scholarly journals The Potential of iPS Cells in Synucleinopathy Research

2012 ◽  
Vol 2012 ◽  
pp. 1-6 ◽  
Author(s):  
Leonhard Linta ◽  
Marianne Stockmann ◽  
Tobias M. Boeckers ◽  
Alexander Kleger ◽  
Stefan Liebau

α-synuclein is a protein involved in the pathogenesis of several so-called synucleinopathies including Parkinson's disease. A variety of models have been so far assessed. Human induced pluripotent stem cells provide a patient- and disease-specific model forin vitrostudies, pharmacotoxicological screens, and hope for future cell-based therapies. Initial experimental procedures include the harvest of patients’ material for the reprogramming process, the investigation of the patients genetic background in the cultured cells, and the evaluation of disease-relevant factors/proteins under various cell culture conditions.

2012 ◽  
Vol 2012 ◽  
pp. 1-10 ◽  
Author(s):  
Thekkeparambil Chandrabose Srijaya ◽  
Padmaja Jayaprasad Pradeep ◽  
Rosnah Binti Zain ◽  
Sabri Musa ◽  
Noor Hayaty Abu Kasim ◽  
...  

Induced pluripotent stem cell-based therapy for treating genetic disorders has become an interesting field of research in recent years. However, there is a paucity of information regarding the applicability of induced pluripotent stem cells in dental research. Recent advances in the use of induced pluripotent stem cells have the potential for developing disease-specific iPSC linesin vitrofrom patients. Indeed, this has provided a perfect cell source for disease modeling and a better understanding of genetic aberrations, pathogenicity, and drug screening. In this paper, we will summarize the recent progress of the disease-specific iPSC development for various human diseases and try to evaluate the possibility of application of iPS technology in dentistry, including its capacity for reprogramming some genetic orodental diseases. In addition to the easy availability and suitability of dental stem cells, the approach of generating patient-specific pluripotent stem cells will undoubtedly benefit patients suffering from orodental disorders.


2012 ◽  
Vol 14 (4) ◽  
pp. 574-579 ◽  
Author(s):  
Yong Zhu ◽  
Hong-Liang Hu ◽  
Peng Li ◽  
Shi Yang ◽  
Wei Zhang ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3233-3233
Author(s):  
Sarah S Rozelle ◽  
Brenden W Smith ◽  
Efthymia Melista ◽  
Ehimen Aneni ◽  
Paola Sebastiani ◽  
...  

Abstract Abstract 3233 As they can be generated from the somatic cells of any individual, induced pluripotent stem cells (iPSC) represent renewable, potentially unlimited cell sources that circumvent the possibility of inappropriate immune response and open the door to the advent of patient-specific, personalized medicine. Disease-specific iPSCs have the potential to elucidate disease mechanisms, revolutionize drug discovery, and improve patient care. We have built a large library of sickle cell disease-specific iPSCs containing more than 100 individual lines from both African American and Saudi Arab patients with different HbS gene haplotypes and HbF-modulating quantitative trait loci (QTL) genotypes. The differentiation of these lines into the erythroid lineage offers a novel opportunity to study erythroid development, the regulation of globin switching, small molecule drug development and the modeling of red blood cell linked diseases in vitro. Although several teams have published proof-of-principle examples for the derivation of hematopoietic cells from pluripotent stem cells, these protocols are technically demanding and result in the production of limited numbers of cells. Our conceptual approach has been to mimic the natural sequences of development in vitro in order to derive the range and number of cell types needed for the creation of a robust iPSC-based platform. We have developed a novel, chemically defined and feeder-free methodology for the production of large numbers of functionally mature red blood cells (RBCs) from both normal and disease-specific human iPSCs. This protocol utilizes a 2D/adherent approach and eliminates the need for embryoid body formation or xenogeneic agents resulting in a shorter production time (∼10 days). Large numbers of clinically relevant, high purity hematopoietic cells can be generated such that 15,000 cells yield 1 billion cells in two weeks. This protocol produces bipotential megakaryocyte-erythroid progenitors (MEPs) that co-express the surface markers CD235 (red cells) and CD41 (megakaryocytes) and demonstrate expression of accepted panels of both erythroid and megakaryocyte-specific genes. Use of an erythroid maturation media results in efficient maturation of MEPs to erythrocytes. Due to this novel approach and the robust nature of the methodology, we are able to generate large numbers of functionally mature RBCs that produce hemoglobin, respond to oxygen deprivation, and enucleate. Furthermore, these human iPSC-derived directly differentiated erythroid-lineage cells engraft robustly in Nod-SCID-Gamma (NSG) immunocompromised mice and demonstrate detectable chimerism in peripheral blood. Importantly, these cells respond to hydroxyurea (HU), the only FDA approved drug that increases HbF levels in sickle cell anemia. Our goals are to use these cells to further understand hemoglobin switching in carriers of varied HbS haplotypes and to harness our library of sickle cell disease-specific lines in combination with the developed differentiation protocol in order to create correlations between genetics and response to new and available HbF inducing agents, furthering the clinician's capability to personalize treatment plans for each patient. Disclosures: No relevant conflicts of interest to declare.


2011 ◽  
Vol 2011 ◽  
pp. 1-11 ◽  
Author(s):  
Reto Eggenschwiler ◽  
Komal Loya ◽  
Malte Sgodda ◽  
Francoise André ◽  
Tobias Cantz

Direct reprogramming of somatic cells into pluripotent cells by retrovirus-mediated expression of OCT4, SOX2, KLF4, and C-MYC is a promising approach to derive disease-specific induced pluripotent stem cells (iPSCs). In this study, we focused on three murine models for metabolic liver disorders: the copper storage disorder Wilson's disease (toxic-milk mice), tyrosinemia type 1 (fumarylacetoacetate-hydrolase deficiency, FAH−/−mice), and alpha1-antitrypsin deficiency (PiZ mice). Colonies of iPSCs emerged 2-3 weeks after transduction of fibroblasts, prepared from each mouse strain, and were maintained as individual iPSC lines. RT-PCR and immunofluorescence analyses demonstrated the expression of endogenous pluripotency markers. Hepatic precursor cells could be derived from these disease-specific iPSCs applying anin vitrodifferentiation protocol and could be visualized after transduction of a lentiviral albumin-GFP reporter construct. Functional characterization of these cells allowed the recapitulation of the disease phenotype for further studies of underlying molecular mechanisms of the respective disease.


2009 ◽  
Vol 390 (9) ◽  
Author(s):  
Alexandra Rolletschek ◽  
Anna M. Wobus

Abstract Adult cells have been reprogrammed into induced pluripotent stem (iPS) cells by introducing pluripotency-associated transcription factors. Here, we discuss recent advances and challenges of in vitro reprogramming and future prospects of iPS cells for their use in diagnosis and cell therapy. The generation of patient-specific iPS cells for clinical application requires alternative strategies, because genome-integrating viral vectors may cause insertional mutagenesis. Moreover, when suitable iPS cell lines will be available, efficient and selective differentiation protocols are needed to generate transplantable grafts. Finally, we point to the requirement of a regulatory framework necessary for the commercial use of iPS cells.


2011 ◽  
Vol 366 (1575) ◽  
pp. 2286-2296 ◽  
Author(s):  
Gabsang Lee ◽  
Lorenz Studer

Induced pluripotent stem (iPS) cells have considerable promise as a novel tool for modelling human disease and for drug discovery. While the generation of disease-specific iPS cells has become routine, realizing the potential of iPS cells in disease modelling poses challenges at multiple fronts. Such challenges include selecting a suitable disease target, directing the fate of iPS cells into symptom-relevant cell populations, identifying disease-related phenotypes and showing reversibility of such phenotypes using genetic or pharmacological approaches. Finally, the system needs to be scalable for use in modern drug discovery. Here, we will discuss these points in the context of modelling familial dysautonomia (FD, Riley–Day syndrome, hereditary sensory and autonomic neuropathy III (HSAN-III)), a rare genetic disorder in the peripheral nervous system. We have demonstrated three disease-specific phenotypes in FD-iPS-derived cells that can be partially rescued by treating cells with the plant hormone kinetin. Here, we will discuss how to use FD-iPS cells further in high throughput drug discovery assays, in modelling disease severity and in performing mechanistic studies aimed at understanding disease pathogenesis. FD is a rare disease but represents an important testing ground for exploring the potential of iPS cell technology in modelling and treating human disease.


2016 ◽  
Vol 113 (7) ◽  
pp. 1889-1894 ◽  
Author(s):  
Salvatore Iovino ◽  
Alison M. Burkart ◽  
Laura Warren ◽  
Mary Elizabeth Patti ◽  
C. Ronald Kahn

Induced pluripotent stem cells (iPS cells) represent a unique tool for the study of the pathophysiology of human disease, because these cells can be differentiated into multiple cell types in vitro and used to generate patient- and tissue-specific disease models. Given the critical role for skeletal muscle insulin resistance in whole-body glucose metabolism and type 2 diabetes, we have created a novel cellular model of human muscle insulin resistance by differentiating iPS cells from individuals with mutations in the insulin receptor (IR-Mut) into functional myotubes and characterizing their response to insulin in comparison with controls. Morphologically, IR-Mut cells differentiated normally, but had delayed expression of some muscle differentiation-related genes. Most importantly, whereas control iPS-derived myotubes exhibited in vitro responses similar to primary differentiated human myoblasts, IR-Mut myotubes demonstrated severe impairment in insulin signaling and insulin-stimulated 2-deoxyglucose uptake and glycogen synthesis. Transcriptional regulation was also perturbed in IR-Mut myotubes with reduced insulin-stimulated expression of metabolic and early growth response genes. Thus, iPS-derived myotubes from individuals with genetically determined insulin resistance demonstrate many of the defects observed in vivo in insulin-resistant skeletal muscle and provide a new model to analyze the molecular impact of muscle insulin resistance.


