Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ibon Garitaonandia is active.

Publication


Featured researches published by Ibon Garitaonandia.


Cell Stem Cell | 2012

Recurrent Variations in DNA Methylation in Human Pluripotent Stem Cells and Their Differentiated Derivatives

Kristopher L. Nazor; Gulsah Altun; Candace L. Lynch; Ha Tran; Julie V. Harness; Ileana Slavin; Ibon Garitaonandia; Franz Josef Müller; Yu Chieh Wang; Francesca S. Boscolo; Eyitayo Fakunle; Biljana Dumevska; S.J Lee; Hyun Sook Park; Tsaiwei Olee; Darryl D. D'Lima; Ruslan Semechkin; Mana M. Parast; Vasiliy Galat; Andrew L. Laslett; Uli Schmidt; Hans S. Keirstead; Jeanne F. Loring; Louise C. Laurent

Human pluripotent stem cells (hPSCs) are potential sources of cells for modeling disease and development, drug discovery, and regenerative medicine. However, it is important to identify factors that may impact the utility of hPSCs for these applications. In an unbiased analysis of 205 hPSC and 130 somatic samples, we identified hPSC-specific epigenetic and transcriptional aberrations in genes subject to X chromosome inactivation (XCI) and genomic imprinting, which were not corrected during directed differentiation. We also found that specific tissue types were distinguished by unique patterns of DNA hypomethylation, which were recapitulated by DNA demethylation during in vitro directed differentiation. Our results suggest that verification of baseline epigenetic status is critical for hPSC-based disease models in which the observed phenotype depends on proper XCI or imprinting and that tissue-specific DNA methylation patterns can be accurately modeled during directed differentiation of hPSCs, even in the presence of variations in XCI or imprinting.


Nature Methods | 2011

Induced pluripotent stem cells from highly endangered species

Inbar Friedrich Ben-Nun; Susanne C. Montague; Marlys L. Houck; Ha T. Tran; Ibon Garitaonandia; Trevor R. Leonardo; Yu-Chieh Wang; Suellen J. Charter; Louise C. Laurent; Oliver A. Ryder; Jeanne F. Loring

For some highly endangered species there are too few reproductively capable animals to maintain adequate genetic diversity, and extraordinary measures are necessary to prevent extinction. We report generation of induced pluripotent stem cells (iPSCs) from two endangered species: a primate, the drill, Mandrillus leucophaeus and the nearly extinct northern white rhinoceros, Ceratotherium simum cottoni. iPSCs may eventually facilitate reintroduction of genetic material into breeding populations.


Cell Research | 2011

Specific lectin biomarkers for isolation of human pluripotent stem cells identified through array-based glycomic analysis

Yu Chieh Wang; Masato Nakagawa; Ibon Garitaonandia; Ileana Slavin; Gulsah Altun; Robert M. Lacharite; Kristopher L. Nazor; Ha T. Tran; Candace L. Lynch; Trevor R. Leonardo; Ying Liu; Suzanne E. Peterson; Louise C. Laurent; Shinya Yamanaka; Jeanne F. Loring

Rapid and dependable methods for isolating human pluripotent stem cell (hPSC) populations are urgently needed for quality control in basic research and in cell-based therapy applications. Using lectin arrays, we analyzed glycoproteins extracted from 26 hPSC samples and 22 differentiated cell samples, and identified a small group of lectins with distinctive binding signatures that were sufficient to distinguish hPSCs from a variety of non-pluripotent cell types. These specific biomarkers were shared by all the 12 human embryonic stem cell and the 14 human induced pluripotent stem cell samples examined, regardless of the laboratory of origin, the culture conditions, the somatic cell type reprogrammed, or the reprogramming method used. We demonstrated a practical application of specific lectin binding by detecting hPSCs within a differentiated cell population with lectin-mediated staining followed by fluorescence microscopy and flow cytometry, and by enriching and purging viable hPSCs from mixed cell populations using lectin-mediated cell separation. Global gene expression analysis showed pluripotency-associated differential expression of specific fucosyltransferases and sialyltransferases, which may underlie these differences in protein glycosylation and lectin binding. Taken together, our results show that protein glycosylation differs considerably between pluripotent and non-pluripotent cells, and demonstrate that lectins may be used as biomarkers to monitor pluripotency in stem cell populations and for removal of viable hPSCs from mixed cell populations.


