Network


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

Hotspot


Dive into the research topics where Tomonari Hayama is active.

Publication


Featured researches published by Tomonari Hayama.


Nature | 2017

Correction of a pathogenic gene mutation in human embryos

Hong Ma; Nuria Marti-Gutierrez; Sang Wook Park; Jun Wu; Yeonmi Lee; Keiichiro Suzuki; Amy Koski; Dongmei Ji; Tomonari Hayama; Riffat Ahmed; Hayley Darby; Crystal Van Dyken; Ying Li; Eunju Kang; A. Reum Park; Daesik Kim; Jianhui Gong; Ying Gu; Xun Xu; David Battaglia; Sacha Krieg; David M. Lee; Diana H. Wu; Don P. Wolf; Stephen Heitner; Juan Carlos Izpisua Belmonte; Paula Amato; Jin-Soo Kim; Sanjiv Kaul; Shoukhrat Mitalipov

Genome editing has potential for the targeted correction of germline mutations. Here we describe the correction of the heterozygous MYBPC3 mutation in human preimplantation embryos with precise CRISPR–Cas9-based targeting accuracy and high homology-directed repair efficiency by activating an endogenous, germline-specific DNA repair response. Induced double-strand breaks (DSBs) at the mutant paternal allele were predominantly repaired using the homologous wild-type maternal gene instead of a synthetic DNA template. By modulating the cell cycle stage at which the DSB was induced, we were able to avoid mosaicism in cleaving embryos and achieve a high yield of homozygous embryos carrying the wild-type MYBPC3 gene without evidence of off-target mutations. The efficiency, accuracy and safety of the approach presented suggest that it has potential to be used for the correction of heritable mutations in human embryos by complementing preimplantation genetic diagnosis. However, much remains to be considered before clinical applications, including the reproducibility of the technique with other heterozygous mutations.


Nature | 2015

Metabolic rescue in pluripotent cells from patients with mtDNA disease

Hong Ma; Clifford D.L. Folmes; Jun Wu; Robert Morey; Sergio Mora-Castilla; Alejandro Ocampo; Li Ma; Joanna Poulton; Xinjian Wang; Riffat Ahmed; Eunju Kang; Yeonmi Lee; Tomonari Hayama; Ying Li; Crystal Van Dyken; Nuria Marti Gutierrez; Rebecca Tippner-Hedges; Amy Koski; Nargiz Mitalipov; Paula Amato; Don P. Wolf; Taosheng Huang; Andre Terzic; Louise C. Laurent; Juan Carlos Izpisua Belmonte; Shoukhrat Mitalipov

Mitochondria have a major role in energy production via oxidative phosphorylation, which is dependent on the expression of critical genes encoded by mitochondrial (mt)DNA. Mutations in mtDNA can cause fatal or severely debilitating disorders with limited treatment options. Clinical manifestations vary based on mutation type and heteroplasmy (that is, the relative levels of mutant and wild-type mtDNA within each cell). Here we generated genetically corrected pluripotent stem cells (PSCs) from patients with mtDNA disease. Multiple induced pluripotent stem (iPS) cell lines were derived from patients with common heteroplasmic mutations including 3243A>G, causing mitochondrial encephalomyopathy and stroke-like episodes (MELAS), and 8993T>G and 13513G>A, implicated in Leigh syndrome. Isogenic MELAS and Leigh syndrome iPS cell lines were generated containing exclusively wild-type or mutant mtDNA through spontaneous segregation of heteroplasmic mtDNA in proliferating fibroblasts. Furthermore, somatic cell nuclear transfer (SCNT) enabled replacement of mutant mtDNA from homoplasmic 8993T>G fibroblasts to generate corrected Leigh-NT1 PSCs. Although Leigh-NT1 PSCs contained donor oocyte wild-type mtDNA (human haplotype D4a) that differed from Leigh syndrome patient haplotype (F1a) at a total of 47 nucleotide sites, Leigh-NT1 cells displayed transcriptomic profiles similar to those in embryo-derived PSCs carrying wild-type mtDNA, indicative of normal nuclear-to-mitochondrial interactions. Moreover, genetically rescued patient PSCs displayed normal metabolic function compared to impaired oxygen consumption and ATP production observed in mutant cells. We conclude that both reprogramming approaches offer complementary strategies for derivation of PSCs containing exclusively wild-type mtDNA, through spontaneous segregation of heteroplasmic mtDNA in individual iPS cell lines or mitochondrial replacement by SCNT in homoplasmic mtDNA-based disease.


Nature | 2016

Mitochondrial replacement in human oocytes carrying pathogenic mitochondrial DNA mutations

Eunju Kang; Jun Wu; Nuria Marti Gutierrez; Amy Koski; Rebecca Tippner-Hedges; Karen Agaronyan; Aida Platero-Luengo; Paloma Martínez-Redondo; Hong Ma; Yeonmi Lee; Tomonari Hayama; Crystal Van Dyken; Xinjian Wang; Shiyu Luo; Riffat Ahmed; Ying Li; Dongmei Ji; Refik Kayali; Cengiz Cinnioglu; Susan B. Olson; Jeffrey T. Jensen; David Battaglia; David M. Lee; Diana Wu; Taosheng Huang; Don P. Wolf; Dmitry Temiakov; Juan Carlos Izpisua Belmonte; Paula Amato; Shoukhrat Mitalipov

Maternally inherited mitochondrial (mt)DNA mutations can cause fatal or severely debilitating syndromes in children, with disease severity dependent on the specific gene mutation and the ratio of mutant to wild-type mtDNA (heteroplasmy) in each cell and tissue. Pathogenic mtDNA mutations are relatively common, with an estimated 778 affected children born each year in the United States. Mitochondrial replacement therapies or techniques (MRT) circumventing mother–to–child mtDNA disease transmission involve replacement of oocyte maternal mtDNA. Here we report MRT outcomes in several families with common mtDNA syndromes. The mother’s oocytes were of normal quality and mutation levels correlated with those in existing children. Efficient replacement of oocyte mutant mtDNA was performed by spindle transfer, resulting in embryos containing >99% donor mtDNA. Donor mtDNA was stably maintained in embryonic stem cells (ES cells) derived from most embryos. However, some ES cell lines demonstrated gradual loss of donor mtDNA and reversal to the maternal haplotype. In evaluating donor–to–maternal mtDNA interactions, it seems that compatibility relates to mtDNA replication efficiency rather than to mismatch or oxidative phosphorylation dysfunction. We identify a polymorphism within the conserved sequence box II region of the D-loop as a plausible cause of preferential replication of specific mtDNA haplotypes. In addition, some haplotypes confer proliferative and growth advantages to cells. Hence, we propose a matching paradigm for selecting compatible donor mtDNA for MRT.


Cell Stem Cell | 2017

Functional Human Oocytes Generated by Transfer of Polar Body Genomes

Hong Ma; Ryan C. O’Neil; Nuria Marti Gutierrez; Manoj Hariharan; Zhuzhu Z. Zhang; Yupeng He; Cengiz Cinnioglu; Refik Kayali; Eunju Kang; Yeonmi Lee; Tomonari Hayama; Amy Koski; Joseph R. Nery; Rosa Castanon; Rebecca Tippner-Hedges; Riffat Ahmed; Crystal Van Dyken; Ying Li; Susan B. Olson; David Battaglia; David M. Lee; Diana H. Wu; Paula Amato; Don P. Wolf; Joseph R. Ecker; Shoukhrat Mitalipov

Oocyte defects lie at the heart of some forms of infertility and could potentially be addressed therapeutically by alternative routes for oocyte formation. Here, we describe the generation of functional human oocytes following nuclear transfer of first polar body (PB1) genomes from metaphase II (MII) oocytes into enucleated donor MII cytoplasm (PBNT). The reconstructed oocytes supported the formation of de novo meiotic spindles and, after fertilization with sperm, meiosis completion and formation of normal diploid zygotes. While PBNT zygotes developed to blastocysts less frequently (42%) than controls (75%), genome-wide genetic, epigenetic, and transcriptional analyses of PBNT and control ESCs indicated comparable numbers of structural variations and markedly similar DNA methylation and transcriptome profiles. We conclude that rescue of PB1 genetic material via introduction into donor cytoplasm may offer a source of oocytes for infertility treatment or mitochondrial replacement therapy for mtDNA disease.


The EMBO Journal | 2017

Mitochondrial genome inheritance and replacement in the human germline

Don P. Wolf; Tomonari Hayama; Shoukhrat Mitalipov

Mitochondria, the ubiquitous power packs in nearly every eukaryotic cell, contain their own DNA, known as mtDNA, which is inherited exclusively from the mother. The number of mitochondrial genomes varies depending on the cells energy needs. The mature oocyte contains the highest number of mitochondria of any cell type, although there is little if any mtDNA replication after fertilization until the embryo implants. This has potential repercussions for mitochondrial replacement therapy (MRT; see description of currently employed methods below) used to prevent the transmission of mtDNA‐based disorders. If only a few mitochondria with defective mtDNA are left in the embryo and undergo extensive replication, it might therefore thwart the purpose of MRT. In order to improve the safety and efficacy of this experimental therapy, we need a better understanding of how and which mtDNA is tagged for replication versus transcription after fertilization of the oocyte.


Stem Cells | 2017

Concise Review: Embryonic Stem Cells Derived by Somatic Cell Nuclear Transfer: A Horse in the Race?

Don P. Wolf; Robert Morey; Eunju Kang; Hong Ma; Tomonari Hayama; Louise C. Laurent; Shoukhrat Mitalipov

Embryonic stem cells (ESC) hold promise for the treatment of human medical conditions but are allogeneic. Here, we consider the differences between autologous pluripotent stem cells produced by nuclear transfer (NT‐ESCs) and transcription factor‐mediated, induced pluripotent stem cells (iPSCs) that impact the desirability of each of these cell types for clinical use. The derivation of NT‐ESCs is more cumbersome and requires donor oocytes; however, the use of oocyte cytoplasm as the source of reprogramming factors is linked to a key advantage of NT‐ESCs—the ability to replace mutant mitochondrial DNA in a patient cell (due to either age or inherited disease) with healthy donor mitochondria from an oocyte. Moreover, in epigenomic and transcriptomic comparisons between isogenic iPSCs and NT‐ESCs, the latter produced cells that more closely resemble bona fide ESCs derived from fertilized embryos. Thus, although NT‐ESCs are more difficult to generate than iPSCs, the ability of somatic cell nuclear transfer to replace aged or diseased mitochondria and the closer epigenomic and transcriptomic similarity between NT‐ESCs and bona fide ESCs may make NT‐ESCs superior for future applications in regenerative medicine. Stem Cells 2017;35:26–34


Stem Cells | 2016

Embryonic Stem Cells Derived by Somatic Cell Nuclear Transfer: A Horse in the Race?

Don P. Wolf; Robert Morey; Eunju Kang; Hong Ma; Tomonari Hayama; Louise C. Laurent; Shoukhrat Mitalipov

Embryonic stem cells (ESC) hold promise for the treatment of human medical conditions but are allogeneic. Here, we consider the differences between autologous pluripotent stem cells produced by nuclear transfer (NT‐ESCs) and transcription factor‐mediated, induced pluripotent stem cells (iPSCs) that impact the desirability of each of these cell types for clinical use. The derivation of NT‐ESCs is more cumbersome and requires donor oocytes; however, the use of oocyte cytoplasm as the source of reprogramming factors is linked to a key advantage of NT‐ESCs—the ability to replace mutant mitochondrial DNA in a patient cell (due to either age or inherited disease) with healthy donor mitochondria from an oocyte. Moreover, in epigenomic and transcriptomic comparisons between isogenic iPSCs and NT‐ESCs, the latter produced cells that more closely resemble bona fide ESCs derived from fertilized embryos. Thus, although NT‐ESCs are more difficult to generate than iPSCs, the ability of somatic cell nuclear transfer to replace aged or diseased mitochondria and the closer epigenomic and transcriptomic similarity between NT‐ESCs and bona fide ESCs may make NT‐ESCs superior for future applications in regenerative medicine. Stem Cells 2017;35:26–34


PLOS ONE | 2018

Germline and somatic mtDNA mutations in mouse aging

Hong Ma; Yeonmi Lee; Tomonari Hayama; Crystal Van Dyken; Nuria Marti-Gutierrez; Ying Li; Riffat Ahmed; Amy Koski; Eunju Kang; Hayley Darby; Thanasup Gonmanee; Younjung Park; Don P. Wolf; Chong Jai Kim; Shoukhrat Mitalipov

The accumulation of acquired mitochondrial genome (mtDNA) mutations with aging in somatic cells has been implicated in mitochondrial dysfunction and linked to age-onset diseases in humans. Here, we asked if somatic mtDNA mutations are also associated with aging in the mouse. MtDNA integrity in multiple organs and tissues in young and old (2–34 months) wild type (wt) mice was investigated by whole genome sequencing. Remarkably, no acquired somatic mutations were detected in tested tissues. However, we identified several non-synonymous germline mtDNA variants whose heteroplasmy levels (ratio of normal to mutant mtDNA) increased significantly with aging suggesting clonal expansion of inherited mtDNA mutations. Polg mutator mice, a model for premature aging, exhibited both germline and somatic mtDNA mutations whose numbers and heteroplasmy levels increased significantly with age implicating involvement in premature aging. Our results suggest that, in contrast to humans, acquired somatic mtDNA mutations do not accompany the aging process in wt mice.


Stem Cells | 2016

NT‐ESCs: A Horse in the Race?

Don P. Wolf; Robert Morey; Eunju Kang; Hong Ma; Tomonari Hayama; Louise C. Laurent; Shoukhrat Mitalipov

Embryonic stem cells (ESC) hold promise for the treatment of human medical conditions but are allogeneic. Here, we consider the differences between autologous pluripotent stem cells produced by nuclear transfer (NT‐ESCs) and transcription factor‐mediated, induced pluripotent stem cells (iPSCs) that impact the desirability of each of these cell types for clinical use. The derivation of NT‐ESCs is more cumbersome and requires donor oocytes; however, the use of oocyte cytoplasm as the source of reprogramming factors is linked to a key advantage of NT‐ESCs—the ability to replace mutant mitochondrial DNA in a patient cell (due to either age or inherited disease) with healthy donor mitochondria from an oocyte. Moreover, in epigenomic and transcriptomic comparisons between isogenic iPSCs and NT‐ESCs, the latter produced cells that more closely resemble bona fide ESCs derived from fertilized embryos. Thus, although NT‐ESCs are more difficult to generate than iPSCs, the ability of somatic cell nuclear transfer to replace aged or diseased mitochondria and the closer epigenomic and transcriptomic similarity between NT‐ESCs and bona fide ESCs may make NT‐ESCs superior for future applications in regenerative medicine. Stem Cells 2017;35:26–34


Cell Stem Cell | 2016

Age-Related Accumulation of Somatic Mitochondrial DNA Mutations in Adult-Derived Human iPSCs.

Eunju Kang; Xinjian Wang; Rebecca Tippner-Hedges; Hong Ma; Clifford D.L. Folmes; Nuria Marti Gutierrez; Yeonmi Lee; Crystal Van Dyken; Riffat Ahmed; Ying Li; Amy Koski; Tomonari Hayama; Shiyu Luo; Cary O. Harding; Paula Amato; Jeffrey T. Jensen; David Battaglia; David M. Lee; Diana Wu; Andre Terzic; Don P. Wolf; Taosheng Huang; Shoukhrat Mitalipov

Collaboration


Dive into the Tomonari Hayama's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong Ma

Oregon National Primate Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge