Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Environ Sci Technol ; 58(28): 12313-12319, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38958666

ABSTRACT

Southern Hemisphere humpback whales accumulate persistent and toxic chemicals, which are transported to Antarctica through distant sources and in situ usage. The extreme seasonal migration-associated fast of humpback whales results in the remobilization of persistent and lipophilic environmental contaminants from liberated fat stores. Mitochondria play a key role in lipid metabolism, and any disruption to mitochondrial function is expected to influence whole-organism bioenergetics. It is therefore of interest to advance understanding of the impact of known contaminants of the Antarctic sea-ice ecosystem upon humpback whale cellular bioenergetics. Using cell line-based in vitro testing, this study employed the Seahorse Extracellular Flux Analyzer to study cellular metabolic activity in live humpback whale fibroblast cells. The assay, based on oxygen consumption rate, provides insights into the cause of cellular bioenergetic disruption. Immortalized skin fibroblasts were exposed to four priority environmental chemicals found in the Antarctic sea-ice ecosystem. Our findings reveal chemical-dependent functional alterations and varying bioenergetic profile responses. Chlorpyrifos was observed to decrease mitochondrial basal oxygen consumption; dieldrin increased basal oxygen consumption; trifluralin's impact was dose-specific, and endosulfan displayed no effect. Our results provide unique insights into environmental chemical mechanisms of action on cellular bioenergetics, generating much-needed taxa-specific chemical effect data in support of evidence-based conservation policy and management.


Subject(s)
Energy Metabolism , Fibroblasts , Humpback Whale , Animals , Fibroblasts/drug effects , Fibroblasts/metabolism , Energy Metabolism/drug effects , Humpback Whale/metabolism , Oxygen Consumption/drug effects , Water Pollutants, Chemical/toxicity
2.
Environ Sci Technol ; 57(24): 8975-8982, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37272882

ABSTRACT

Cetaceans are at elevated risk of accumulating persistent and lipophilic environmental contaminants due to their longevity and high proportion of body fat. Despite this, there is a paucity of taxa-specific chemical effect data, in part due to the ethical and logistical constraints in working with highly mobile aquatic species. Advances in cetacean cell culture have opened the door to the application of mainstream in vitro toxicological effect assessment approaches. Image-based cell profiling is a high-throughput, microscopy-based system commonly applied in drug development. It permits the analysis of the xenobiotic effect on multiple cell organelles simultaneously, hereby flagging its potential utility in the evaluation of chemical toxicodynamics. Here we exposed immortalized humpback whale skin fibroblasts (HuWaTERT) to six priority environmental contaminants known to accumulate in the Southern Ocean food web, in order to explore their subcellular organelle responses. Results revealed chemical-dependent modulation of mitochondrial texture, with the lowest observed effect concentrations for chlorpyrifos, dieldrin, trifluralin, and p,p'-dichlorodiphenyldichloroethane of 0.3, 4.1, 9.3, and 19.8 nM, respectively. By contrast, no significant changes were observed upon exposure to endosulfan and lindane. This study contributes the first fixed mitochondrial images of HuWaTERT and constitutes novel, taxa-specific chemical effect data in support of evidence-based conservation policy and management.


Subject(s)
Humpback Whale , Hydrocarbons, Chlorinated , Pesticides , Animals , Humpback Whale/physiology , Hydrocarbons, Chlorinated/analysis , Hydrocarbons, Chlorinated/metabolism , Pesticides/analysis , Mitochondria/chemistry , Fibroblasts/chemistry , Fibroblasts/metabolism
3.
Int J Mol Sci ; 23(20)2022 Oct 20.
Article in English | MEDLINE | ID: mdl-36293487

ABSTRACT

Marsupials have been a powerful comparative model to understand mammalian biology. However, because of the unique characteristics of their embryology, marsupial pluripotency architecture remains to be fully understood, and nobody has succeeded in developing embryonic stem cells (ESCs) from any marsupial species. We have developed an integration-free iPSC reprogramming method and established validated iPSCs from two inbred strains of a marsupial, Monodelphis domestica. The monoiPSCs showed a significant (6181 DE-genes) and highly uniform (r2 [95% CI] = 0.973 ± 0.007) resetting of the cellular transcriptome and were similar to eutherian ESCs and iPSCs in their overall transcriptomic profiles. However, monoiPSCs showed unique regulatory architecture of the core pluripotency transcription factors and were more like marsupial epiblasts. Our results suggest that POU5F1 and the splice-variant-specific expression of POU5F3 synergistically regulate the opossum pluripotency gene network. It is plausible that POU5F1, POU5F3 splice variant XM_016427856.1, and SOX2 form a self-regulatory network. NANOG expression, however, was specific to monoiPSCs and epiblasts. Furthermore, POU5F1 was highly expressed in trophectoderm cells, whereas all other pluripotency transcription factors were significantly downregulated, suggesting that the regulatory architecture of core pluripotency genes of marsupials may be distinct from that of eutherians.


