79 research outputs found

    The future of direct cardiac reprogramming: any GMT cocktail variety?

    Get PDF
    Direct cardiac reprogramming has emerged as a novel therapeutic approach to treat and regenerate injured hearts through the direct conversion of fibroblasts into cardiac cells. Most studies have focused on the reprogramming of fibroblasts into induced cardiomyocytes (iCMs). The first study in which this technology was described, showed that at least a combination of three transcription factors, GATA4, MEF2C and TBX5 (GMT cocktail), was required for the reprogramming into iCMs in vitro using mouse cells. However, this was later demonstrated to be insufficient for the reprogramming of human cells and additional factors were required. Thereafter, most studies have focused on implementing reprogramming efficiency and obtaining fully reprogrammed and functional iCMs, by the incorporation of other transcription factors, microRNAs or small molecules to the original GMT cocktail. In this respect, great advances have been made in recent years. However, there is still no consensus on which of these GMT-based varieties is best, and robust and highly reproducible protocols are still urgently required, especially in the case of human cells. On the other hand, apart from CMs, other cells such as endothelial and smooth muscle cells to form new blood vessels will be fundamental for the correct reconstruction of damaged cardiac tissue. With this aim, several studies have centered on the direct reprogramming of fibroblasts into induced cardiac progenitor cells (iCPCs) able to give rise to all myocardial cell lineages. Especially interesting are reports in which multipotent and highly expandable mouse iCPCs have been obtained, suggesting that clinically relevant amounts of these cells could be created. However, as of yet, this has not been achieved with human iCPCs, and exactly what stage of maturity is appropriate for a cell therapy product remains an open question. Nonetheless, the major concern in regenerative medicine is the poor retention, survival, and engraftment of transplanted cells in the cardiac tissue. To circumvent this issue, several cell pre-conditioning approaches are currently being explored. As an alternative to cell injection, in vivo reprogramming may face fewer barriers for its translation to the clinic. This approach has achieved better results in terms of efficiency and iCMs maturity in mouse models, indicating that the heart environment can favor this process. In this context, in recent years some studies have focused on the development of safer delivery systems such as Sendai virus, Adenovirus, chemical cocktails or nanoparticles. This article provides an in-depth review of the in vitro and in vivo cardiac reprograming technology used in mouse and human cells to obtain iCMs and iCPCs, and discusses what challenges still lie ahead and what hurdles are to be overcome before results from this field can be transferred to the clinical settings

    Generation of two transgene-free human iPSC lines from CD133+ cord blood cells

    Get PDF
    We have generated two human induced pluripotent stem cell (iPSC) lines from CD133+ cells isolated from umbilical cord blood (CB) of a female child using non-integrative Sendai virus. Here we describe the complete characterization of these iPSC lines: PRYDi-CB5 and PRYDi-CB40

    One-step in vitro generation of ETV2-null pig embryos

    Get PDF
    Each year, tens of thousands of people worldwide die of end-stage organ failure due to the limited availability of organs for use in transplantation. To meet this clinical demand, one of the last frontiers of regenerative medicine is the generation of humanized organs in pigs from pluripotent stem cells (PSCs) via blastocyst complementation. For this, organ-disabled pig models are needed. As endothelial cells (ECs) play a critical role in xenotransplantation rejection in every organ, we aimed to produce hematoendothelial-disabled pig embryos targeting the master transcription factor ETV2 via CRISPR-Cas9-mediated genome modification. In this study, we designed five different guide RNAs (gRNAs) against the DNA-binding domain of the porcine ETV2 gene, which were tested on porcine fibroblasts in vitro. Four out of five guides showed cleavage capacity and, subsequently, these four guides were microinjected individually as ribonucleoprotein complexes (RNPs) into one-cell-stage porcine embryos. Next, we combined the two gRNAs that showed the highest targeting efficiency and microinjected them at higher concentrations. Under these conditions, we significantly improved the rate of biallelic mutation. Hence, here, we describe an efficient one-step method for the generation of hematoendothelial-disabled pig embryos via CRISPR-Cas9 microinjection in zygotes. This model could be used in experimentation related to the in vivo generation of humanized organs

