Pluripotent stem cell-derived kidney organoids: An in vivo-like in vitro technology

2016 ◽  
Vol 790 ◽  
pp. 12-20 ◽  
Author(s):  
Frans Schutgens ◽  
Marianne C. Verhaar ◽  
Maarten B. Rookmaaker
2021 ◽  
Author(s):  
Jitske Jansen ◽  
Bartholomeus T van den Berge ◽  
Martijn van den Broek ◽  
Rutger J Maas ◽  
Brigith Willemsen ◽  
...  

Nephrotic syndrome (NS) is characterized by severe proteinuria as a consequence of kidney glomerular injury due to podocyte damage. In vitro models mimicking in vivo podocyte characteristics are a prerequisite to resolve NS pathogenesis. Here, we report human induced pluripotent stem cell derived kidney organoids containing a podocyte population that heads towards adult podocytes and were superior compared to 2D counterparts, based on scRNA sequencing, super-resolution imaging and electron microscopy. In this study, these next-generation podocytes in kidney organoids enabled personalized idiopathic nephrotic syndrome modeling as shown by activated slit diaphragm signaling and podocyte injury following protamine sulfate treatment and exposure to NS plasma containing pathogenic permeability factors. Organoids cultured from cells of a patient with heterozygous NPHS2 mutations showed poor NPHS2 expression and aberrant NPHS1 localization, which was reversible after genetic correction. Repaired organoids displayed increased VEGFA pathway activity and transcription factor activity known to be essential for podocyte physiology, as shown by RNA sequencing. This study shows that organoids are the preferred model of choice to study idiopathic and congenital podocytopathies.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Gabriel Peinkofer ◽  
Martina Maass ◽  
Kurt Pfannkuche ◽  
Agapios Sachinidis ◽  
Stephan Baldus ◽  
...  

Abstract Background Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) are regarded as promising cell type for cardiac cell replacement therapy, but it is not known whether the developmental stage influences their persistence and functional integration in the host tissue, which are crucial for a long-term therapeutic benefit. To investigate this, we first tested the cell adhesion capability of murine iPSC-CM in vitro at three different time points during the differentiation process and then examined cell persistence and quality of electrical integration in the infarcted myocardium in vivo. Methods To test cell adhesion capabilities in vitro, iPSC-CM were seeded on fibronectin-coated cell culture dishes and decellularized ventricular extracellular matrix (ECM) scaffolds. After fixed periods of time, stably attached cells were quantified. For in vivo experiments, murine iPSC-CM expressing enhanced green fluorescent protein was injected into infarcted hearts of adult mice. After 6–7 days, viable ventricular tissue slices were prepared to enable action potential (AP) recordings in transplanted iPSC-CM and surrounding host cardiomyocytes. Afterwards, slices were lysed, and genomic DNA was prepared, which was then used for quantitative real-time PCR to evaluate grafted iPSC-CM count. Results The in vitro results indicated differences in cell adhesion capabilities between day 14, day 16, and day 18 iPSC-CM with day 14 iPSC-CM showing the largest number of attached cells on ECM scaffolds. After intramyocardial injection, day 14 iPSC-CM showed a significant higher cell count compared to day 16 iPSC-CM. AP measurements revealed no significant difference in the quality of electrical integration and only minor differences in AP properties between d14 and d16 iPSC-CM. Conclusion The results of the present study demonstrate that the developmental stage at the time of transplantation is crucial for the persistence of transplanted iPSC-CM. iPSC-CM at day 14 of differentiation showed the highest persistence after transplantation in vivo, which may be explained by a higher capability to adhere to the extracellular matrix.


2021 ◽  
Vol 12 ◽  
Author(s):  
Kriti Joshi ◽  
Fergus Cameron ◽  
Swasti Tiwari ◽  
Stuart I. Mannering ◽  
Andrew G. Elefanty ◽  
...  

Induced pluripotent stem cell (iPSC) technology is increasingly being used to create in vitro models of monogenic human disorders. This is possible because, by and large, the phenotypic consequences of such genetic variants are often confined to a specific and known cell type, and the genetic variants themselves can be clearly identified and controlled for using a standardized genetic background. In contrast, complex conditions such as autoimmune Type 1 diabetes (T1D) have a polygenic inheritance and are subject to diverse environmental influences. Moreover, the potential cell types thought to contribute to disease progression are many and varied. Furthermore, as HLA matching is critical for cell-cell interactions in disease pathogenesis, any model that seeks to test the involvement of particular cell types must take this restriction into account. As such, creation of an in vitro model of T1D will require a system that is cognizant of genetic background and enables the interaction of cells representing multiple lineages to be examined in the context of the relevant environmental disease triggers. In addition, as many of the lineages critical to the development of T1D cannot be easily generated from iPSCs, such models will likely require combinations of cell types derived from in vitro and in vivo sources. In this review we imagine what an ideal in vitro model of T1D might look like and discuss how the required elements could be feasibly assembled using existing technologies. We also examine recent advances towards this goal and discuss potential uses of this technology in contributing to our understanding of the mechanisms underlying this autoimmune condition.


Biomedicines ◽  
2020 ◽  
Vol 8 (9) ◽  
pp. 346 ◽  
Author(s):  
Roya Rasaei ◽  
Eunbi Kim ◽  
Ji-Young Kim ◽  
Sunghun Na ◽  
Jung-Hyun Kim ◽  
...  

Hyperglycemia is a causative factor in the pathogenesis of respiratory diseases, known to induce fibrosis and inflammation in the lung. However, little attention has been paid to genes related to hyperglycemic-induced lung alterations and stem cell applications for therapeutic use. In this study, our microarray data revealed significantly increased levels of junctional adhesion molecule 2 (JAM2) in the high glucose (HG)-induced transcriptional profile in human perivascular cells (hPVCs). The elevated level of JAM2 in HG-treated hPVCs was transcriptionally and epigenetically reversible when HG treatment was removed. We further investigated the expression of JAM2 using in vivo and in vitro hyperglycemic models. Our results showed significant upregulation of JAM2 in the lungs of streptozotocin (STZ)-induced diabetic mice, which was greatly suppressed by the administration of conditioned medium obtained from human mesenchymal stem cell cultures. Furthermore, JAM2 was found to be significantly upregulated in human pluripotent stem cell-derived multicellular alveolar organoids by exposure to HG. Our results suggest that JAM2 may play an important role in STZ-induced lung alterations and could be a potential indicator for predicting the therapeutic effects of stem cells and drugs in diabetic lung complications.


Author(s):  
Xiaohua Duan ◽  
Yuling Han ◽  
Liuliu Yang ◽  
Benjamin E. Nilsson-Payant ◽  
Pengfei Wang ◽  
...  

Summary ParagraphThe current COVID-19 pandemic is caused by SARS-coronavirus 2 (SARS-CoV-2). There are currently no therapeutic options for mitigating this disease due to lack of a vaccine and limited knowledge of SARS-CoV-2 biology. As a result, there is an urgent need to create new disease models to study SARS-CoV-2 biology and to screen for therapeutics using human disease-relevant tissues. COVID-19 patients typically present with respiratory symptoms including cough, dyspnea, and respiratory distress, but nearly 25% of patients have gastrointestinal indications including anorexia, diarrhea, vomiting, and abdominal pain. Moreover, these symptoms are associated with worse COVID-19 outcomes1. Here, we report using human pluripotent stem cell-derived colonic organoids (hPSC-COs) to explore the permissiveness of colonic cell types to SARS-CoV-2 infection. Single cell RNA-seq and immunostaining showed that the putative viral entry receptor ACE2 is expressed in multiple hESC-derived colonic cell types, but highly enriched in enterocytes. Multiple cell types in the COs can be infected by a SARS-CoV-2 pseudo-entry virus, which was further validated in vivo using a humanized mouse model. We used hPSC-derived COs in a high throughput platform to screen 1280 FDA-approved drugs against viral infection. Mycophenolic acid and quinacrine dihydrochloride were found to block the infection of SARS-CoV-2 pseudo-entry virus in COs both in vitro and in vivo, and confirmed to block infection of SARS-CoV-2 virus. This study established both in vitro and in vivo organoid models to investigate infection of SARS-CoV-2 disease-relevant human colonic cell types and identified drugs that blocks SARS-CoV-2 infection, suitable for rapid clinical testing.


2020 ◽  
Vol 9 (13) ◽  
Author(s):  
Evgeniya Vaskova ◽  
Gentaro Ikeda ◽  
Yuko Tada ◽  
Christine Wahlquist ◽  
Marc Mercola ◽  
...  

Background Exosomes are small extracellular vesicles that function as intercellular messengers and effectors. Exosomal cargo contains regulatory small molecules, including mi RNA s, mRNA s, lnc RNA s, and small peptides that can be modulated by different pathological stimuli to the cells. One of the main mechanisms of action of drug therapy may be the altered production and/or content of the exosomes. Methods and Results We studied the effects on exosome production and content by neprilysin inhibitor/angiotensin receptor blockers, sacubitril/valsartan and valsartan alone, using human‐induced pluripotent stem cell‐derived cardiomyocytes under normoxic and hypoxic injury model in vitro , and assessed for physiologic correlation using an ischemic myocardial injury rodent model in vivo. We demonstrated that the treatment with sacubitril/valsartan and valsartan alone resulted in the increased production of exosomes by induced pluripotent stem cell‐derived cardiomyocytes in vitro in both conditions as well as in the rat plasma in vivo. Next‐generation sequencing of these exosomes exhibited downregulation of the expression of rno‐miR‐181a in the sacubitril/valsartan treatment group. In vivo studies employing chronic rodent myocardial injury model demonstrated that miR‐181a antagomir has a beneficial effect on cardiac function. Subsequently, immunohistochemical and molecular studies suggested that the downregulation of miR‐181a resulted in the attenuation of myocardial fibrosis and hypertrophy, restoring the injured rodent heart after myocardial infarction. Conclusions We demonstrate that an additional mechanism of action of the pleiotropic effects of sacubitril/valsartan may be mediated by the modulation of the mi RNA expression level in the exosome payload.


2019 ◽  
Vol 2019 ◽  
pp. 1-17 ◽  
Author(s):  
Teresa P. Silva ◽  
João P. Cotovio ◽  
Evguenia Bekman ◽  
Maria Carmo-Fonseca ◽  
Joaquim M. S. Cabral ◽  
...  

Human morphogenesis is a complex process involving distinct microenvironmental and physical signals that are manipulated in space and time to give rise to complex tissues and organs. Advances in pluripotent stem cell (PSC) technology have promoted the in vitro recreation of processes involved in human morphogenesis. The development of organoids from human PSCs represents one reliable source for modeling a large spectrum of human disorders, as well as a promising approach for drug screening and toxicological tests. Based on the “self-organization” capacity of stem cells, different PSC-derived organoids have been created; however, considerable differences between in vitro-generated PSC-derived organoids and their in vivo counterparts have been reported. Advances in the bioengineering field have allowed the manipulation of different components, including cellular and noncellular factors, to better mimic the in vivo microenvironment. In this review, we focus on different examples of bioengineering approaches used to promote the self-organization of stem cells, including assembly, patterning, and morphogenesis in vitro, contributing to tissue-like structure formation.


2021 ◽  
Vol 129 (Suppl_1) ◽  
Author(s):  
Olivia T Ly ◽  
Grace Brown ◽  
Hanna Chen ◽  
Liang Hong ◽  
Xinge Wang ◽  
...  

Introduction: The limited success of pharmacological approaches to atrial fibrillation ( AF ) is due to limitations of in vitro and in vivo models and inaccessibility of human atrial tissue. Patient-specific induced pluripotent stem cell-derived atrial cardiomyocytes (iPSC-aCMs) are a robust platform to model the heterogeneous myocardial substrate of AF, but their immaturity limits their fidelity. Objective: We hypothesized that a combinatorial approach of biochemical (triiodothyronine [ T3 ], insulin-like growth factor-1 [ IGF-1 ], and dexamethasone; collectively TID ), bioenergetic (fatty acids [ FA ]), and electrical stimulation ( ES ) will enhance electrophysiological ( EP ), structural, and metabolic maturity of iPSC- a CMs. Methods: We assessed maturation with whole cell patch clamping, calcium transients, immunofluorescence (IF), Seahorse Analyzer, contractility assay, RT-PCR, Western Blotting, and RNA sequencing (RNAseq). Using a time series with RNAseq we identified signaling pathways and transcriptional regulation that drive EP, structural, and metabolic atrial development and compared iPSC-aCM maturity with human aCMs (haCMs) obtained from the same patient. Results: TID+FA+ES significantly improved structural organization and cell morphology ( Fig. 1a ), enhanced membrane potential stability and improved depolarization ( Fig. 1b ), improved Ca 2+ kinetics with faster and increased Ca 2+ release from sarcoplasmic reticulum ( Fig. 1c ), and increased expression of Na + , Ca 2+ , and K + channels, markers of structural maturity, FA metabolism, and oxidative phosphorylation ( Fig. 1d ). There was no difference in each parameter between TID+FA+ES iPSC-aCMs and haCMs from the same patient. Conclusion: Our optimized, combinatorial TID+FA+ES approach markedly enhanced EP, structural, and metabolic maturity of human iPSC-aCMs, which will be useful for elucidating the genetic basis of AF developing precision drug therapies.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2047-2047
Author(s):  
Chuanfeng Wu ◽  
So Gun Hong ◽  
Alexander Jares ◽  
Vicky Guo ◽  
Thomas Winkler ◽  
...  

Abstract Abstract 2047 Induced pluripotent stem cell (iPSC) based therapies offer a promising path for patient-specific regenerative medicine. However, residual undifferentiated iPSCs within the graft present a potential risk of tumor development. Including a suicide gene system in the transplanted cells would allow the elimination of the transplant in case of an adverse event. For this purpose we propose the use of an inducible caspase 9 (iCasp9) suicide gene system (Di Stasi et al., 2011), consisting of human caspase 9 fused to a modified human FK-binding protein. After conditional iCasp9 dimerization by a synthetic small molecule chemical inducer (AP1903) the marked cells undergo rapid apoptosis (Clackson et al., 1998). To explore the system in the context of iPSC, we studied in vitro and in vivo purging strategies as well as therapeutically modalities using lentiviral constructs expressing iCasp9 either from a constitutional promoters or a pluripotency-specific promoter in a teratoma mouse model. We designed lentiviral vectors consisting of the iCasp9 gene linked via a 2A peptide to human CD19, driven by either CMV, EF1α, or embryonic stem cell-specific EOS (3+) promoters (Hotta et al., 2009). The iCasp9 lentiviral constructs were introduced into either into mouse iPSCs (C57Bl/6) or rhesus monkey fibroblasts that were subsequently used to derive monkey iCasp9-iPSCs for future nonhuman primate model. In mature rhesus monkey fibroblasts, cells expressing iCasp9/CD19 driven by either CMV or EF1α promoters underwent rapid and complete apoptosis with exposure to the AP1903 dimerizer (0.5μM to 10 μM), whereas the EOS (3+) promoter drove minimal iCasp9-CD19 expression in fibroblasts, as expected. Murine iPSCs transduced with the CMV-iCasp9 construct demonstrated silencing of the transgenes and no significant apoptosis induction with exposure to AP1903 (10 μM). In contrast, both the EF1α and the EOS (3+) promoters highly expressed iCasp9, allowing for successful induction of apoptosis in iPSCs following AP1903 treatment, with dosages ranging from 0.5μM to 10 μM in vitro, cell apoptosis were analyzed by Annexin V/7AAD using flow cytometry. We next tested the iCasp9 suicide gene system in an in vivo NSG mouse teratoma assay. For this we chose three different approaches: 1) In vitro treatment with AP1903(10 μM) of the iPSCs 4 hours prior to cells injection to NSG mouse (in vitro purging iCasp9-CD19 expressing cells); 2) Immediate I.V. injection of AP1903 (1mg/kg, single does) after injection of the iPSCs into NSG mouse (in vivo purging) ; and 3) Treatment with AP1903(1mg/kg) one week after iPSC injection (in vivo purging after initial teratoma formation). A non-iCasp9 mouse iPSC clone was used as a control in the assay. The results showed that mice injected with iPSCs expressing EOS (3+) or EF1α iCasp9 that underwent in vitro purging with 10 μM AP1903 did not show teratoma formation until 2 months, whereas the non-iCasp9 mouse iPSCs control mice reached the end point size of the teratoma after two weeks(1.494±0.29 g, mean±SD). Mice treated with a single dose of 1mg/kg AP1903 post iPSC cell injection demonstrated markedly delayed teratoma formation. Mice treated one week post iPSC injection with a daily AP1903 regimen (1mg/kg) for four days demonstrated inhibition of further tumor growth, but did not fully ablate existing tumors. In conclusion, we have shown that the iCasp9/AP1903 system is effective in eliminating iPSCs in vitro and in vivo. Furthermore, we show that efficient transgene expression and subsequent elimination of pluripotent cells depends on the internal promoter of the viral construct. Currently, we are investigating why the induction of the suicide gene failed to ablate existing tumors efficiently (insufficient dosage, silencing of transgenes in vivo). Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document