Subject(s)
Induced Pluripotent Stem Cells , Monodelphis , Animals , Monodelphis/genetics , Induced Pluripotent Stem Cells/metabolism , Embryonic Stem Cells , Transcription Factors/genetics , Transcription Factors/metabolism , Mammals , Cellular Reprogramming/genetics
4.
Front Immunol ; 12: 599014, 2021.
Article in English | MEDLINE | ID: mdl-33746946

ABSTRACT

Schistosomes infect about 250 million people globally causing the devastating and persistent disease of schistosomiasis. These blood flukes have a complicated life cycle involving alternating infection of freshwater snail intermediate and definitive mammalian hosts. To survive and flourish in these diverse environments, schistosomes transition through a number of distinct life-cycle stages as a result of which they change their body plan in order to quickly adapt to each new environment. Current research suggests that stem cells, present in adults and larvae, are key in aiding schistosomes to facilitate these changes. Given the recent advances in our understanding of schistosome stem cell biology, we review the key roles that two major classes of cells play in the different life cycle stages during intramolluscan and intramammalian development; these include the germinal cells of sporocysts involved in asexual reproduction in molluscan hosts and the neoblasts of adult worms involved in sexual reproduction in human and other mammalian hosts. These studies shed considerable new light in revealing the stem cell heterogeneity driving the propagation of the schistosome life cycle. We also consider the possibility and value of establishing stem cell lines in schistosomes to advance schistosomiasis research. The availability of such self-renewable resources will provide new platforms to study stem cell behavior and regulation, and to address fundamental aspects of schistosome biology, reproductive development and survival. In turn, such studies will create new avenues to unravel individual gene function and to optimize genome-editing processes in blood flukes, which may lead to the design of novel intervention strategies for schistosomiasis.


Subject(s)
Schistosoma , Schistosomiasis/immunology , Stem Cells , Animals , Biomedical Research , Schistosoma/cytology , Schistosoma/immunology , Stem Cells/cytology , Stem Cells/immunology
5.
Sci Rep ; 11(1): 3486, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33568729

ABSTRACT

With a view towards harnessing the therapeutic potential of canine mesenchymal stromal cells (cMSCs) as modulators of inflammation and the immune response, and to avoid the issues of the variable quality and quantity of harvested cMSCs, we examined the immunomodulatory properties of cMSCs derived from canine induced pluripotent stem cells (ciMSCs), and compared them to cMSCs harvested from adipose tissue (cAT-MSC) and bone marrow (cBM-MSC). A combination of deep sequencing and quantitative RT-PCR of the ciMSC transcriptome confirmed that ciMSCs express more genes in common with cBM-MSCs and cAT-MSCs than with the ciPSCs from which they were derived. Both ciMSCs and harvested cMSCs express a range of pluripotency factors in common with the ciPSCs including NANOG, POU5F1 (OCT-4), SOX-2, KLF-4, LIN-28A, MYC, LIF, LIFR, and TERT. However, ESRRB and PRDM-14, both factors associated with naïve, rather than primed, pluripotency were expressed only in the ciPSCs. CXCR-4, which is essential for the homing of MSCs to sites of inflammation, is also detectable in ciMSCs, cAT- and cBM-MSCs, but not ciPSCs. ciMSCs constitutively express the immunomodulatory factors iNOS, GAL-9, TGF-ß1, PTGER-2α and VEGF, and the pro-inflammatory mediators COX-2, IL-1ß and IL-8. When stimulated with the canine pro-inflammatory cytokines tumor necrosis factor-α (cTNF-α), interferon-γ (cIFN-γ), or a combination of both, ciMSCs upregulated their expression of IDO, iNOS, GAL-9, HGF, TGF-ß1, PTGER-2α, VEGF, COX-2, IL-1ß and IL-8. When co-cultured with mitogen-stimulated lymphocytes, ciMSCs downregulated their expression of iNOS, HGF, TGF-ß1 and PTGER-2α, while increasing their expression of COX-2, IDO and IL-1ß. Taken together, these findings suggest that ciMSCs possess similar immunomodulatory capabilities as harvested cMSCs and support further investigation into their potential use for the management of canine immune-mediated and inflammatory disorders.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Mesenchymal Stem Cells/physiology , Adipose Tissue/cytology , Animals , Anti-Inflammatory Agents/metabolism , Bone Marrow Cells/physiology , Cells, Cultured , Coculture Techniques , Cytokines/immunology , Cytokines/metabolism , Dogs , Gene Expression Regulation , Immunologic Factors/metabolism , Lymphocyte Activation , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Transcriptome
6.
Anim Reprod Sci ; 219: 106546, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32828417