2015 ◽  
Vol 27 (1) ◽  
pp. 252
Author(s):  
J. Secher ◽  
K. Freude ◽  
S. Petkov ◽  
A. Ceylan ◽  
M. Schmidt ◽  
...  

Porcine-induced pluripotent stem cells (piPSC) have been established since 2009, but only 1 report demonstrated contribution to germline chimeras. One well-established in vivo pluripotency assay is the teratoma assay, which has recently been questioned due to the lack of standardized guidelines. In the present study we have characterised GFP-tagged in vitro and in vivo tetracycline-inducible piPSC [porcine MYC, SOX2, KLF4 (pOMSK)] and their capacity to form teratomas. We injected 1.5 million cells in 250 µL of PBS subcutaneous into NOD/SCID mice and followed them up to 6 weeks. The teratomas were analysed by immunohistochemistry for the 3 germlayer markers β3 tubulin, α fetoprotein, and smooth muscle actin. We not only found our teratomas positive for these markers, but also co-positive for GFP, clearly showing that the teratoma was made from porcine cells, which was not sufficiently proven in former studies. Our H&E staining revealed the following structures: cuboidal ephitelium, thyroid-like structure, renal corpuscle, and steroid producing cells. We continued to test the capacity of our venus iPS cells to contribute to in vitro chimeras. To achieve this we used a micromanipulator to inject 15 cells into Day 5 parthenotes, and subsequently cultured them in PZM3 with 10% FCS, cultured with or without doxycycline. These in vitro chimeras were followed until Day 7 in Nikons Biostation IM and used for differential staining. In all groups we saw good survival, hatching, and maintenance of GFP, indicating integration of these cells in our in vitro assay. We only found differences between survivals of the cell lines in the group cultured with doxycycline. Finally, in order to assess if the naïve type venus iPS cells could possibly be a truly naïve piPSC, we tested their capacity to form in vivo germline chimeras. This was tested by injecting 15 piPSC into Day 4 to 5 in vivo embryos. The injected embryos were transferred into 5 surrogate mothers, 3 of them were fed doxycycline before the transfer and 5 days after, and the last 2 recipient sows were not fed doxycycline. The pregnancies were terminated at Day 32 and the embryos were examined for fluorescence and the GFP transgene by PCR. In summary, it appears that both naïve type and primed type venus iPS cells are still strongly dependent on the pOMSK transgene expression, and the ultimate proof for pluripotency, the chimera production, seems to be not achievable under the condition we have chosen.


2017 ◽  
Vol 58 (4) ◽  
pp. 430-438 ◽  
Author(s):  
Shoki Inui ◽  
Kazumasa Minami ◽  
Emiko Ito ◽  
Hiromasa Imaizumi ◽  
Seiji Mori ◽  
...  

Abstract Induced pluripotent stem (iPS) cells have demonstrated they can undergo self-renewal, attain pluripotency, and differentiate into various types of functional cells. In clinical transplantation of iPS cells, however, a major problem is the prevention of tumorigenesis. We speculated that tumor formation could be inhibited by means of irradiation. Since the main purpose of this study was to explore the prevention of tumor formation in human iPS (hiPS) cells, we tested the effects of irradiation on tumor-associated factors such as radiosensitivity, pluripotency and cell death in hiPS cells. The irradiated hiPS cells showed much higher radiosensitivity, because the survival fraction of hiPS cells irradiated with 2 Gy was < 10%, and there was no change of pluripotency. Irradiation with 2 and 4 Gy caused substantial cell death, which was mostly the result of apoptosis. Irradiation with 2 Gy was detrimental enough to cause loss of proliferation capability and trigger substantial cell death in vitro. The hiPS cells irradiated with 2 Gy were injected into NOG mice (NOD/Shi-scid, IL-2 Rγnull) for the analysis of tumor formation. The group of mice into which hiPS cells irradiated with 2 Gy was transplanted showed significant suppression of tumor formation in comparison with that of the group into which non-irradiated hiPS cells were transplanted. It can be presumed that this diminished rate of tumor formation was due to loss of proliferation and cell death caused by irradiation. Our findings suggest that tumor formation following cell therapy or organ transplantation induced by hiPS cells may be prevented by irradiation.


Sign in / Sign up

Export Citation Format

Share Document