PLOS ONE | 2015

Increased Risk of Genetic and Epigenetic Instability in Human Embryonic Stem Cells Associated with Specific Culture Conditions

Ibon Garitaonandia; Hadar Amir; Francesca S. Boscolo; Gerald Wambua; Heather L. Schultheisz; Karen Sabatini; Robert Morey; Shannon Waltz; Yu-Chieh Wang; Ha Tran; Trevor R. Leonardo; Kristopher L. Nazor; Ileana Slavin; Candace L. Lynch; Yingchun Li; Ronald Coleman; Irene Gallego Romero; Gulsah Altun; David Reynolds; Stephen Dalton; Mana M. Parast; Jeanne F. Loring; Louise C. Laurent

The self-renewal and differentiation capacities of human pluripotent stem cells (hPSCs) make them a promising source of material for cell transplantation therapy, drug development, and studies of cellular differentiation and development. However, the large numbers of cells necessary for many of these applications require extensive expansion of hPSC cultures, a process that has been associated with genetic and epigenetic alterations. We have performed a combinatorial study on both hESCs and hiPSCs to compare the effects of enzymatic vs. mechanical passaging, and feeder-free vs. mouse embryonic fibroblast feeder substrate, on the genetic and epigenetic stability and the phenotypic characteristics of hPSCs. In extensive experiments involving over 100 continuous passages, we observed that both enzymatic passaging and feeder-free culture were associated with genetic instability, higher rates of cell proliferation, and persistence of OCT4/POU5F1-positive cells in teratomas, with enzymatic passaging having the stronger effect. In all combinations of culture conditions except for mechanical passaging on feeder layers, we noted recurrent deletions in the genomic region containing the tumor suppressor gene TP53, which was associated with decreased mRNA expression of TP53, as well as alterations in the expression of several downstream genes consistent with a decrease in the activity of the TP53 pathway. Among the hESC cultures, we also observed culture-associated variations in global gene expression and DNA methylation. The effects of enzymatic passaging and feeder-free conditions were also observed in hiPSC cultures. Our results highlight the need for careful assessment of the effects of culture conditions on cells intended for clinical therapies.


Cell Transplantation | 2015

Proof of concept studies exploring the safety and functional activity of human parthenogenetic-derived neural stem cells for the treatment of Parkinson's disease.

Rodolfo Gonzalez; Ibon Garitaonandia; Andrew M. Crain; Poustovoitov M; Tatiana Abramihina; Alexander Noskov; Jiang C; Robert Morey; Louise C. Laurent; John D. Elsworth; Evan Y. Snyder; D.E. Redmond; Ruslan Semechkin

Recent studies indicate that human pluripotent stem cell (PSC)-based therapies hold great promise in Parkinsons disease (PD). Clinical studies have shown that grafted fetal neural tissue can achieve considerable biochemical and clinical improvements in PD. However, the source of fetal tissue grafts is limited and ethically controversial. Human parthenogenetic stem cells offer a good alternative because they are derived from unfertilized oocytes without destroying viable human embryos and can be used to generate an unlimited supply of neural stem cells for transplantation. Here we evaluate for the first time the safety and engraftment of human parthenogenetic stem cell-derived neural stem cells (hpNSCs) in two animal models: 6-hydroxydopamine (6-OHDA)-lesioned rodents and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated nonhuman primates (NHPs). In both rodents and nonhuman primates, we observed successful engraftment and higher dopamine levels in hpNSC-transplanted animals compared to vehicle control animals, without any adverse events. These results indicate that hpNSCs are safe, well tolerated, and could potentially be a source for cell-based therapies in PD.


Scientific Reports | 2013

Deriving dopaminergic neurons for clinical use. A practical approach

Rodolfo Gonzalez; Ibon Garitaonandia; Tatiana Abramihina; Gerald Wambua; Alina Ostrowska; Mathew Brock; Alexander Noskov; Francesca S. Boscolo; John S. Craw; Louise C. Laurent; Evan Y. Snyder; Ruslan Semechkin

New small molecules that regulate the step-wise differentiation of human pluripotent stem cells into dopaminergic neurons have been identified. The steroid, guggulsterone, was found to be the most effective inducer of neural stem cells into dopaminergic neurons. These neurons are extensively characterized and shown to be functional. We believe this new approach offers a practical route to creating neurons of sufficient quality to be used to treat Parkinsons disease patients.


Cell Transplantation | 2016

Neural Stem Cells Derived from Human Parthenogenetic Stem Cells Engraft and Promote Recovery in a Nonhuman Primate Model of Parkinson's Disease.

Rodolfo Gonzalez; Ibon Garitaonandia; Poustovoitov M; Tatiana Abramihina; Caleb R.S. McEntire; Culp B; Attwood J; Alexander Noskov; Christiansen-Weber T; Marwa Khater; Sergio Mora-Castilla; Cuong To; Crain A; Sherman G; Andrey Semechkin; Louise C. Laurent; John D. Elsworth; John R. Sladek; Evan Y. Snyder; Kern Ra

Cell therapy has attracted considerable interest as a promising therapeutic alternative for patients with Parkinsons disease (PD). Clinical studies have shown that grafted fetal neural tissue can achieve considerable biochemical and clinical improvements in PD. However, the source of fetal tissue grafts is limited and ethically controversial. Human parthenogenetic stem cells offer a good alternative because they are derived from unfertilized oocytes without destroying potentially viable human embryos and can be used to generate an unlimited supply of neural cells for transplantation. We have previously reported that human parthenogenetic stem cell-derived neural stem cells (hpNSCs) successfully engraft, survive long term, and increase brain dopamine (DA) levels in rodent and nonhuman primate models of PD. Here we report the results of a 12-month transplantation study of hpNSCs in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned African green monkeys with moderate to severe clinical parkinsonian symptoms. The hpNSCs manufactured under current good manufacturing practice (cGMP) conditions were injected bilaterally into the striatum and substantia nigra of immunosuppressed monkeys. Transplantation of hpNSCs was safe and well tolerated by the animals with no dyskinesia, tumors, ectopic tissue formation, or other test article-related serious adverse events. We observed that hpNSCs promoted behavioral recovery; increased striatal DA concentration, fiber innervation, and number of dopaminergic neurons; and induced the expression of genes and pathways downregulated in PD compared to vehicle control animals. These results provide further evidence for the clinical translation of hpNSCs and support the approval of the worlds first pluripotent stem cell-based phase I/IIa study for the treatment of PD (Clinical Trial Identifier NCT02452723).


Journal of Visualized Experiments | 2011

Teratoma Generation in the Testis Capsule

Suzanne E. Peterson; Ha T. Tran; Ibon Garitaonandia; Sangyoon Han; Kyle S. Nickey; Trevor R. Leonardo; Louise C. Laurent; Jeanne F. Loring

