Network


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

Hotspot


Dive into the research topics where Momoko Yoshimoto is active.

Publication


Featured researches published by Momoko Yoshimoto.


Nature | 2009

Biomechanical forces promote embryonic haematopoiesis

Luigi Adamo; Olaia Naveiras; Pamela L. Wenzel; Shannon McKinney-Freeman; Peter Mack; Jorge Gracia-Sancho; Astrid Suchy-Dicey; Momoko Yoshimoto; M. William Lensch; Mervin C. Yoder; Guillermo García-Cardeña; George Q. Daley

Biomechanical forces are emerging as critical regulators of embryogenesis, particularly in the developing cardiovascular system. After initiation of the heartbeat in vertebrates, cells lining the ventral aspect of the dorsal aorta, the placental vessels, and the umbilical and vitelline arteries initiate expression of the transcription factor Runx1 (refs 3–5), a master regulator of haematopoiesis, and give rise to haematopoietic cells. It remains unknown whether the biomechanical forces imposed on the vascular wall at this developmental stage act as a determinant of haematopoietic potential. Here, using mouse embryonic stem cells differentiated in vitro, we show that fluid shear stress increases the expression of Runx1 in CD41+c-Kit+ haematopoietic progenitor cells, concomitantly augmenting their haematopoietic colony-forming potential. Moreover, we find that shear stress increases haematopoietic colony-forming potential and expression of haematopoietic markers in the para-aortic splanchnopleura/aorta–gonads–mesonephros of mouse embryos and that abrogation of nitric oxide, a mediator of shear-stress-induced signalling, compromises haematopoietic potential in vitro and in vivo. Collectively, these data reveal a critical role for biomechanical forces in haematopoietic development.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Embryonic day 9 yolk sac and intra-embryonic hemogenic endothelium independently generate a B-1 and marginal zone progenitor lacking B-2 potential

Momoko Yoshimoto; Encarnacion Montecino-Rodriguez; Michael J. Ferkowicz; Prashanth Porayette; W. Christopher Shelley; Simon J. Conway; Kenneth Dorshkind; Mervin C. Yoder

The majority of B lymphocytes in the adult mouse are generated in the bone marrow from hematopoietic stem cells (HSCs) that first appear in the aorta-gonado-mesonephros region of the fetus on embryonic day (E) 10.5–11. Comparatively less is known about B-cell development during embryogenesis. For example, which specific embryonic tissues participate in B lymphopoiesis and whether hematopoietic differentiation is skewed toward specific B-cell subsets in the embryo are unanswered questions, because the systemic circulation is initiated early during embryogenesis, resulting in an admixture of cells potentially originating from multiple sites. We demonstrate, using Ncx1−/− mice that lack systemic blood circulation, that the E9 yolk sac (YS) and the intra-embryonic para-aortic splanchnopleura (P-Sp) tissues independently give rise to AA4.1+CD19+B220lo-neg B progenitor cells that preferentially differentiate into innate type B-1 and marginal zone (MZ) B cells but not into B-2 cells upon transplantation. We have further demonstrated that these B-1 progenitor cells arise directly from YS and P-Sp hemogenic endothelium. These results document the initial wave of innate B lymphopoietic progenitor cells available for seeding the fetal liver at E11. The results of these studies expand our knowledge of hemogenic endothelial sites specifying distinct B-1 and MZ cell fates apart from B-2 cells and independent of an HSC origin during development.


Nature Communications | 2013

Early dynamic fate changes in haemogenic endothelium characterized at the single-cell level

Gemma Swiers; Claudia Baumann; O'Rourke Jf; Eleni Giannoulatou; Stephen Taylor; Anagha Joshi; Victoria Moignard; Cristina Pina; Thomas Bee; Konstantinos D. Kokkaliaris; Momoko Yoshimoto; Mervin C. Yoder; Jon Frampton; Timm Schroeder; Tariq Enver; Berthold Göttgens; Marella de Bruijn

Haematopoietic stem cells (HSCs) are the founding cells of the adult haematopoietic system, born during ontogeny from a specialized subset of endothelium, the haemogenic endothelium (HE) via an endothelial-to-haematopoietic transition (EHT). Although recently imaged in real time, the underlying mechanism of EHT is still poorly understood. We have generated a Runx1 +23 enhancer-reporter transgenic mouse (23GFP) for the prospective isolation of HE throughout embryonic development. Here we perform functional analysis of over 1,800 and transcriptional analysis of 268 single 23GFP(+) HE cells to explore the onset of EHT at the single-cell level. We show that initiation of the haematopoietic programme occurs in cells still embedded in the endothelial layer, and is accompanied by a previously unrecognized early loss of endothelial potential before HSCs emerge. Our data therefore provide important insights on the timeline of early haematopoietic commitment.


Nature Biotechnology | 2014

Differentiation of human pluripotent stem cells to cells similar to cord-blood endothelial colony–forming cells

Nutan Prasain; Man Ryul Lee; Sasidhar Vemula; Jonathan Luke Meador; Momoko Yoshimoto; Michael J. Ferkowicz; Alexa Fett; Manav Gupta; Brian M. Rapp; Mohammad Reza Saadatzadeh; Michael Ginsberg; Olivier Elemento; Younghee Lee; Sherry L. Voytik-Harbin; Hyung Min Chung; Ki Sung Hong; Emma Reid; Christina O'Neill; Reinhold Medina; Alan W. Stitt; Michael P. Murphy; Shahin Rafii; Hal E. Broxmeyer; Mervin C. Yoder

The ability to differentiate human pluripotent stem cells into endothelial cells with properties of cord-blood endothelial colony–forming cells (CB-ECFCs) may enable the derivation of clinically relevant numbers of highly proliferative blood vessel–forming cells to restore endothelial function in patients with vascular disease. We describe a protocol to convert human induced pluripotent stem cells (hiPSCs) or embryonic stem cells (hESCs) into cells similar to CB-ECFCs at an efficiency of >108 ECFCs produced from each starting pluripotent stem cell. The CB-ECFC-like cells display a stable endothelial phenotype with high clonal proliferative potential and the capacity to form human vessels in mice and to repair the ischemic mouse retina and limb, and they lack teratoma formation potential. We identify Neuropilin-1 (NRP-1)-mediated activation of KDR signaling through VEGF165 as a critical mechanism for the emergence and maintenance of CB-ECFC-like cells.


Development | 2004

Development of primitive and definitive hematopoiesis from nonhuman primate embryonic stem cells in vitro

Katsutsugu Umeda; Toshio Heike; Momoko Yoshimoto; Mitsutaka Shiota; Hirofumi Suemori; Hong-Yuan Luo; David H.K. Chui; Ryuzo Torii; Masabumi Shibuya; Norio Nakatsuji; Tatsutoshi Nakahata

Although information about the development of primitive and definitive hematopoiesis has been elucidated in murine embryos and embryonic stem (ES) cells, there have been few in vitro studies of these processes in primates. In this study, we investigated hematopoietic differentiation from cynomolgus monkey ES cells grown on OP9, a stromal cell line deficient in macrophage colony-stimulating factor. Primitive erythrocytes (EryP) and definitive erythrocytes (EryD) developed sequentially from ES cells in the culture system; this was confirmed by immunostaining and reverse transcriptase-polymerase chain reaction (RT-PCR) analysis of embryonic, fetal and adult globin genes. EryP were detected on day 8 without exogenous erythropoietin (EPO), whereas EryD appeared on day 16 and had an indispensable requirement for exogenous EPO. RT-PCR analysis of the cultures revealed a sequential expression of genes associated with primitive and definitive hematopoietic development that was equivalent to that seen during primate ontogeny in vivo. Vascular endothelial growth factor (VEGF) increased, in a dose-dependent manner, not only the number of floating hematopoietic cells, but also the number of adherent hematopoietic cell clusters containing CD34-positive immature progenitors. In colony assays, exogenous VEGF also had a dose-dependent stimulatory effect on the generation of primitive erythroid colonies. More efficient primitive and definitive erythropoiesis was induced by re-plating sorted CD34-positive cells. Thus, this system reproduces early hematopoietic development in vitro and can serve as a model for analyzing the mechanisms of hematopoietic development in primates.


The FASEB Journal | 2009

Generation of transplantable, functional satellite-like cells from mouse embryonic stem cells

Hsi Chang; Momoko Yoshimoto; Katsutsugu Umeda; Toru Iwasa; Yuta Mizuno; So-ichiro Fukada; Hiroshi Yamamoto; Norio Motohashi; Yuko Miyagoe-Suzuki; Shin'ichi Takeda; Toshio Heike; Tatsutoshi Nakahata

Satellite cells are myogenic stem cells responsible for the postnatal regeneration of skeletal muscle. Here we report the successful in vitro induction of Pax7‐positive satellite‐like cells from mouse embryonic stem (mES) cells. Embryoid bodies were generated from mES cells and cultured on Matrigel‐coated dishes with Dulbeccos modified Eagle medium containing fetal bovine serum and horse serum. Pax7‐positive satellite‐like cells were enriched by fluorescence‐activated cell sorting using a novel anti‐satellite cell antibody, SM/C‐2.6. SM/C‐2.6‐positive cells efficiently differentiate into skeletal muscle fibers both in vitro and in vivo. Furthermore, the cells demonstrate satellite cell characteristics such as extensive self‐renewal capacity in subsequent muscle injury model, long‐term engraft‐ment up to 24 wk, and the ability to be secondarily transplanted with remarkably high engraftment efficiency compared to myoblast transplantation. This is the first report of transplantable, functional satellite‐like cells derived from mES cells and will provide a foundation for new therapies for degenerative muscle disor‐ders.—Chang, H.,Yoshimoto, M., Umeda, K., Iwasa, T., Mizuno, Y., Fukada, S., Yamamoto, H., Motohashi, N., Yuko‐Miyagoe‐Suzuki, Takeda, S., Heike, T., Nakahata, T. Generation of transplantable, functional satellite‐like cells from mouse embryonic stem cells. FASEB J. 23, 1907–1919 (2009)


Blood | 2012

Autonomous murine T-cell progenitor production in the extra-embryonic yolk sac before HSC emergence

Momoko Yoshimoto; Prashanth Porayette; Nicole L. Glosson; Simon J. Conway; Nadia Carlesso; Angelo A. Cardoso; Mark H. Kaplan; Mervin C. Yoder

The extra-embryonic yolk sac (YS) is the first hematopoietic site in the mouse embryo and is thought to generate only primitive erythroid and myeloerythroid progenitor cells before definitive HSC emergence within the embryo on E10.5. Here, we have shown the existence of T cell-restricted progenitors in the E9.5 YS that directly engraft in recipient immunodeficient mice. T-cell progenitors were also produced in vitro from both YS and para-aortic splanchnopleura hemogenic endothelial cells, and these T-cell progenitors repopulated the thymus and differentiated into mature T-cell subsets in vivo on transplantation. Our data confirm that the YS produces T-lineage-restricted progenitors that are available to colonize the thymus and provide new insight into the YS as a definitive hematopoietic site in the mouse embryo.


Development | 2013

Fkbp1a controls ventricular myocardium trabeculation and compaction by regulating endocardial Notch1 activity

Hanying Chen; Wenjun Zhang; Xiaoxin Sun; Momoko Yoshimoto; Zhuang Chen; Wuqiang Zhu; Jijia Liu; Yadan Shen; Weidong Yong; Deqiang Li; Jin Zhang; Yang Lin; Bai-Yan Li; Nathan J. VanDusen; Paige Snider; Robert J. Schwartz; Simon J. Conway; Loren J. Field; Mervin C. Yoder; Anthony B. Firulli; Nadia Carlesso; Jeffrey A. Towbin; Weinian Shou

Trabeculation and compaction of the embryonic myocardium are morphogenetic events crucial for the formation and function of the ventricular walls. Fkbp1a (FKBP12) is a ubiquitously expressed cis-trans peptidyl-prolyl isomerase. Fkbp1a-deficient mice develop ventricular hypertrabeculation and noncompaction. To determine the physiological function of Fkbp1a in regulating the intercellular and intracellular signaling pathways involved in ventricular trabeculation and compaction, we generated a series of Fkbp1a conditional knockouts. Surprisingly, cardiomyocyte-restricted ablation of Fkbp1a did not give rise to the ventricular developmental defect, whereas endothelial cell-restricted ablation of Fkbp1a recapitulated the ventricular hypertrabeculation and noncompaction observed in Fkbp1a systemically deficient mice, suggesting an important contribution of Fkbp1a within the developing endocardia in regulating the morphogenesis of ventricular trabeculation and compaction. Further analysis demonstrated that Fkbp1a is a novel negative modulator of activated Notch1. Activated Notch1 (N1ICD) was significantly upregulated in Fkbp1a-ablated endothelial cells in vivo and in vitro. Overexpression of Fkbp1a significantly reduced the stability of N1ICD and direct inhibition of Notch signaling significantly reduced hypertrabeculation in Fkbp1a-deficient mice. Our findings suggest that Fkbp1a-mediated regulation of Notch1 plays an important role in intercellular communication between endocardium and myocardium, which is crucial in controlling the formation of the ventricular walls.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2004

Role of Bone Marrow-Derived Progenitor Cells in Cuff-Induced Vascular Injury in Mice

Yang Xu; Hidenori Arai; Xin Zhuge; Hideto Sano; Toshinori Murayama; Momoko Yoshimoto; Toshio Heike; Tatsutoshi Nakahata; Shin-Ichi Nishikawa; Toru Kita; Masayuki Yokode

Objectives—Arterial injury results in vascular remodeling associated with proliferation and migration of smooth muscle cells (SMCs) and the development of intimal hyperplasia, which is a critical component of restenosis after angioplasty of human coronary arteries and an important feature of atherosclerotic lesions. However, the origin of SMCs and other cells in the development of vascular remodeling is not yet fully understood. Methods and Results—We utilized a cuff-induced vascular injury model after transplantation of the bone marrow (BM) from green fluorescent protein (GFP)-transgenic mice. We found that macrophages were major cells recruited to the adventitia of the vascular injury lesion along with SMCs and endothelial cells (ECs). While investigating whether those cells are derived from the donor, we found that most of the macrophages were GFP-positive, and some of the SMCs and ECs were also GFP-positive. Administration of the anti–c-fms antibody resulted in a marked decrease in macrophages and a relative increase of SMCs, while administration of antibodies against the platelet-derived growth factor receptor-&bgr; caused a prominent decrease in SMCs and a relative increase in macrophages. Conclusions—The current study indicates that BM-derived cells play an important role in vascular injury, and that differentiation of macrophages and SMCs might be dependent on each other.


The FASEB Journal | 2007

Human cord blood CD34+ cells develop into hepatocytes in the livers of NOD/SCID/γcnull mice through cell fusion

Hisanori Fujino; Hidefumi Hiramatsu; Atsunori Tsuchiya; Akira Niwa; Haruyoshi Noma; Mitsutaka Shiota; Katsutsugu Umeda; Momoko Yoshimoto; Mamoru Ito; Toshio Heike; Tatsutoshi Nakahata

Several studies have shown that hepatocytes can be generated from hematopoietic stem cells, but this event is believed to be rare and to require hepatic damage. To investigate this phenomenon in human cells, we used a NOD/SCID/γcnull (NOG) mouse model that can achieve a tremendously high level of chimerism when transplanted with human hematopoietic cells. Even without hepatotoxic treatment other than irradiation, human albumin and α‐1‐antitrypsin‐positive cells were invariably detected in the livers of NOG mice after i.v. transplantation of human cord blood CD34+ cells. Human albumin was detected in the murine sera, indicating functional maturation of the human hepatocytes. Flow cytometric analysis of recipient liver cells in single‐cell suspension demonstrated that human albumin‐positive cells were also positive for both murine and human MHC and were negative for human CD45. PCR analysis of recipient livers revealed the expression of a wide variety of human hepatocyte‐ or cholangiocyte‐specific mRNAs. These results show that human CD34+ cells fuse with hepatocytes of NOG mice without liver injury, lose their hematopoietic phenotype, and begin hepatocyte‐specific gene transcription. These phenomena were not observed when CD34− cells were transplanted. Thus, our model revealed a previously unidentified pathway of human hematopoietic stem/progenitor cell differentiation.—Fujino, H., Hiramatsu, H., Tsuchiya, A., Niwa, A., Noma, H., Shiota, M., Umeda, K., Yoshimoto, M., Ito, M., Heike, T., Nakahata, T. Human cord blood CD34+ cells develop into hepatocytes in the livers of NOD/SCID/γcnull mice through cell fusion. FASEB J. 21, 3499–3510 (2007)

Collaboration


Dive into the Momoko Yoshimoto's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yan Liu

European Organisation for Research and Treatment of Cancer

View shared research outputs
Researchain Logo
Decentralizing Knowledge