ABSTRACT

Whilst adoption of in vitro production (IVP) of cattle embryos and subsequent biopsy for genetic evaluation is increasing, biopsy techniques primarily used were developed to sample in vivo-produced blastocysts. This study was conducted to develop a laser-assisted blastomere extrusion approach for rapid and minimal-invasive biopsy of IVP cattle embryos at pre-morula to morula stages of development (Day 5 or 6 post-fertilisation). Embryo development into blastocysts was not compromised when ≤3 cells were collected by blastomere extrusion on Day 5 (44.4 ± 4.4 % and 34.3 ± 4.6 %) or Day 6 (58.0 ± 4.3 % and 57.5 ± 5.3 %) post-fertilisation compared with non-biopsied control embryos. Similarly, capacity to withstand cryopreservation was not different between embryos biopsied at Day 5 and 6 post-fertilisation and control-embryos (58.8 ± 6.0 %, 63.5 ± 5.6 %, and 56.0 ± 4.8 %, respectively). When more cells were collected from embryos at Day 6 post-fertilisation (≥8 compared to ≤3 cells), subsequent embryo development was not different (63.6 ± 6.1 % and 73.1 ± 6.2 %, respectively) nor was the capacity to withstand cryopreservation (67.9 ± 9.0 % and 62.5 ± 8.7 %, respectively). For biopsies on Day 6 post-fertilization, 95 % of samples produced a PCR product; however, when compared to the whole embryo PCR results, approximately 11 % of biopsy-samples classified as being from a male embryo were from female embryos (false positive), indicating DNA contamination between samples. In conclusion, results of this study indicate laser-assisted blastomere extrusion is a time efficient and minimally invasive approach to biopsy IVP morula and pre-morula cattle embryos to facilitate genetic analysis.


Subject(s)
Blastomeres/pathology , Cattle/embryology , Cleavage Stage, Ovum/pathology , Lasers , Morula/pathology , Animals , Biopsy/methods , Biopsy/veterinary , Blastocyst/pathology , Cells, Cultured , Cryopreservation/veterinary , Embryo Culture Techniques/veterinary , Embryo, Mammalian/pathology , Embryonic Development/physiology , Female , Fertilization in Vitro/veterinary , Lasers/adverse effects , Male , Polymerase Chain Reaction/veterinary , Preimplantation Diagnosis/methods , Preimplantation Diagnosis/veterinary
7.
Gen Comp Endocrinol ; 291: 113436, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32057910

ABSTRACT

Baleen whales are vulnerable to environmental impacts due to low fecundity, capital breeding strategies, and their reliance on a large amount of prey resources over large spatial scales. There has been growing interest in monitoring health and physiological stress in these species but, to date, few measures have been validated. The purpose of this study was to examine whether blubber cortisol could be used as a measure of physiological stress in humpback whales. Cortisol concentrations were initially compared between live, presumably 'healthy' whales (n = 187) and deceased whales (n = 35), which had died after stranding or entanglement, or washed ashore as a carcass. Deceased whales were found to have significantly higher cortisol levels (mean ± SD; 5.47 ± 4.52 ng/g) than live whales (0.51 ± 0.14 ng/g; p < 0.001), particularly for those animals that had experienced prolonged trauma (e.g. stranding) prior to death. Blubber cortisol levels in live whales were then examined for evidence of life history-related, seasonal, or sampling-related effects. Life history group and sampling-related factors, such as encounter time and the number of biopsy sampling attempts per animal, were found to be poor predictors of blubber cortisol levels in live whales. In contrast, blubber cortisol levels varied seasonally, with whales migrating north towards the breeding grounds in winter having significantly higher levels (0.54 ± 0.21 ng/g, p = 0.016) than those migrating south towards the feeding grounds in spring (0.48 ± 1.23 ng/g). These differences could be due to additional socio-physiological stress experienced by whales during peaks in breeding activity. Overall, blubber cortisol appears to be a suitable measure of chronic physiological stress in humpback whales.


Subject(s)
Animal Structures/metabolism , Humpback Whale/anatomy & histology , Hydrocortisone/metabolism , Stress, Physiological , Adipose Tissue/metabolism , Animals , Female , Geography , Humpback Whale/physiology , Male , Progesterone/metabolism , Queensland , Seasons
8.
Stem Cells Dev ; 29(1): 25-37, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31709909