    Cure of ADPKD by Selection for Spontaneous Genetic Repair Events in Pkd1-Mutated iPS Cells

    Get PDF
    Induced pluripotent stem cells (iPSCs) generated by epigenetic reprogramming of personal somatic cells have limited therapeutic capacity for patients suffering from genetic disorders. Here we demonstrate restoration of a genomic mutation heterozygous for Pkd1 (polycystic kidney disease 1) deletion (Pkd1(+/−) to Pkd1(+/R+)) by spontaneous mitotic recombination. Notably, recombination between homologous chromosomes occurred at a frequency of 1∼2 per 10,000 iPSCs. Southern blot hybridization and genomic PCR analyses demonstrated that the genotype of the mutation-restored iPSCs was indistinguishable from that of the wild-type cells. Importantly, the frequency of cyst generation in kidneys of adult chimeric mice containing Pkd1(+/R+) iPSCs was significantly lower than that of adult chimeric mice with parental Pkd1(+/−) iPSCs, and indistinguishable from that of wild-type mice. This repair step could be directly incorporated into iPSC development programmes prior to cell transplantation, offering an invaluable step forward for patients carrying a wide range of genetic disorders

    Development Refractoriness of MLL-Rearranged Human B Cell Acute Leukemias to Reprogramming into Pluripotency

    Get PDF
    Induced pluripotent stem cells (iPSCs) are a powerful tool for disease modeling. They are routinely generated from healthy donors and patients from multiple cell types at different developmental stages. However, reprogramming leukemias is an extremely inefficient process. Few studies generated iPSCs from primary chronic myeloid leukemias, but iPSC generation from acute myeloid or lymphoid leukemias (ALL) has not been achieved. We attempted to generate iPSCs from different subtypes of B-ALL to address the developmental impact of leukemic fusion genes. OKSM(L)-expressing mono/polycistronic-, retroviral/lentiviral/episomal-, and Sendai virus vector-based reprogramming strategies failed to render iPSCs in vitro and in vivo. Addition of transcriptomic-epigenetic reprogramming ‘‘boosters’’ also failed to generate iPSCs from B cell blasts and B-ALL lines, and when iPSCs emerged they lacked leukemic fusion genes, demonstrating non-leukemic myeloid origin. Conversely, MLL-AF4-overexpressing hematopoietic stem cells/B progenitors were successfully reprogrammed, indicating that B cell origin and leukemic fusion gene were not reprogramming barriers. Global transcriptome/DNA methylome profiling suggested a developmental/differentiation refractoriness of MLL-rearranged B-ALL to reprogramming into pluripotency

    Generation of Healthy Mice from Gene-Corrected Disease-Specific Induced Pluripotent Stem Cells

    Get PDF
    Using the murine model of tyrosinemia type 1 (fumarylacetoacetate hydrolase [FAH] deficiency; FAH−/− mice) as a paradigm for orphan disorders, such as hereditary metabolic liver diseases, we evaluated fibroblast-derived FAH−/−-induced pluripotent stem cells (iPS cells) as targets for gene correction in combination with the tetraploid embryo complementation method. First, after characterizing the FAH−/− iPS cell lines, we aggregated FAH−/−-iPS cells with tetraploid embryos and obtained entirely FAH−/−-iPS cell–derived mice that were viable and exhibited the phenotype of the founding FAH−/− mice. Then, we transduced FAH cDNA into the FAH−/−-iPS cells using a third-generation lentiviral vector to generate gene-corrected iPS cells. We could not detect any chromosomal alterations in these cells by high-resolution array CGH analysis, and after their aggregation with tetraploid embryos, we obtained fully iPS cell–derived healthy mice with an astonishing high efficiency for full-term development of up to 63.3%. The gene correction was validated functionally by the long-term survival and expansion of FAH-positive cells of these mice after withdrawal of the rescuing drug NTBC (2-(2-nitro-4-fluoromethylbenzoyl)-1,3-cyclohexanedione). Furthermore, our results demonstrate that both a liver-specific promoter (transthyretin, TTR)-driven FAH transgene and a strong viral promoter (from spleen focus-forming virus, SFFV)-driven FAH transgene rescued the FAH-deficiency phenotypes in the mice derived from the respective gene-corrected iPS cells. In conclusion, our data demonstrate that a lentiviral gene repair strategy does not abrogate the full pluripotent potential of fibroblast-derived iPS cells, and genetic manipulation of iPS cells in combination with tetraploid embryo aggregation provides a practical and rapid approach to evaluate the efficacy of gene correction of human diseases in mouse models

    Nuclear Reprogramming Strategy Modulates Differentiation Potential of Induced Pluripotent Stem Cells

    Get PDF
    Bioengineered by ectopic expression of stemness factors, induced pluripotent stem (iPS) cells demonstrate embryonic stem cell-like properties and offer a unique platform for derivation of autologous pluripotent cells from somatic tissue sources. In the process of nuclear reprogramming, somatic tissues are converted to a pluripotent ground state, thus unlocking an unlimited potential to expand progenitor pools. Molecular dissection of nuclear reprogramming suggests that a residual memory derived from the original parental source, along with the remnants of the reprogramming process itself, leads to a biased potential of the bioengineered progeny to differentiate into target tissues such as cardiac cytotypes. In this way, iPS cells that fulfill pluripotency criteria may display heterogeneous profiles for lineage specification. Small molecule-based strategies have been identified that modulate the epigenetic state of reprogrammed cells and are optimized to erase the residual memory and homogenize the differentiation potential of iPS cells derived from distinct backgrounds. Here, we describe the salient components of the reprogramming process and their effect on the downstream differentiation capacity of the iPS populations in the context of cardiovascular regenerative applications

    Present state and future perspectives of using pluripotent stem cells in toxicology research

    Get PDF
    The use of novel drugs and chemicals requires reliable data on their potential toxic effects on humans. Current test systems are mainly based on animals or in vitro–cultured animal-derived cells and do not or not sufficiently mirror the situation in humans. Therefore, in vitro models based on human pluripotent stem cells (hPSCs) have become an attractive alternative. The article summarizes the characteristics of pluripotent stem cells, including embryonic carcinoma and embryonic germ cells, and discusses the potential of pluripotent stem cells for safety pharmacology and toxicology. Special attention is directed to the potential application of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) for the assessment of developmental toxicology as well as cardio- and hepatotoxicology. With respect to embryotoxicology, recent achievements of the embryonic stem cell test (EST) are described and current limitations as well as prospects of embryotoxicity studies using pluripotent stem cells are discussed. Furthermore, recent efforts to establish hPSC-based cell models for testing cardio- and hepatotoxicity are presented. In this context, methods for differentiation and selection of cardiac and hepatic cells from hPSCs are summarized, requirements and implications with respect to the use of these cells in safety pharmacology and toxicology are presented, and future challenges and perspectives of using hPSCs are discussed

    Identification of novel regulators of transcription in iPSC-derived cardiovascular progenitors

    Get PDF
    Stem cells allow to investigate about the basic mechanisms that regulate embryonic development, cellular plasticity, and organ maintenance and regeneration. Induced pluripotent stem cells (iPSCs) are a powerful source of cells for diverse applications such as developmental and disease modeling, drug discovery and regenerative medicine. Cardiac development is coordinated by complex interactions between cardiac progenitor cell populations, different molecular signaling pathways, and spatially and temporally regulated gene expression. Cardiovascular progenitors (CVPs), with similar potential to these present in early stages of embryonic development, can be obtained from iPSCs by mimicking signaling during cardiogenesis, creating an ideal cell source to treat the damaged heart. However, the mechanisms and conditions for long-term self-renewal and maintenance of CVPs remain elusive. The generation of new iPSC models for tracing CVP lineages permits to delve into the biology of CVPs and discover novel potential regulators of their fate. We have established three different Cre/LoxP mouse models for lineage tracing of CVPs and their cell progeny by the expression of ZsGreen (ZsG) protein: Ai6-Mesp1-Cre (Mesp1 tracer), Ai6-Isl1-Cre (Isl1 tracer) and Ai6-Mef2c-AHF-Cre (AHF tracer) mice. Multiple iPSC clones have been derived from Ai6-Isl1-Cre and Ai6-Mef2c-AHF-Cre reporter mice. Several generated iPSC lines have been fully characterized, demonstrating embryonic stem-like features. iPSCs encoded the expected genomic insertions, showed normal karyotypes, transgenes were silenced, and expressed endogenous pluripotency-associated markers. Moreover, iPSCs were capable to differentiate into the three germ layers both in vitro and in vivo. We have verified the utility of established AHFiPSCs to track CVPs and their differentiated progeny. Upon differentiation, ZsG+ cells derived from AHFiPSCs appeared from embryoid body (EB) day 6 onwards, expressed cardiovascular-related markers, and were able to differentiate into cardiomyocytes, endothelial and smooth muscle cells. Comparative gene expression analysis using four different AHFiPSC lines revealed distinct molecular signatures in three particular stages of differentiation: undifferentiated iPSCs (AHFiPS-D0), sorted ZsG+ cells at day 6 (AHFiPS-D6.ZsG+) and sorted ZsG+ cells at day 13 of differentiation (AHFiPS-D13.ZsG+), that expressed pluripotency-, CVP- and cardiac/vascular lineages-associated markers, respectively. Gapdh and Polr2a have been determined the most stable housekeeping genes along the differentiation of AHFiPSCs, being optimal for accurate normalization of gene expression in our samples. We have identified novel regulators of transcription specifically upregulated in CVPs: Lin28a (and its paralog Lin28b), Lhx1 and Nr6a1. The expression of these genes was also found increased in the corresponding CVP-enriched samples from two different public analyses using mouse and human pluripotent stem cells. Moreover, using bioinformatic analysis of biological pathways we have found p53 as an interconnecting molecule of all these selected regulators of transcription. In order to explore novel insights into the biological role of the selected regulators in CVP fate, we have generated an inducible vector (pTRE-CDS-IRES-Puro-REX1-Blast) to carry out gain-of-function (GOF) analyses. This Tet-On system worked properly in AHFiPSC clones, but unfortunately it failed to work in EBs of certain size along differentiation. In contrast, this GOF system correctly functioned in human iPSCs differentiated in monolayer cultures. We have established four CBiPS1sv-4F-5 cell lines carrying Tet-On systems for the inducible expression of LIN28A, LIN28B, NR6A1 and LHX1, and preliminary results indicated that these regulators of transcription might have a role in CVP fate determination

    Generation of Functional Human NKX2.5GFP Reporter Cell Lines for Direct Reprogramming into Proliferative Cardiac Progenitors

    No full text
    The mammalian heart is the first organ formed in the embryo and is composed of four chambers: right atrium (RA), right ventricle (RV), left atrium (LA), and left ventricle (LV), and three layers: the endocardium, the myocardium, and the pericardium. The endocardium is formed by simple squamous epithelium known as endothelium, which lines the chambers and is joined to myocardium by a thin layer of connective tissue. The pericardium is comprised of two distinct sublayers: the inner serous pericardium, which is formed by visceral pericardium or epicardium and parietal pericardium separated by pericardial cavity, and the outer fibrous pericardium made of dense connective tissue. The myocardium is the muscular layer of the heart, it is the thickest one and is located between the endocardium and the pericardium. With regard to cellular composition, the heart is principally formed by cardiomyocytes (CM), cardiac fibroblasts (cFib) , vascular smooth muscle cells (localized fundamentally within the myocardium), vascular endothelial cells (located within the myocardium and endocardium) and mesothelial cells present in the pericardium 1–3 . CM are specialized cells with a complex filament structure responsible for the control of the rhythmic beating of the heart and present heterogeneity depending on the location, morphology, and function, including atrial, ventricular, sinoatrial nodal, atrioventricular nodal, His bundle, and Purkinje fibers 4,5
    corecore