <span class="sep">; </span><span class="author" itemprop="author">Bao, Xiaoping</span> </span> <span class="year">( <time itemprop="datePublished" datetime="2022-11-18">November 2022</time> , Frontiers in Bioengineering and Biotechnology) </span> </div> <div style="cursor: pointer;-webkit-line-clamp: 5;" class="abstract" itemprop="description"> Heart diseases are leading cause of death around the world. Given their unique capacity to self-renew and differentiate into all types of somatic cells, human pluripotent stem cells (hPSCs) hold great promise for heart disease modeling and cardiotoxic drug screening. hPSC-derived cardiac organoids are emerging biomimetic models for studying heart development and cardiovascular diseases, but it remains challenging to make mature organoids with a native-like structure in vitro . In this study, temporal modulation of Wnt signaling pathway co-differentiated hPSCs into beating cardiomyocytes and cardiac endothelial-like cells in 3D organoids, resulting in cardiac endothelial-bounded chamber formation. These chambered cardiac organoids exhibited more mature membrane potential compared to cardiac organoids composed of only cardiomyocytes. Furthermore, a better response to toxic drugs was observed in chamber-contained cardiac organoids. In summary, spatiotemporal signaling pathway modulation may lead to more mature cardiac organoids for studying cardiovascular development and diseases. </div> <a href='#' class='show open-abstract' style='margin-left:10px;'>more »</a> <a href='#' class='hide close-abstract' style='margin-left:10px;'>« less</a> </div><div class="clearfix"></div> </div> </li> <li> <div class="article item document" itemscope itemtype="http://schema.org/TechArticle"> <div class="item-info"> <div class="title"> <a href="https://par.nsf.gov/biblio/10368904-microstructured-hydrogels-guide-selfassembly-function-lung-alveolospheres" itemprop="url"> <span class='span-link' itemprop="name">Microstructured Hydrogels to Guide Self‐Assembly and Function of Lung Alveolospheres</span> </a> </div> <div> <strong> <a class="misc external-link" href="https://doi.org/10.1002/adma.202202992" target="_blank" title="Link to document DOI">https://doi.org/10.1002/adma.202202992  <span class="fas fa-external-link-alt"></span></a> </strong> </div> <div class="metadata"> <span class="authors"> <span class="author" itemprop="author">Loebel, Claudia</span> <span class="sep">; </span><span class="author" itemprop="author">Weiner, Aaron I.</span> <span class="sep">; </span><span class="author" itemprop="author">Eiken, Madeline K.</span> <span class="sep">; </span><span class="author" itemprop="author">Katzen, Jeremy B.</span> <span class="sep">; </span><span class="author" itemprop="author">Morley, Michael P.</span> <span class="sep">; </span><span class="author" itemprop="author">Bala, Vikram</span> <span class="sep">; </span><span class="author" itemprop="author">Cardenas‐Diaz, Fabian L.</span> <span class="sep">; </span><span class="author" itemprop="author">Davidson, Matthew D.</span> <span class="sep">; </span><span class="author" itemprop="author">Shiraishi, Kazushige</span> <span class="sep">; </span><span class="author" itemprop="author">Basil, Maria C.</span> <span class="sep">; </span><span class="author">et al</span></span> <span class="year">( <time itemprop="datePublished" datetime="2022-06-07">June 2022</time> , Advanced Materials) </span> </div> <div style="cursor: pointer;-webkit-line-clamp: 5;" class="abstract" itemprop="description"> <title>Abstract

Epithelial cell organoids have increased opportunities to probe questions on tissue development and disease in vitro and for therapeutic cell transplantation. Despite their potential, current protocols to grow these organoids almost exclusively depend on culture within 3D Matrigel, which limits defined culture conditions, introduces animal components, and results in heterogenous organoids (i.e., shape, size, composition). Here, a method is described that relies on hyaluronic acid hydrogels for the generation and expansion of lung alveolar organoids (alveolospheres). Using synthetic hydrogels with defined chemical and physical properties, human‐induced pluripotent stem cell (iPSC)‐derived alveolar type 2 cells (iAT2s) self‐assemble into alveolospheres and propagate in Matrigel‐free conditions. By engineering predefined microcavities within these hydrogels, the heterogeneity of alveolosphere size and structure is reduced when compared to 3D culture, while maintaining the alveolar type 2 cell fate of human iAT2‐derived progenitor cells. This hydrogel system is a facile and accessible system for the culture of iPSC‐derived lung progenitors and the method can be expanded to the culture of primary mouse tissue derived AT2 and other epithelial progenitor and stem cell aggregates.

 
more » « less
  • The mechanism that causes the Alzheimer’s disease (AD) pathologies, including amyloid plaque, neurofibrillary tangles, and neuron death, is not well understood due to the lack of robust study models for human brain. Three-dimensional organoid systems based on human pluripotent stem cells (hPSCs) have shown a promising potential to model neurodegenerative diseases, including AD. These systems, in combination with engineering tools, allow in vitro generation of brain-like tissues that recapitulate complex cell-cell and cell-extracellular matrix (ECM) interactions. Brain ECMs play important roles in neural differentiation, proliferation, neuronal network, and AD progression. In this contribution related to brain ECMs, recent advances in modeling AD pathology and progression based on hPSC-derived neural cells, tissues, and brain organoids were reviewed and summarized. In addition, the roles of ECMs in neural differentiation of hPSCs and the influences of heparan sulfate proteoglycans, chondroitin sulfate proteoglycans, and hyaluronic acid on the progression of neurodegeneration were discussed. The advantages that use stem cell-based organoids to study neural degeneration and to investigate the effects of ECM development on the disease progression were highlighted. The contents of this article are significant for understanding cell-matrix interactions in stem cell microenvironment for treating neural degeneration. 
    more » « less
  • Background: Recently, the in vitro blood–brain barrier (BBB) models derived from human pluripotent stem cells have been given extensive attention in therapeutics due to the implications they have with the health of the central nervous system. It is essential to create an accurate BBB model in vitro in order to better understand the properties of the BBB, and how it can respond to inflammatory stimulation and be passed by targeted or non-targeted cell therapeutics, more specifically extracellular vesicles. Methods: Brain-specific pericytes (iPCs) were differentiated from iPSK3 cells using dual SMAD signaling inhibitors and Wnt activation plus fibroblast growth factor 2 (FGF-2). The derived cells were characterized by immunostaining, flow cytometry, and RT-PCR. In parallel, blood vessels organoids were derived using Wnt activation, BMP4, FGF2, VEGF, and SB431542. The organoids were replated and treated with retinoic acid to enhance the blood–brain barrier (BBB) features in the differentiated brain endothelial cells (iECs). Co-culture was performed for iPCs and iECs in the transwell system and 3D microfluidics channels. Results: The derived iPCs expressed common markers PDGFRb and NG2, and brain-specific genes FOXF2 , ABCC9 , KCNJ8 , and ZIC1 . The derived iECs expressed common endothelial cell markers CD31, VE-cadherin, and BBB-associated genes BRCP , GLUT-1 , PGP , ABCC1 , OCLN , and SLC2A1 . The co-culture of the two cell types responded to the stimulation of amyloid β42 oligomers by the upregulation of the expression of TNFa , IL6 , NFKB , Casp3 , SOD2 , and TP53 . The co-culture also showed the property of trans-endothelial electrical resistance. The proof of concept vascularization strategy was demonstrated in a 3D microfluidics-based device. Conclusion: The derived iPCs and iECs have brain-specific properties, and the co-culture of iPCs and iECs provides an in vitro BBB model that show inflammatory response. This study has significance in establishing micro-physiological systems for neurological disease modeling and drug screening. 
    more » « less