ABSTRACT

Marsupials have long attracted scientific interest because of their unique biological features and their position in mammalian evolution. Mesenchymal stem cells (MSCs) are of considerable research interest in translational medicine due to their immunomodulatory, anti-inflammatory, and regenerative properties. MSCs have been harvested from various tissues in numerous eutherian species; however, there are no descriptions of MSCs derived from a marsupial. In this study, we have generated Tasmanian devil (Sarcophilus harrisii) MSCs from devil induced pluripotent stem cells (iPSCs), thus providing an unlimited source of devil MSCs and circumventing the need to harvest tissues from live animals. Devil iPSCs were differentiated into MSCs (iMSCs) through both embryoid body formation assays (EB-iMSCs) and through inhibition of the transforming growth factor beta/activin signaling pathway (SB-iMSCs). Both EB-iMSCs and SB-iMSCs are highly proliferative and express the MSC-specific surface proteins CD73, CD90, and CD105, in addition to the pluripotency transcription factors OCT4/POU5F1, SOX2, and NANOG. Expression of the marsupial pluripotency factor POU5F3, a paralogue of OCT4/POU5F1, is significantly reduced in association with the transition from pluripotency to multipotency. Devil iMSCs readily differentiate along the adipogenic, osteogenic, and chondrogenic pathways in vitro, confirming their trilineage differentiation potential. Importantly, in vitro teratoma assays confirmed their multipotency, rather than pluripotency, since the iMSCs only formed derivatives of the mesodermal germ layer. Devil iMSCs show a tropism toward medium conditioned by devil facial tumor cells and express a range of immunomodulatory and anti-inflammatory factors. Therefore, devil iMSCs will be a valuable tool for further studies on marsupial biology and may facilitate the development of an MSC-based treatment strategy against Devil Facial Tumor Disease.


Subject(s)
Facial Neoplasms/genetics , Immunologic Factors/genetics , Induced Pluripotent Stem Cells/metabolism , Marsupialia/genetics , Mesenchymal Stem Cells/metabolism , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Adipogenesis/genetics , Animals , Chondrogenesis/genetics , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Endoglin/genetics , Endoglin/metabolism , Facial Neoplasms/metabolism , Facial Neoplasms/pathology , Gene Expression , Immunologic Factors/metabolism , Induced Pluripotent Stem Cells/cytology , Marsupialia/metabolism , Mesenchymal Stem Cells/cytology , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Osteogenesis/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Thy-1 Antigens/genetics , Thy-1 Antigens/metabolism , Tropism/genetics
9.
Cell Biol Toxicol ; 35(4): 387-398, 2019 08.
Article in English | MEDLINE | ID: mdl-30627956

ABSTRACT

Marine mammals, such as whales, have a high proportion of body fat and so are susceptible to the accumulation, and associated detrimental health effects, of lipophilic environmental contaminants. Recently, we developed a wild-type cell line from humpback whale fibroblasts (HuWa). Extensive molecular assessments with mammalian wild-type cells are typically constrained by a finite life span, with cells eventually becoming senescent. Thus, the present work explored the possibility of preventing senescence in the HuWa cell line by transfection with plasmids encoding the simian virus large T antigen (SV40T) or telomerase reverse transcriptase (TERT). No stable expression was achieved upon SV40 transfection. Transfection with TERT, on the other hand, activated the expression of telomerase in HuWa cells. At the time of manuscript preparation, the transfected HuWa cells (HuWaTERT) have been stable for at least 59 passages post-transfection. HuWaTERT proliferate rapidly and maintain initial cell characteristics, such as morphology and chromosomal stability. The response of HuWaTERT cells to an immune stimulant (lipopolysaccharide (LPS)) and an immunotoxicant (Aroclor1254) was assessed by measurement of intracellular levels of the pro-inflammatory cytokines interleukin (IL)-6, IL-1ß and tumour necrosis factor (TNF)-α. HuWaTERT cells constitutively express IL-6, IL-1ß and TNFα. Exposure to neither LPS nor Aroclor1254 had an effect on the levels of these cytokines. Overall, this work supports the diverse applicability of HuWa cell lines in that they display reliable long-term preservation, susceptibility to exogenous gene transfer and enable the study of humpback whale-specific cellular response mechanisms.


Subject(s)
Fibroblasts/metabolism , Humpback Whale/metabolism , Adipose Tissue , Animals , Aroclors/analysis , Cell Line/physiology , Cell Line, Transformed/metabolism , Gene Transfer Techniques , Lipopolysaccharides , Polychlorinated Biphenyls/analysis , Telomerase/metabolism , Transfection/methods
10.
Stem Cells Dev ; 28(3): 151-164, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30417748

ABSTRACT

The platypus (Ornithorhynchus anatinus) is an egg-laying monotreme mammal whose ancestors diverged ∼166 million years ago from the evolutionary pathway that eventually gave rise to both marsupial and eutherian mammals. Consequently, its genome is an extraordinary amalgam of both ancestral reptilian and derived mammalian features. To gain insight into the evolution of mammalian pluripotency, we have generated induced pluripotent stem cells from the platypus (piPSCs). Deep sequencing of the piPSC transcriptome revealed that piPSCs robustly express the core eutherian pluripotency factors POU5F1/OCT4, SOX2, and NANOG. Given the more extensive role of SOX3 over SOX2 in avian pluripotency, our data indicate that between 315 and 166 million years ago, primitive mammals replaced the role of SOX3 in the vertebrate pluripotency network with SOX2. DAX1/NR0B1 is not expressed in piPSCs and an analysis of the platypus DAX1 promoter revealed the absence of a proximal SOX2-binding DNA motif known to be critical for DAX1 expression in eutherian pluripotent stem cells, suggesting that the acquisition of SOX2 responsiveness by DAX1 has facilitated its recruitment into the pluripotency network of eutherians. Using the RNAseq data, we were also able to demonstrate that in both fibroblasts and piPSCs, the expression ratio of X chromosomes to autosomes (X1-5 X1-5:AA) is approximately equal to 1, indicating that there is no upregulation of X-linked genes. Finally, the RNAseq data also allowed us to explore the process of X-linked gene inactivation in the platypus, where we determined that for any given gene, there is no preference for silencing of the maternal or paternal allele; that is, within a population of cells, the silencing of X-linked genes is not imprinted.


Subject(s)
Cell Differentiation , Platypus , Pluripotent Stem Cells/cytology , Transcriptome , Animals , Cells, Cultured , DAX-1 Orphan Nuclear Receptor/genetics , DAX-1 Orphan Nuclear Receptor/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Genomic Imprinting , Pluripotent Stem Cells/metabolism , SOX Transcription Factors/genetics , SOX Transcription Factors/metabolism , X Chromosome Inactivation
11.
Stem Cells Dev ; 27(10): 704-715, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29562867

ABSTRACT

Horses are susceptible to a number of neurotropic viruses, including West Nile virus (WNV), which is a pathogen of global significance in both horses and humans. However, there are no in vitro models with which to study infectious neuropathic diseases in the horse. In an effort to redress this, we have generated neurons from equine induced pluripotent stem cells (equiPSCs) that express a range of cortical neuron-specific markers, in addition to the membrane-bound ligand ephrin B3, which plays an important role in axon guidance as well as functioning as the receptor through which henipaviruses, such as Hendra virus, enter mammalian neurons. EquiPSC-derived neurons spontaneously depolarize with waves of depolarization conducted unidirectionally to adjacent neurons. We sought to confirm that equiPSC-derived neurons are a possible in vitro model for viral neuropathic diseases in the horse by examining their susceptibility to infection with flaviviruses that are known to be neurotropic in horses, including WNV and Murray Valley encephalitis virus (MVEV), and to compare these to nonpathogenic flaviviruses such as Fitzroy River virus (FRV) and Bamaga virus (BgV). All three strains of WNV tested in this study grew to high titres in the equiPSC-derived neurons, inducing a strong cytopathic effect (cpe), as did MVEV. In contrast, FRV showed restricted replication, and no cpe, which is consistent with the observation that FRV infects, but does not cause disease, in horses. BgV, which is thought to infect only marsupials, did not replicate in the equiPSC-derived neurons. Hence, our equiPSC-derived neurons display virus-specific differences in terms of viral titre and cpe that are similar to observations made in vivo, thus supporting their use as an in vitro model for neurotropic viral infection in horses.


Subject(s)
Flavivirus Infections/virology , Flavivirus/pathogenicity , Horse Diseases/virology , Induced Pluripotent Stem Cells/virology , Neurons/virology , Animals , Horses , Virus Replication/physiology , West Nile Fever/virology , West Nile virus/pathogenicity
12.
APL Bioeng ; 2(4): 046103, 2018 Dec.
Article in English | MEDLINE | ID: mdl-31069325

ABSTRACT

Precise and accurate gene correction is crucial for enabling iPSC-based therapies, and Cas9-Nickase based approaches are increasingly considered for in vivo correction of diseases such as beta-thalassemia. Here, we generate footprint-free induced pluripotent stem cells from a patient with a beta-thalassemia mutation (IVSII-1 G > A) and employ a double Cas9nickase-mediated correction strategy combined with a piggyBac transposon-modified donor vector for gene correction. Our approach further aimed to minimize the formation of adjacent single-strand breaks at the targeted allele through the destruction of the binding site for one guide and the use of a synonymous protospacer adjacent motif blocking mutation (canonical PAM sequence 5'-NGG-3' is changed to 5'-NCG-3', where N indicates any nucleobase) for the other guide. We show that this strategy indeed not only permits bi-allelic seamless repair of the beta-globin gene splice site mutation and negligible off-target mutagenesis or re-editing of the targeted allele but also results in unexpected on-target mutagenesis with some guide RNAs (gRNAs) in several targeted clones. This study thus not only validates a framework for seamless gene correction with enhanced specificity and accuracy but also highlights potential safety concerns associated with Cas9-nickase based gene correction.

13.
Stem Cells Dev ; 27(2): 112-122, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29161957

ABSTRACT

We demonstrate the generation of Tasmanian devil (Sarcophilus harrisii) induced pluripotent stem cells (DeviPSCs) from dermal fibroblasts by lentiviral delivery of human transcription factors. DeviPSCs display characteristic pluripotent stem cell colony morphology, with individual cells having a high nuclear-to-cytoplasmic ratio and alkaline phosphatase activity. DeviPSCs are leukemia inhibitory factor dependent and have reactivated endogenous octamer-binding transcription factor 4 [OCT4, POU domain, class 5, transcription factor 1 (POU5F1)], POU2 [POU domain, class 5, transcription factor 3 (POU5F3)], sex determining region Y-box 2 (SOX2), Nanog homeobox (NANOG) and dosage-sensitive sex reversal, adrenal hypoplasia congenita critical region on the X chromosome, gene 1 (DAX1) genes, retained a normal karyotype, and concurrently silenced exogenous human transgenes. Notably, co-expression of both OCT4 and POU2 suggests that they are representative of cells of the epiblast, the marsupial equivalent of the inner cell mass. DeviPSCs readily form embryoid bodies and in vitro teratomas containing derivatives of all three embryonic germ layers. To date, DeviPSCs have been stably maintained for more than 45 passages. Our DeviPSCs provide an invaluable resource for studies into marsupial pluripotency and development, and they may also serve as an important tool in efforts to combat the threat of devil facial tumor disease.


Subject(s)
Biological Evolution , Cellular Reprogramming Techniques , Induced Pluripotent Stem Cells/metabolism , Marsupialia/metabolism , Transcription Factors/biosynthesis , Transduction, Genetic , Animals , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Marsupialia/genetics , Transcription Factors/genetics
14.
Semin Cell Dev Biol ; 56: 117-121, 2016 08.
Article in English | MEDLINE | ID: mdl-26806635

ABSTRACT

Marsupials and monotremes represent evolutionarily divergent lineages from the majority of extant mammals which are eutherian, or placental, mammals. Monotremes possess multiple X and Y chromosomes that appear to have arisen independently of eutherian and marsupial sex chromosomes. Dosage compensation of X-linked genes occurs in monotremes on a gene-by-gene basis, rather than through chromosome-wide silencing, as is the case in eutherians and marsupials. Specifically, studies in the platypus have shown that for any given X-linked gene, a specific proportion of nuclei within a cell population will silence one locus, with the percentage of cells undergoing inactivation at that locus being highly gene-specific. Hence, it is perhaps not surprising that the expression level of X-linked genes in female platypus is almost double that in males. This is in contrast to the situation in marsupials where one of the two X chromosomes is inactivated in females by the long non-coding RNA RSX, a functional analogue of the eutherian XIST. However, marsupial X chromosome inactivation differs from that seen in eutherians in that it is exclusively the paternal X chromosome that is silenced. In addition, marsupials appear to have globally upregulated X-linked gene expression in both sexes, thus balancing their expression levels with those of the autosomes, a process initially proposed by Ohno in 1967 as being a fundamental component of the X chromosome dosage compensation mechanism but which may not have evolved in eutherians.


Subject(s)
Biological Evolution , Dosage Compensation, Genetic , Marsupialia/genetics , X Chromosome/genetics , Animals , Humans
15.
Aquat Toxicol ; 167: 240-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26363275

ABSTRACT

This paper reports the first successful derivation and characterization of humpback whale fibroblast cell lines. Primary fibroblasts were isolated from the dermal connective tissue of skin biopsies, cultured at 37 °C and 5% CO2 in the standard mammalian medium DMEM/F12 supplemented with 10% fetal bovine serum (FBS). Of nine initial biopsies, two cell lines were established from two different animals and designated HuWa1 and HuWa2. The cells have a stable karyotype with 2n=44, which has commonly been observed in other baleen whale species. Cells were verified as being fibroblasts based on their spindle-shaped morphology, adherence to plastic and positive immunoreaction to vimentin. Population doubling time was determined to be ∼41 h and cells were successfully cryopreserved and thawed. To date, HuWa1 cells have been propagated 30 times. Cells proliferate at the tested temperatures, 30, 33.5 and 37 °C, but show the highest rate of proliferation at 37 °C. Short-term exposure to para,para'-dichlorodiphenyldichloroethylene (p,p'-DDE), a priority compound accumulating in southern hemisphere humpback whales, resulted in a concentration-dependent loss of cell viability. The effective concentration which caused a 50% reduction in HuWa1 cell viability (EC50 value) was approximately six times greater than the EC50 value for the same chemical measured with human dermal fibroblasts. HuWa1 exposed to a natural, p,p'-DDE-containing, chemical mixture extracted from whale blubber showed distinctively higher sensitivity than to p,p'-DDE alone. Thus, we provide the first cytotoxicological data for humpback whales and with establishment of the HuWa cell lines, a unique in vitro model for the study of the whales' sensitivity and cellular response to chemicals and other environmental stressors.


Subject(s)
Humpback Whale/physiology , Adipose Tissue/chemistry , Animals , Cell Line , Cell Survival/drug effects , Dichlorodiphenyl Dichloroethylene/metabolism , Dichlorodiphenyl Dichloroethylene/toxicity , Fibroblasts/cytology , Inhibitory Concentration 50 , Risk Assessment , Water Pollutants, Chemical/metabolism , Water Pollutants, Chemical/toxicity
16.
Vet J ; 202(3): 416-24, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25457267

ABSTRACT

There has been unprecedented interest in recent years in the use of stem cells as therapy for an array of diseases in companion animals. Stem cells have already been deployed therapeutically in a number of clinical settings, in particular the use of mesenchymal stem cells to treat osteoarthritis in horses and dogs. However, an assessment of the scientific literature highlights a marked disparity between the purported benefits of stem cell therapies and their proven abilities as defined by rigorously controlled scientific studies. Although preliminary data generated from clinical trials in human patients are encouraging, therapies currently available to treat animals are supported by very limited clinical evidence, and the commercialisation of these treatments may be premature. This review introduces the three main types of stem cells relevant to veterinary applications, namely, embryonic stem cells, induced pluripotent stem cells, and mesenchymal stem cells, and draws together research findings from in vitro and in vivo studies to give an overview of current stem cell therapies for the treatment of osteoarthritis in animals. Recent advances in tissue engineering, which is proposed as the future direction of stem cell-based therapy for osteoarthritis, are also discussed.


Subject(s)
Embryonic Stem Cells/transplantation , Induced Pluripotent Stem Cells/transplantation , Osteoarthritis/veterinary , Stem Cell Transplantation/veterinary , Tissue Engineering/veterinary , Animals , Mesenchymal Stem Cell Transplantation/veterinary , Osteoarthritis/prevention & control , Stem Cell Transplantation/statistics & numerical data
17.
Stem Cell Res ; 13(2): 251-61, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108530

ABSTRACT

Optimization of pluripotent stem cell expansion and differentiation is facilitated by biological tools that permit non-invasive and dynamic monitoring of pluripotency, and the ability to select for an undifferentiated input cell population. Here we report on the generation and characterisation of clonal human embryonic stem (HES3, H9) and human induced pluripotent stem cell lines (UQEW01i-epifibC11) that have been stably modified with an artificial EOS(C3+) promoter driving expression of EGFP and puromycin resistance-conferring proteins. We show that EGFP expression faithfully reports on the pluripotency status of the cells in these lines and that antibiotic selection allows for an efficient elimination of differentiated cells from the cultures. We demonstrate that the extinction of the expression of the pluripotency reporter during differentiation closely correlates with the decrease in expression of conventional pluripotency markers, such as OCT4 (POU5F1), TRA-1-60 and SSEA4 when screening across conditions with various levels of pluripotency-maintaining or differentiation-inducing signals. We further illustrate the utility of these lines for real-time monitoring of pluripotency in embryoid bodies and microfluidic bioreactors.


Subject(s)
Embryonic Stem Cells/metabolism , Genes, Reporter , Induced Pluripotent Stem Cells/metabolism , Transgenes , Anti-Bacterial Agents/pharmacology , Bioreactors , Cell Culture Techniques/instrumentation , Cell Differentiation , Cell Line , Cell Proliferation , Drug Resistance, Bacterial/genetics , Gene Expression Regulation, Developmental , Genetic Markers , Genetic Vectors , Genotype , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Lentivirus/genetics , Microfluidic Analytical Techniques , Phenotype , Puromycin/pharmacology , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transduction, Genetic
18.
Stem Cells Dev ; 23(24): 3021-33, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25055193

ABSTRACT

In this study we have generated canine mesenchymal stromal cells (MSCs), also known as mesenchymal stem cells, from canine induced pluripotent stem cells (ciPSCs) by small-molecule inhibition of the transforming growth factor beta (TGFß)/activin signaling pathway. These ciPSC-derived MSCs (ciPSC-MSCs) express the MSC markers CD73, CD90, CD105, STRO1, cPDGFRß and cKDR, in addition to the pluripotency factors OCT4, NANOG and REX1. ciPSC-MSCs lack immunostaining for H3K27me3, suggesting that they possess two active X chromosomes. ciPSC-MSCs are highly proliferative and undergo robust differentiation along the osteo-, chondro- and adipogenic pathways, but do not form teratoma-like tissues in vitro. Of further significance for the translational potential of ciPSC-MSCs, we show that these cells can be encapsulated and maintained within injectable hydrogel matrices that, when functionalized with bound pentosan polysulfate, dramatically enhance chondrogenesis and inhibit osteogenesis. The ability to efficiently derive large numbers of highly proliferative canine MSCs from ciPSCs that can be incorporated into injectable, functionalized hydrogels that enhance their differentiation along a desired lineage constitutes an important milestone towards developing an effective MSC-based therapy for osteoarthritis in dogs, but equally provides a model system for assessing the efficacy and safety of analogous approaches for treating human degenerative joint diseases.


Subject(s)
Activins/metabolism , Chondrogenesis , Induced Pluripotent Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Osteogenesis , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cells, Cultured , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Dogs , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Histones/genetics , Histones/metabolism , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pentosan Sulfuric Polyester/pharmacology , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
Stem Cells Dev ; 23(13): 1515-23, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24555755

ABSTRACT

In this study we have reprogrammed dermal fibroblasts from an adult female horse into equine induced pluripotent stem cells (equiPSCs). These equiPSCs are dependent only on leukemia inhibitory factor (LIF), placing them in striking contrast to previously derived equiPSCs that have been shown to be co-dependent on both LIF and basic fibroblast growth factor (bFGF). These equiPSCs have a normal karyotype and have been maintained beyond 60 passages. They possess alkaline phosphatase activity and express eqNANOG, eqOCT4, and eqTERT mRNA. Immunocytochemistry confirmed that they produce NANOG, REX1, SSEA4, TRA1-60, and TRA1-81. While our equiPSCs are LIF dependent, bFGF co-stimulates their proliferation via the PI3K/AKT pathway. EquiPSCs lack expression of eqXIST and immunostaining for H3K27me3, suggesting that during reprogramming the inactive X chromosome has likely been reactivated to generate cells that have two active X chromosomes. EquiPSCs form embryoid bodies and in vitro teratomas that contain derivatives of all three germ layers. These LIF-dependent equiPSCs likely reflect a more naive state of pluripotency than equiPSCs that are co-dependent on both LIF and bFGF and so provide a novel resource for understanding pluripotency in the horse.


Subject(s)
Fibroblasts/physiology , Induced Pluripotent Stem Cells/physiology , Leukemia Inhibitory Factor/physiology , Skin/cytology , Animals , Biomarkers/metabolism , Cell Proliferation , Coculture Techniques , Feeder Cells , Female , Fibroblast Growth Factors/physiology , Gene Expression , Histones/metabolism , Horses , Transcription Factors/metabolism , X Chromosome/genetics
20.
Stem Cells Dev ; 21(12): 2288-97, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22221227

ABSTRACT

Dogs provide a more clinically relevant model of human disease than rodents, particularly with respect to hereditary diseases. Thus, the availability of canine stem cells will greatly facilitate the use of the dog in the development of stem cell-based gene therapies and regenerative medicine. In this study we describe the production of canine induced pluripotent stem cells (ciPSCs) from adult dermal fibroblasts. These cells have a morphology resembling previously described canine embryonic stem cells, a normal karyotype, and express pluripotency markers including alkaline phosphatase, Nanog, Oct4, Telomerase, SSEA1, SSEA4, TRA1-60, TRA1-81, and Rex1. Furthermore, the inactive X chromosome is reactivated indicating a ground-state pluripotency. In culture they readily form embryoid bodies, which in turn give rise to cell types from all 3 embryonic germ layers, as indicated by expression of the definitive endoderm markers Cxcr4 and α-fetoprotein, mesoderm markers Collagen IIA and Gata2, and ectoderm markers ßIII-tubulin, Enolase, and Nestin. Of particular significance is the observation that these ciPSCs are dependent only on leukemia inhibitory factor (LIF), making them similar to mouse and canine embryonic stem cells, but strikingly unlike the ciPSCs recently described in two other studies, which were dependent on both basic fibroblast growth factor and LIF in order to maintain their pluripotency. Thus, our ciPSCs closely resemble mouse ESCs derived from the inner cell mass of preimplantation embryos, while the previously described ciPSCs appear to be more representative of cells from the epiblast of mouse postimplantation embryos.


Subject(s)
Fibroblasts/physiology , Induced Pluripotent Stem Cells/metabolism , Lymphokines/physiology , Skin/pathology , Alkaline Phosphatase/metabolism , Animals , Cell Differentiation , Cell Nucleus/metabolism , Cell Shape , Coculture Techniques , Dogs , Female , Gene Expression , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/transplantation , Mice , Mice, Inbred NOD , Neoplasms, Experimental/pathology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Teratoma/pathology , Transgenes , X Chromosome Inactivation
SELECTION OF CITATIONS
SEARCH DETAIL
...