Pluripotent stem cells (PSCs) have the unique characteristic that they can differentiate into cells from all three germ layers. This makes them a potentially valuable tool for the treatment of many different diseases. With the advent of induced pluripotent stem cells (iPSCs) and continuing research with human embryonic stem cells (hESCs) there is a need for assays that can demonstrate that a particular cell line is pluripotent. Germline transmission has been the gold standard for demonstrating the pluripotence of mouse embryonic stem cell (mESC) lines1,2,3. Using this assay, researchers can show that a mESC line can make all cell types in the embryo including germ cells4. With the generation of human ESC lines5,6, the appropriate assay to prove pluripotence of these cells was unclear since human ESCs cannot be tested for germline transmission. As a surrogate, the teratoma assay is currently used to demonstrate the pluripotency of human pluripotent stem cells (hPSCs)7,8,9. Though this assay has recently come under scrutiny and new technologies are being actively explored, the teratoma assay is the current gold standard7. In this assay, the cells in question are injected into an immune compromised mouse. If the cells are pluripotent, a teratoma will eventually develop and sections of the tumor will show tissues from all 3 germ layers10. In the teratoma assay, hPSCs can be injected into different areas of the mouse. The most common injection sites include the testis capsule, the kidney capsule, the liver; or into the leg either subcutaneously or intramuscularly11. Here we describe a robust protocol for the generation of teratomas from hPSCs using the testis capsule as the site for tumor growth.


Regenerative Medicine | 2012

In vitro differentiation of human parthenogenetic stem cells into neural lineages

Dmitry Isaev; Ibon Garitaonandia; Tatiana Abramihina; Tatjana Zogovic-Kapsalis; Richard West; Andrey Semechkin; Albrecht M. Müller; Ruslan Semechkin

Human parthenogenetic stem cells are derived from the inner cell mass of blastocysts obtained from unfertilized oocytes that have been stimulated to develop without any participation of male gamete. As parthenogenesis does not involve the destruction of a viable human embryo, the derivation and use of human parthenogenetic stem cells does not raise the same ethical concerns as conventional embryonic stem cells. Human parthenogenetic stem cells are similar to embryonic stem cells in their proliferation and multilineage in vitro differentiation capacity. The aim of this study is to derive multipotent neural stem cells from human parthenogenetic stem cells that are stable to passaging and cryopreservation, and have the ability to further differentiate into functional neurons. Immunocytochemistry, quantitative real-time PCR, or FACS were used to confirm that the derived neural stem cells express neural markers such as NES, SOX2 and MS1. The derived neural stem cells keep uniform morphology for at least 30 passages and can be spontaneously differentiated into cells with neuron morphology that express TUBB3 and MAP2, and fire action potentials. These results suggest that parthenogenetic stem cells are a very promising and potentially unlimited source for the derivation of multipotent neural stem cells that can be used for therapeutic applications.


Stem Cells | 2017

Spontaneous Single-Copy Loss of TP53 in Human Embryonic Stem Cells Markedly Increases Cell Proliferation and Survival.

Hadar Amir; Thomas Touboul; Karen Sabatini; Divya Chhabra; Ibon Garitaonandia; Jeanne F. Loring; Robert Morey; Louise C. Laurent

Genomic aberrations have been identified in many human pluripotent stem cell (hPSC) cultures. Commonly observed duplications in portions of chromosomes 12p and 17q have been associated with increases in genetic instability and resistance to apoptosis, respectively. However, the phenotypic consequences related to sporadic mutations have not been evaluated to date. Here, we report on the effects of a single‐copy deletion of the chr17p13.1 region, a sporadic mutation that spontaneously arose independently in several subclones of a human embryonic stem cell culture. Compared to cells with two normal copies of chr17p13.1 (“wild‐type”), the cells with a single‐copy deletion of this region (“mutant”) displayed a selective advantage when exposed to stressful conditions, and retained a higher percentage of cells expressing the pluripotency marker POU5F1/OCT4 after 2 weeks of in vitro differentiation. Knockdown of TP53, which is a gene encompassed by the deleted region, in wild‐type cells mimicked the chr17p13.1 deletion phenotype. Thus, sporadic mutations in hPSCs can have phenotypic effects that may impact their utility for clinical applications. Stem Cells 2017;35:872–885

Collaboration


Dive into the Ibon Garitaonandia's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeanne F. Loring

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerald Wambua

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Ha T. Tran

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Candace L. Lynch

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gulsah Altun

Georgia State University

View shared research outputs
Top Co-Authors

Avatar

Ileana Slavin

Scripps Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge