Network


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

Hotspot


Dive into the research topics where Koichi Nishijo is active.

Publication


Featured researches published by Koichi Nishijo.


Cancer Cell | 2011

Evidence for an Unanticipated Relationship between Undifferentiated Pleomorphic Sarcoma and Embryonal Rhabdomyosarcoma

Brian P. Rubin; Koichi Nishijo; Hung I Harry Chen; Xiaolan Yi; David P. Schuetze; Ranadip Pal; Suresh I. Prajapati; Jinu Abraham; Benjamin R. Arenkiel; Qing Rong Chen; Sean Davis; Amanda T. McCleish; Mario R. Capecchi; Joel E. Michalek; Lee Ann Zarzabal; Javed Khan; Zhongxin Yu; David M. Parham; Frederic G. Barr; Paul S. Meltzer; Yidong Chen; Charles Keller

Embryonal rhabdomyosarcoma (eRMS) shows the most myodifferentiation among sarcomas, yet the precise cell of origin remains undefined. Using Ptch1, p53 and/or Rb1 conditional mouse models and controlling prenatal or postnatal myogenic cell of origin, we demonstrate that eRMS and undifferentiated pleomorphic sarcoma (UPS) lie in a continuum, with satellite cells predisposed to giving rise to UPS. Conversely, p53 loss in maturing myoblasts gives rise to eRMS, which have the highest myodifferentiation potential. Regardless of origin, Rb1 loss modifies tumor phenotype to mimic UPS. In human sarcomas that lack pathognomic chromosomal translocations, p53 loss of function is prevalent, whereas Shh or Rb1 alterations likely act primarily as modifiers. Thus, sarcoma phenotype is strongly influenced by cell of origin and mutational profile.


The FASEB Journal | 2009

Biomarker system for studying muscle, stem cells, and cancer in vivo

Koichi Nishijo; Tohru Hosoyama; Christopher R.R. Bjornson; Beverly S. Schaffer; Suresh I. Prajapati; Ali N. Bahadur; Mark S. Hansen; Mary C. Blandford; Amanda T. McCleish; Brian P. Rubin; Jonathan A. Epstein; Thomas A. Rando; Mario R. Capecchi; Charles Keller

Bioluminescent reporter genes are sensitive in situ tools for following disease progression in preclinical models, albeit they are subject to scattering and absorption in deep tissues. We have generated a bicistronic Cre/LoxP reporter mouse line that pairs the expression of firefly luciferase with quantifiable expression of a human placental alkaline phosphatase that is secreted into the serum (SeAP). With the use of this dual‐modality bioreporter with a novel, inducible Pax7‐ CreER line for tracking muscle satellite cells, we demonstrate the longitudinal kinetics of muscle stem cell turnover, accounting for a doubling of the signal from satellite cell and progeny every 3.93 wk in the transition from adolescence to early adulthood. We also show that this dual‐modality bioreporter can be incorporated in preclinical cancer models, whereby SeAP activity is reflective of tumor burden. Thus, this dual bioreporter permits both spatial localization and accurate quantification of biological processes in vivo even when the tissue of interest is deep within the animal.— Nishijo, K.,Hosoyama, T., Bjornson, C. R. R., Schaffer, B. S., Prajapati, S. I., Bahadur, A. N., Hansen, M. S., Blandford, M. C., McCleish, A. T., Rubin, B. P., Epstein, J. A., Rando, T. A., Capecchi, M. R., Keller, C. Biomarker system for studying muscle, stem cells, and cancer in vivo. FASEB J. 23, 2681–2690 (2009)


Oncogene | 2008

PDGFR-A is a therapeutic target in alveolar rhabdomyosarcoma

Eri Taniguchi; Koichi Nishijo; Amanda T. McCleish; Joel E. Michalek; Marcia Grayson; Anthony J. Infante; H. E. Abboud; Robin D. LeGallo; Stephen J. Qualman; Brian P. Rubin; Charles Keller

Alveolar rhabdomyosarcoma is an aggressive skeletal muscle cancer of childhood. Our initial studies of rhabdomyosarcoma gene expression for patients enrolled in a national clinical trial suggested that platelet-derived growth factor receptor A (PDGFR-A) may be a mediator of disease progression and metastasis. Using our conditional mouse tumor models that authentically recapitulate the primary mutations and metastatic progression of alveolar rhabdomyosarcomas in humans, we found by immunoblotting and immunokinase assays that PDGFR-A and its downstream effectors, mitogen-activated protein kinase and Akt, were highly activated in both primary and metastatic tumors. Inhibition of PDGFR-A by RNA interference, small molecule inhibitor or neutralizing antibody had a dramatic effect on tumor cell growth both in vitro and in vivo, although resistance evolved in one-third of tumors. These results establish proof-of-principal for PDGFR-A as a therapeutic target in alveolar rhabdomyosarcoma.


Genes & Cancer | 2010

A Postnatal Pax7+ Progenitor Gives Rise to Pituitary Adenomas

Tohru Hosoyama; Koichi Nishijo; Melinda M. Garcia; Beverly S. Schaffer; Sachiko Ohshima-Hosoyama; Suresh I. Prajapati; Michael Duff Davis; Wilmon F. Grant; Bernd W. Scheithauer; Daniel L. Marks; Brian P. Rubin; Charles Keller

Pituitary adenomas are classified into functioning and nonfunctioning (silent) tumors on the basis of hormone secretion. However, the mechanism of tumorigenesis and the cell of origin for pituitary adenoma subtypes remain to be elucidated. Employing a tamoxifen-inducible mouse model, we demonstrate that a novel postnatal Pax7(+) progenitor cell population in the pituitary gland gives rise to silent corticotroph macro-adenomas when the retinoblastoma tumor suppressor is conditionally deleted. While Pax transcriptional factors are critical for embryonic patterning as well as postnatal stem cell renewal for many organs, we have discovered that Pax7 marks a restricted cell population in the postnatal pituitary intermediate lobe. This Pax7(+) early progenitor cell population is overlapping but ontologically downstream of the Nestin(+) pituitary stem cell population, yet upstream of another newly discovered Myf6(+) late progenitor cell population. Interestingly, the Pax7(+) progenitor cell population is evolutionarily conserved in primates and humans, and Pax7 expression is maintained not only in murine tumors but also in human functioning and silent corticotropinomas. Taken together, our results strongly suggest that human silent corticotroph adenomas may in fact arise from a Pax7 lineage of the intermediate lobe, a region of the human pituitary bearing closer scientific interest as a reservoir of pituitary progenitor cells.


Molecular Cancer Therapeutics | 2011

Evasion Mechanisms to Igf1r Inhibition in Rhabdomyosarcoma

Jinu Abraham; Suresh I. Prajapati; Koichi Nishijo; Beverly S. Schaffer; Eri Taniguchi; Aoife Kilcoyne; Amanda T. McCleish; Laura D. Nelon; Francis G. Giles; Argiris Efstratiadis; Robin D. LeGallo; Brent M. Nowak; Brian P. Rubin; Suman Malempati; Charles Keller

Inhibition of the insulin-like growth factor 1 receptor (Igf1r) is an approach being taken in clinical trials to overcome the dismal outcome for metastatic alveolar rhabdomyosarcoma (ARMS), an aggressive muscle cancer of children and young adults. In our study, we address the potential mechanism(s) of Igf1r inhibitor resistance that might be anticipated for patients. Using a genetically engineered mouse model of ARMS, validated for active Igf1r signaling, we show that the prototypic Igf1r inhibitor NVP-AEW541 can inhibit cell growth and induce apoptosis in vitro in association with decreased Akt and Mapk phosphorylation. However, drug resistance in vivo is more common and is accompanied by Igf1r overexpression, Mapk reactivation, and Her2 overexpression. Her2 is found to form heterodimers with Igf1r in resistant primary tumor cell cultures, and stimulation with Igf2 leads to Her2 phosphorylation. The Her2 inhibitor lapatinib cooperates with NVP-AEW541 to reduce Igf1r phosphorylation and to inhibit cell growth even though lapatinib alone has little effect on growth. These results point to the potential therapeutic importance of simultaneous targeting of Igf1r and Her2 to abrogate resistance. Mol Cancer Ther; 10(4); 697–707. ©2011 AACR.


Developmental Biology | 2013

Myf5 expression during fetal myogenesis defines the developmental progenitors of adult satellite cells

Stefano Biressi; Christopher R.R. Bjornson; Poppy M.M. Carlig; Koichi Nishijo; Charles Keller; Thomas A. Rando

Myf5 is a member of the muscle-specific determination genes and plays a critical role in skeletal muscle development. Whereas the expression of Myf5 during embryonic and fetal myogenesis has been extensively studied, its expression in progenitors that will ultimately give rise to adult satellite cells, the stem cells responsible for muscle repair, is still largely unexplored. To investigate this aspect, we have generated a mouse strain carrying a CreER coding sequence in the Myf5 locus. In this strain, Tamoxifen-inducible Cre activity parallels endogenous Myf5 expression. Combining Myf5(CreER) and Cre reporter alleles, we were able to evaluate the contribution of cells expressing Myf5 at distinct developmental stages to the pool of satellite cells in adult hindlimb muscles. Although it was possible to trace back the origin of some rare satellite cells to a subpopulation of Myf5(+ve) progenitors in the limb buds at the late embryonic stage (∼E12), a significant number of satellite cells arise from cells which expressed Myf5 for the first time at the fetal stage (∼E15). These studies provide direct evidence that adult satellite cells derive from progenitors that first express the myogenic determination gene Myf5 during fetal stages of myogenesis.


Clinical Cancer Research | 2011

IL-4R drives dedifferentiation, mitogenesis, and metastasis in rhabdomyosarcoma.

Tohru Hosoyama; Mohammed Imran Aslam; Jinu Abraham; Suresh I. Prajapati; Koichi Nishijo; Joel E. Michalek; Lee Ann Zarzabal; Laura D. Nelon; Denis C. Guttridge; Brian P. Rubin; Charles Keller

Purpose: Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in childhood. The alveolar subtype of rhabdomyosarcoma (ARMS) is a paradigm for refractory and incurable solid tumors because more than half of the children at diagnosis have either regional lymph node or distant metastases. These studies follow our previous observation that Interleukin-4 receptor α (IL-4Rα) is upregulated in both human and murine ARMS, and that the IL-4R signaling pathway may be a target for abrogating tumor progression. Experimental Design: By in vitro biochemical and cell biology studies as well as preclinical studies using a genetically engineered mouse model, we evaluated the role of IL-4 and IL-13 in IL-4R–mediated mitogenesis, myodifferentiation, and tumor progression. Results: IL-4 and IL-13 ligands accelerated tumor cell growth and activated STAT6, Akt, or MAPK signaling pathways in the human RMS cell lines, RD and Rh30, as well as in mouse primary ARMS cell cultures. IL-4 and IL-13 treatment also decreased protein expression of myogenic differentiation factors MyoD and Myogenin, indicating a loss of muscle differentiation. Using a genetically engineered mouse model of ARMS, we have shown that inhibition of IL-4R signaling pathway with a neutralizing antibody has a profound effect on the frequency of lymph node and pulmonary metastases, resulting in significant survival extension in vivo. Conclusions: Our results indicate that an IL-4R-dependent signaling pathway regulates tumor cell progression in RMS, and inhibition of this pathway could be a promising adjuvant therapeutic approach. Clin Cancer Res; 17(9); 2757–66. ©2011 AACR.


Genes & Development | 2014

Lineage of origin in rhabdomyosarcoma informs pharmacological response

Jinu Abraham; Yaiza Núñez-Álvarez; Simone Hettmer; Elvira Carrió; Hung I Harry Chen; Koichi Nishijo; Elaine T. Huang; Suresh I. Prajapati; Robert L. Walker; Sean Davis; Jennifer Rebeles; Hunter Wiebush; Amanda T. McCleish; Sheila T. Hampton; Christopher R.R. Bjornson; Andrew S. Brack; Amy J. Wagers; Thomas A. Rando; Mario R. Capecchi; Frank C. Marini; Benjamin Ehler; Lee Ann Zarzabal; Martin Goros; Joel E. Michalek; Paul S. Meltzer; David M. Langenau; Robin D. LeGallo; Atiya Mansoor; Yidong Chen; Mònica Suelves

Lineage or cell of origin of cancers is often unknown and thus is not a consideration in therapeutic approaches. Alveolar rhabdomyosarcoma (aRMS) is an aggressive childhood cancer for which the cell of origin remains debated. We used conditional genetic mouse models of aRMS to activate the pathognomonic Pax3:Foxo1 fusion oncogene and inactivate p53 in several stages of prenatal and postnatal muscle development. We reveal that lineage of origin significantly influences tumor histomorphology and sensitivity to targeted therapeutics. Furthermore, we uncovered differential transcriptional regulation of the Pax3:Foxo1 locus by tumor lineage of origin, which led us to identify the histone deacetylase inhibitor entinostat as a pharmacological agent for the potential conversion of Pax3:Foxo1-positive aRMS to a state akin to fusion-negative RMS through direct transcriptional suppression of Pax3:Foxo1.


Journal of Biological Chemistry | 2011

Rb1 Gene Inactivation Expands Satellite Cell and Postnatal Myoblast Pools

Tohru Hosoyama; Koichi Nishijo; Suresh I. Prajapati; Guangheng Li; Charles Keller

Satellite cells are well known as a postnatal skeletal muscle stem cell reservoir that under injury conditions participate in repair. However, mechanisms controlling satellite cell quiescence and activation are the topic of ongoing inquiry by many laboratories. In this study, we investigated whether loss of the cell cycle regulatory factor, pRb, is associated with the re-entry of quiescent satellite cells into replication and subsequent stem cell expansion. By ablation of Rb1 using a Pax7CreER,Rb1 conditional mouse line, satellite cell number was increased 5-fold over 6 months. Furthermore, myoblasts originating from satellite cells lacking Rb1 were also increased 3-fold over 6 months, while terminal differentiation was greatly diminished. Similarly, Pax7CreER,Rb1 mice exhibited muscle fiber hypotrophy in vivo under steady state conditions as well as a delay of muscle regeneration following cardiotoxin-mediated injury. These results suggest that cell cycle re-entry of quiescent satellite cells is accelerated by lack of Rb1, resulting in the expansion of both satellite cells and their progeny in adolescent muscle. Conversely, that sustained Rb1 loss in the satellite cell lineage causes a deficit of muscle fiber formation. However, we also show that pharmacological inhibition of protein phosphatase 1 activity, which will result in pRb inactivation accelerates satellite cell activation and/or expansion in a transient manner. Together, our results raise the possibility that reversible pRb inactivation in satellite cells and inhibition of protein phosphorylation may provide a new therapeutic tool for muscle atrophy by short term expansion of the muscle stem cells and myoblast pool.


Skeletal Muscle | 2013

Rb1 loss modifies but does not initiate alveolar rhabdomyosarcoma

Ken Kikuchi; Eri Taniguchi; Hung-I Harry Chen; Matthew N. Svalina; Jinu Abraham; Elaine T. Huang; Koichi Nishijo; Sean Davis; Christopher Louden; Lee Ann Zarzabal; Olivia Recht; Ayeza Bajwa; Noah Berlow; Mònica Suelves; Sherrie L. Perkins; Paul S. Meltzer; Atiya Mansoor; Joel E. Michalek; Yidong Chen; Brian P. Rubin; Charles Keller

BackgroundAlveolar rhabdomyosarcoma (aRMS) is a myogenic childhood sarcoma frequently associated with a translocation-mediated fusion gene, Pax3:Foxo1a.MethodsWe investigated the complementary role of Rb1 loss in aRMS tumor initiation and progression using conditional mouse models.ResultsRb1 loss was not a necessary and sufficient mutational event for rhabdomyosarcomagenesis, nor a strong cooperative initiating mutation. Instead, Rb1 loss was a modifier of progression and increased anaplasia and pleomorphism. Whereas Pax3:Foxo1a expression was unaltered, biomarkers of aRMS versus embryonal rhabdomyosarcoma were both increased, questioning whether these diagnostic markers are reliable in the context of Rb1 loss. Genome-wide gene expression in Pax3:Foxo1a,Rb1 tumors more closely approximated aRMS than embryonal rhabdomyosarcoma. Intrinsic loss of pRb function in aRMS was evidenced by insensitivity to a Cdk4/6 inhibitor regardless of whether Rb1 was intact or null. This loss of function could be attributed to low baseline Rb1, pRb and phospho-pRb expression in aRMS tumors for which the Rb1 locus was intact. Pax3:Foxo1a RNA interference did not increase pRb or improve Cdk inhibitor sensitivity. Human aRMS shared the feature of low and/or heterogeneous tumor cell pRb expression.ConclusionsRb1 loss from an already low pRb baseline is a significant disease modifier, raising the possibility that some cases of pleomorphic rhabdomyosarcoma may in fact be Pax3:Foxo1a-expressing aRMS with Rb1 or pRb loss of function.

Collaboration


Dive into the Koichi Nishijo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Suresh I. Prajapati

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Amanda T. McCleish

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Joel E. Michalek

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lee Ann Zarzabal

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tohru Hosoyama

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Beverly S. Schaffer

University of Texas Health Science Center at San Antonio

View shared research outputs
Researchain Logo
Decentralizing Knowledge