Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 13(9): e0204580, 2018.
Article in English | MEDLINE | ID: mdl-30261016

ABSTRACT

Following a certain type-specific number of mitotic divisions, terminally differentiated cells undergo proliferative senescence, thwarting efforts to expand different cell populations in vitro for the needs of scientific research or medical therapies. The primary cause of this phenomenon is the progressive shortening of the telomeres and the subsequent activation of cell cycle control pathways leading to a block of cell proliferation. Restoration of telomere length by transgenic expression of telomerase reverse transcriptase (TERT) usually results in bypassing of the replicative senescence and ultimately in cell immortalization. To date, there have not been any reports regarding immortalization of cells from common marmoset (Callithrix jacchus), an important non-human primate model for various human diseases, with the use of exogenous human TERT (hTERT). In this study, marmoset fibroblasts were successfully immortalized with transposon-integrated transgenic hTERT and expanded in vitro for over 500 population doublings. Calculation of population doubling levels (PDL) showed that the derived hTERT-transgenic lines had significantly higher proliferation potential than the wild-type fibroblasts, which reached only a maximum of 46 doublings. However, the immortalized cells exhibited differences in the morphology compared with the control fibroblasts and transcriptome analysis also revealed changes in the gene expression patterns. Finally, the karyotypes of all hTERT-transgenic cell lines showed various aberrations such as presence of extra Chromosome 17, isochromosome 21q, or tetraploidy. By single-cell expansion of the least affected monoclonal immortalized line, one sub-clonal line with normal karyotype was established, suggesting the possibility to derive immortal marmoset cells with normal karyotypes. The results of this study are an important step towards the development and optimization of methods for the production of immortalized cells from common marmoset monkeys.


Subject(s)
Callithrix , Fibroblasts/cytology , Fibroblasts/enzymology , Telomerase/genetics , Animals , Cell Line, Transformed , Cell Proliferation/genetics , Cells, Cultured , Cellular Senescence/genetics , DNA Transposable Elements/genetics , Gene Expression , Gene Expression Profiling , Humans , Karyotyping , Recombinant Proteins/genetics , Telomere Homeostasis/genetics
2.
Primate Biol ; 4(2): 231-240, 2017.
Article in English | MEDLINE | ID: mdl-32110709

ABSTRACT

The protein c-CBL is a ubiquitin ligase. It catalyzes the last step of the transfer of ubiquitin to target proteins. Upon completion of polyubiquitination, the target proteins are degraded. Clinically, it is important that c-CBL is mutated in a subset of patients who develop myeloid malignancies, which are diseases of the hematopoietic stem or progenitor cells. c-CBL has also been shown to be expressed by human spermatogonia. The whole spermatogonial cell population possesses a subset that comprises also the spermatogonial stem cells. Based on these findings we hypothesized that c-CBL might be a general stem cell marker. To test this, we first validated the antibody using marmoset bone marrow and adult testis. In both tissues, the expected staining pattern was observed. Western blot analysis revealed only one band of the expected size. Then, we examined the expression of c-CBL in marmoset monkey embryonic stem (ES) cells, induced pluripotent stem (iPS) cells and adult stem cells. We found that c-CBL is strongly expressed in undifferentiated marmoset iPS cells and ES cells. However, adult stem cells in the gut and the stomach did not express c-CBL, indicating that c-CBL is not a general stem cell marker. In summary, c-CBL is strongly expressed in pluripotent stem cells of the marmoset monkey as well as in selected adult stem cell types. Future studies will define the function of c-CBL in pluripotent stem cells.

3.
Sci Rep ; 6: 29122, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27385131

ABSTRACT

Embryonic stem cells (ESCs) are useful for the study of embryonic development. However, since research on naturally conceived human embryos is limited, non-human primate (NHP) embryos and NHP ESCs represent an excellent alternative to the corresponding human entities. Though, ESC lines derived from naturally conceived NHP embryos are still very rare. Here, we report the generation and characterization of four novel ESC lines derived from natural preimplantation embryos of the common marmoset monkey (Callithrix jacchus). For the first time we document derivation of NHP ESCs derived from morula stages. We show that quantitative chromosome-wise transcriptome analyses precisely reflect trisomies present in both morula-derived ESC lines. We also demonstrate that the female ESC lines exhibit different states of X-inactivation which is impressively reflected by the abundance of the lncRNA X inactive-specific transcript (XIST). The novel marmoset ESC lines will promote basic primate embryo and ESC studies as well as preclinical testing of ESC-based regenerative approaches in NHP.


Subject(s)
Callithrix/genetics , Embryonic Stem Cells/metabolism , Genome , Transcriptome/genetics , Animals , Biomarkers/metabolism , Blastocyst/cytology , Cell Differentiation/genetics , Cell Line , Cell Shape , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Karyotyping , Male , Morula/cytology , Sex Determination Processes/genetics , Teratoma/pathology , X Chromosome Inactivation/genetics
4.
PLoS One ; 11(6): e0157570, 2016.
Article in English | MEDLINE | ID: mdl-27322380

ABSTRACT

Curative approaches for eye cataracts and other eye abnormalities, such as myopia and hyperopia currently suffer from a lack of appropriate models. Here, we present a new approach for in vitro growth of lentoid bodies from induced pluripotent stem (iPS) cells as a tool for ophthalmological research. We generated a transgenic mouse line with lens-specific expression of a fluorescent reporter driven by the alphaA crystallin promoter. Fetal fibroblasts were isolated from transgenic fetuses, reprogrammed to iPS cells, and differentiated to lentoid bodies exploiting the specific fluorescence of the lens cell-specific reporter. The employment of cell type-specific reporters for establishing and optimizing differentiation in vitro seems to be an efficient and generally applicable approach for developing differentiation protocols for desired cell populations.


Subject(s)
Cell Differentiation , Genes, Reporter , Induced Pluripotent Stem Cells/cytology , Lens, Crystalline/cytology , Animals , Cellular Reprogramming , Crystallins/metabolism , DNA Transposable Elements/genetics , Female , Fetus/cytology , Fetus/embryology , Fetus/metabolism , Fibroblasts/metabolism , Fluorescence , Mice , Organ Specificity
5.
Cell Reprogram ; 17(2): 131-40, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25826726

ABSTRACT

Induced pluripotent stem cells (iPSCs) are a seminal breakthrough in stem cell research and are promising tools for advanced regenerative therapies in humans and reproductive biotechnology in farm animals. iPSCs are particularly valuable in species in which authentic embryonic stem cell (ESC) lines are yet not available. Here, we describe a nonviral method for the derivation of bovine iPSCs employing Sleeping Beauty (SB) and piggyBac (PB) transposon systems encoding different combinations of reprogramming factors, each separated by self-cleaving peptide sequences and driven by the chimeric CAGGS promoter. One bovine iPSC line (biPS-1) generated by a PB vector containing six reprogramming genes was analyzed in detail, including morphology, alkaline phosphatase expression, and typical hallmarks of pluripotency, such as expression of pluripotency markers and formation of mature teratomas in immunodeficient mice. Moreover, the biPS-1 line allowed a second round of SB transposon-mediated gene transfer. These results are promising for derivation of germ line-competent bovine iPSCs and will facilitate genetic modification of the bovine genome.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming , DNA Transposable Elements , Fibroblasts/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Cattle , Cell Line , Genetic Vectors , Mice , Teratoma
6.
PLoS One ; 10(3): e0118424, 2015.
Article in English | MEDLINE | ID: mdl-25785453

ABSTRACT

Groundbreaking studies showed that differentiated somatic cells of mouse and human origin could be reverted to a stable pluripotent state by the ectopic expression of only four proteins. The resulting pluripotent cells, called induced pluripotent stem (iPS) cells, could be an alternative to embryonic stem cells, which are under continuous ethical debate. Hence, iPS cell-derived functional cells such as neurons may become the key for an effective treatment of currently incurable degenerative diseases. However, besides the requirement of efficacy testing of the therapy also its long-term safety needs to be carefully evaluated in settings mirroring the clinical situation in an optimal way. In this context, we chose the long-lived common marmoset monkey (Callithrix jacchus) as a non-human primate species to generate iPS cells. The marmoset monkey is frequently used in biomedical research and is gaining more and more preclinical relevance due to the increasing number of disease models. Here, we describe, to our knowledge, the first-time generation of marmoset monkey iPS cells from postnatal skin fibroblasts by non-viral means. We used the transposon-based, fully reversible piggyback system. We cloned the marmoset monkey reprogramming factors and established robust and reproducible reprogramming protocols with a six-factor-in-one-construct approach. We generated six individual iPS cell lines and characterized them in comparison with marmoset monkey embryonic stem cells. The generated iPS cells are morphologically indistinguishable from marmoset ES cells. The iPS cells are fully reprogrammed as demonstrated by differentiation assays, pluripotency marker expression and transcriptome analysis. They are stable for numerous passages (more than 80) and exhibit euploidy. In summary, we have established efficient non-viral reprogramming protocols for the derivation of stable marmoset monkey iPS cells, which can be used to develop and test cell replacement therapies in preclinical settings.


Subject(s)
Cellular Reprogramming Techniques/methods , Fibroblasts/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Callithrix , DNA Transposable Elements , Female , Genetic Vectors , Male , Mice , Skin/cytology , Transgenes
7.
Biochem Biophys Res Commun ; 450(1): 581-7, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24928388

ABSTRACT

The generation of induced pluripotent stem (iPS) cells represents a promising approach for innovative cell therapies. The original method requires viral transduction of several reprogramming factors, which may be associated with an increased risk of tumorigenicity. Transposition of reprogramming cassettes represents a recent alternative to viral approaches. Since binary transposons can be produced as common plasmids they provide a safe and cost-efficient alternative to viral delivery methods. Here, we compared the efficiency of two different transposon systems, Sleeping Beauty (SB) and piggyBac (PB), for the generation of murine iPS. Murine fibroblasts derived from an inbred BL/6 mouse line carrying a pluripotency reporter, Oct4-EGFP, and fibroblasts derived from outbred NMRI mice were employed for reprogramming. Both transposon systems resulted in the successful isolation of murine iPS cell lines. The reduction of the core reprogramming factors to omit the proto-oncogene c-Myc was compatible with iPS cell line derivation, albeit with reduced reprogramming efficiencies. The transposon-derived iPS cells featured typical hallmarks of pluripotency, including teratoma growth in immunodeficient mice. Thus SB and PB transposons represent a promising non-viral approach for iPS cell derivation.


Subject(s)
DNA Transposable Elements/genetics , Fibroblasts/cytology , Fibroblasts/physiology , Genetic Engineering/methods , Nerve Tissue Proteins/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Transposases/genetics , Animals , Cell Culture Techniques/methods , Cell Differentiation , Cell Line , Cells , Genetic Vectors/genetics , Mice , Proto-Oncogene Mas , Transfection , Viruses/genetics
8.
PLoS Genet ; 9(12): e1003960, 2013.
Article in English | MEDLINE | ID: mdl-24339785

ABSTRACT

The second messengers cAMP and cGMP activate their target proteins by binding to a conserved cyclic nucleotide-binding domain (CNBD). Here, we identify and characterize an entirely novel CNBD-containing protein called CRIS (cyclic nucleotide receptor involved in sperm function) that is unrelated to any of the other members of this protein family. CRIS is exclusively expressed in sperm precursor cells. Cris-deficient male mice are either infertile due to a lack of sperm resulting from spermatogenic arrest, or subfertile due to impaired sperm motility. The motility defect is caused by altered Ca(2+) regulation of flagellar beat asymmetry, leading to a beating pattern that is reminiscent of sperm hyperactivation. Our results suggest that CRIS interacts during spermiogenesis with Ca(2+)-regulated proteins that--in mature sperm--are involved in flagellar bending.


Subject(s)
Carrier Proteins/genetics , Cyclic AMP/genetics , Flagella/genetics , Protein Binding/genetics , Spermatogenesis/genetics , Animals , Calcium/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Flagella/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Phosphorylation , Signal Transduction/genetics , Sperm Motility/genetics , Spermatozoa/metabolism
9.
Nat Neurosci ; 16(3): 273-80, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23334579

ABSTRACT

Microglia are crucial for immune responses in the brain. Although their origin from the yolk sac has been recognized for some time, their precise precursors and the transcription program that is used are not known. We found that mouse microglia were derived from primitive c-kit(+) erythromyeloid precursors that were detected in the yolk sac as early as 8 d post conception. These precursors developed into CD45(+) c-kit(lo) CX(3)CR1(-) immature (A1) cells and matured into CD45(+) c-kit(-) CX(3)CR1(+) (A2) cells, as evidenced by the downregulation of CD31 and concomitant upregulation of F4/80 and macrophage colony stimulating factor receptor (MCSF-R). Proliferating A2 cells became microglia and invaded the developing brain using specific matrix metalloproteinases. Notably, microgliogenesis was not only dependent on the transcription factor Pu.1 (also known as Sfpi), but also required Irf8, which was vital for the development of the A2 population, whereas Myb, Id2, Batf3 and Klf4 were not required. Our data provide cellular and molecular insights into the origin and development of microglia.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Interferon Regulatory Factors/metabolism , Microglia/cytology , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Kruppel-Like Factor 4 , Mice , Microglia/metabolism , Proto-Oncogene Proteins c-kit/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...