Network


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

Hotspot


Dive into the research topics where Marc T. Seligson is active.

Publication


Featured researches published by Marc T. Seligson.


Cell | 2013

Lin28 enhances tissue repair by reprogramming cellular metabolism

Ng Shyh-Chang; Hao Zhu; T. Yvanka de Soysa; Gen Shinoda; Marc T. Seligson; Kaloyan M. Tsanov; Liem H. Nguyen; John M. Asara; Lewis C. Cantley; George Q. Daley

Regeneration capacity declines with age, but why juvenile organisms show enhanced tissue repair remains unexplained. Lin28a, a highly conserved RNA-binding protein expressed during embryogenesis, plays roles in development, pluripotency, and metabolism. To determine whether Lin28a might influence tissue repair in adults, we engineered the reactivation of Lin28a expression in several models of tissue injury. Lin28a reactivation improved hair regrowth by promoting anagen in hair follicles and accelerated regrowth of cartilage, bone, and mesenchyme after ear and digit injuries. Lin28a inhibits let-7 microRNA biogenesis; however, let-7 repression was necessary but insufficient to enhance repair. Lin28a bound to and enhanced the translation of mRNAs for several metabolic enzymes, thereby increasing glycolysis and oxidative phosphorylation (OxPhos). Lin28a-mediated enhancement of tissue repair was negated by OxPhos inhibition, whereas a pharmacologically induced increase in OxPhos enhanced repair. Thus, Lin28a enhances tissue repair in some adult tissues by reprogramming cellular bioenergetics. PAPERCLIP:


Cancer Cell | 2014

Lin28b Is Sufficient to Drive Liver Cancer and Necessary for Its Maintenance in Murine Models

Liem H. Nguyen; Daisy A. Robinton; Marc T. Seligson; Linwei Wu; Lin Li; Dinesh Rakheja; Sarah A. Comerford; Saleh Ramezani; Xiankai Sun; Monisha S. Parikh; Erin H. Yang; John T. Powers; Gen Shinoda; Samar P. Shah; Robert E. Hammer; George Q. Daley; Hao Zhu

Lin28a/b are RNA-binding proteins that influence stem cell maintenance, metabolism, and oncogenesis. Poorly differentiated, aggressive cancers often overexpress Lin28, but its role in tumor initiation or maintenance has not been definitively addressed. We report that LIN28B overexpression is sufficient to initiate hepatoblastoma and hepatocellular carcinoma in murine models. We also detected Lin28b overexpression in MYC-driven hepatoblastomas, and liver-specific deletion of Lin28a/b reduced tumor burden, extended latency, and prolonged survival. Both intravenous siRNA against Lin28b and conditional Lin28b deletion reduced tumor burden and prolonged survival. Igf2bp proteins are upregulated, and Igf2bp3 is required in the context of LIN28B overexpression to promote growth. Therefore, multiple murine models demonstrate that Lin28b is both sufficient to initiate liver cancer and necessary for its maintenance.


Stem Cells | 2013

Fetal Deficiency of Lin28 Programs Life-Long Aberrations in Growth and Glucose Metabolism†

Gen Shinoda; Ng Shyh-Chang; T. Yvanka de Soysa; Hao Zhu; Marc T. Seligson; Samar P. Shah; Nora Abo-Sido; Akiko Yabuuchi; John P. Hagan; Richard I. Gregory; John M. Asara; Lewis C. Cantley; Eric G. Moss; George Q. Daley

LIN28A/B are RNA binding proteins implicated by genetic association studies in human growth and glucose metabolism. Mice with ectopic over‐expression of Lin28a have shown related phenotypes. Here, we describe the first comprehensive analysis of the physiologic consequences of Lin28a and Lin28b deficiency in knockout (KO) mice. Lin28a/b‐deficiency led to dwarfism starting at different ages, and compound gene deletions showed a cumulative dosage effect on organismal growth. Conditional gene deletion at specific developmental stages revealed that fetal but neither neonatal nor adult deficiency resulted in growth defects and aberrations in glucose metabolism. Tissue‐specific KO mice implicated skeletal muscle‐deficiency in the abnormal programming of adult growth and metabolism. The effects of Lin28b KO could be rescued by Tsc1 haplo‐insufficiency in skeletal muscles. Our data implicate fetal expression of Lin28a/b in the regulation of life‐long effects on metabolism and growth, and demonstrate that fetal Lin28b acts at least in part via mTORC1 signaling. STEM Cells 2013;31:1563–1573


Nature | 2016

Multiple mechanisms disrupt the let-7 microRNA family in neuroblastoma

John T. Powers; Kaloyan M. Tsanov; Daniel S. Pearson; Frederik Roels; Catherine Spina; Richard H. Ebright; Marc T. Seligson; Yvanka de Soysa; Patrick Cahan; Jessica Theißen; Ho Chou Tu; Areum Han; Kyle C. Kurek; Grace S. LaPier; Jihan K. Osborne; Samantha J. Ross; Marcella Cesana; James J. Collins; Frank Berthold; George Q. Daley

Poor prognosis in neuroblastoma is associated with genetic amplification of MYCN. MYCN is itself a target of let-7, a tumour suppressor family of microRNAs implicated in numerous cancers. LIN28B, an inhibitor of let-7 biogenesis, is overexpressed in neuroblastoma and has been reported to regulate MYCN. Here we show, however, that LIN28B is dispensable in MYCN-amplified neuroblastoma cell lines, despite de-repression of let-7. We further demonstrate that MYCN messenger RNA levels in amplified disease are exceptionally high and sufficient to sponge let-7, which reconciles the dispensability of LIN28B. We found that genetic loss of let-7 is common in neuroblastoma, inversely associated with MYCN amplification, and independently associated with poor outcomes, providing a rationale for chromosomal loss patterns in neuroblastoma. We propose that let-7 disruption by LIN28B, MYCN sponging, or genetic loss is a unifying mechanism of neuroblastoma development with broad implications for cancer pathogenesis.


Stem Cells | 2013

Lin28a Regulates Germ Cell Pool Size and Fertility

Gen Shinoda; T. Yvanka de Soysa; Marc T. Seligson; Akiko Yabuuchi; Yuko Fujiwara; Pei Yi Huang; John P. Hagan; Richard I. Gregory; Eric G. Moss; George Q. Daley

Overexpression of LIN28A is associated with human germ cell tumors and promotes primordial germ cell (PGC) development from embryonic stem cells in vitro and in chimeric mice. Knockdown of Lin28a inhibits PGC development in vitro, but how constitutional Lin28a deficiency affects the mammalian reproductive system in vivo remains unknown. Here, we generated Lin28a knockout (KO) mice and found that Lin28a deficiency compromises the size of the germ cell pool in both males and females by affecting PGC proliferation during embryogenesis. Interestingly however, in Lin28a KO males, the germ cell pool partially recovers during postnatal expansion, while fertility remains impaired in both males and females mated to wild‐type mice. Embryonic overexpression of let‐7, a microRNA negatively regulated by Lin28a, reduces the germ cell pool, corroborating the role of the Lin28a/let‐7 axis in regulating the germ lineage. STEM CELLS 2013;31:1001–1009


Nature Cell Biology | 2017

LIN28 phosphorylation by MAPK/ERK couples signalling to the post-transcriptional control of pluripotency

Kaloyan M. Tsanov; Daniel S. Pearson; Zhaoting Wu; Areum Han; Robinson Triboulet; Marc T. Seligson; John T. Powers; Jihan K. Osborne; Susan Kane; Steven P. Gygi; Richard I. Gregory; George Q. Daley

Signalling and post-transcriptional gene control are both critical for the regulation of pluripotency, yet how they are integrated to influence cell identity remains poorly understood. LIN28 (also known as LIN28A), a highly conserved RNA-binding protein, has emerged as a central post-transcriptional regulator of cell fate through blockade of let-7 microRNA biogenesis and direct modulation of mRNA translation. Here we show that LIN28 is phosphorylated by MAPK/ERK in pluripotent stem cells, which increases its levels via post-translational stabilization. LIN28 phosphorylation had little impact on let-7 but enhanced the effect of LIN28 on its direct mRNA targets, revealing a mechanism that uncouples LIN28’s let-7-dependent and -independent activities. We have linked this mechanism to the induction of pluripotency by somatic cell reprogramming and the transition from naive to primed pluripotency. Collectively, our findings indicate that MAPK/ERK directly impacts LIN28, defining an axis that connects signalling, post-transcriptional gene control, and cell fate regulation.


eLife | 2015

Precise let-7 expression levels balance organ regeneration against tumor suppression

Linwei Wu; Liem H. Nguyen; Kejin Zhou; T. Yvanka de Soysa; Lin Li; Jason B. Miller; Jianmin Tian; Joseph Locker; Shuyuan Zhang; Gen Shinoda; Marc T. Seligson; Lauren R. Zeitels; Asha Acharya; Sam C. Wang; Joshua T. Mendell; Xiaoshun He; Jinsuke Nishino; Sean J. Morrison; Daniel J. Siegwart; George Q. Daley; Ng Shyh-Chang; Hao Zhu

The in vivo roles for even the most intensely studied microRNAs remain poorly defined. Here, analysis of mouse models revealed that let-7, a large and ancient microRNA family, performs tumor suppressive roles at the expense of regeneration. Too little or too much let-7 resulted in compromised protection against cancer or tissue damage, respectively. Modest let-7 overexpression abrogated MYC-driven liver cancer by antagonizing multiple let-7 sensitive oncogenes. However, the same level of overexpression blocked liver regeneration, while let-7 deletion enhanced it, demonstrating that distinct let-7 levels can mediate desirable phenotypes. let-7 dependent regeneration phenotypes resulted from influences on the insulin-PI3K-mTOR pathway. We found that chronic high-dose let-7 overexpression caused liver damage and degeneration, paradoxically leading to tumorigenesis. These dose-dependent roles for let-7 in tissue repair and tumorigenesis rationalize the tight regulation of this microRNA in development, and have important implications for let-7 based therapeutics. DOI: http://dx.doi.org/10.7554/eLife.09431.001


Cancer Research | 2016

Abstract LB-165: Multiple mechanisms disrupt let-7 miRNA biogenesis and function in neuroblastoma

John T. Powers; Kaloyan Tsanov; Frederik Roels; Catherine Spina; Richard H. Ebright; Marc T. Seligson; Yvanka de Soysa; Patrick Cahan; Daniel S. Pearson; Jessica Theißen; Ho-Chou Tu; Grace LaPier; Jihan Osborne; Samantha Ross; James J. Collins; Frank Berthold; George Q. Daley

The let-7 microRNA family are known tumor suppressors often deregulated in cancer, yet the underlying mechanisms of let-7 disruption remain poorly understood. Neuroblastoma, a neural crest derived tumor, is defined in part by poor prognosis associated with genetic amplification of MYCN, itself a let-7 target. The let-7 biogenesis inhibitor LIN28B has recently been implicated as a critical regulator of MYCN, but through CRISPR-mediated gene disruption we show that LIN28B is dispensable for both MYCN protein expression and growth of MYCN-amplified neuroblastoma cell lines despite robust de-repression of let-7, prompting us to explore additional mechanisms for let-7 disruption. Consequently, we have found a novel non-coding role for amplified MYCN mRNA as a potent let-7 sponge that through exceptionally high expression defines a sub-class of self-sponging amplified-competing-endogenous-RNA (aceRNA) and reconciling the dispensability of LIN28B in neuroblastoma cell lines. Furthermore, by analyzing a large cohort of tumor samples from patients, we observe frequent genomic loss of let-7 that inversely associates with MYCN-amplification, providing a functional explanation for the known MYCN-amplification-independent pattern of chromosome 3p and 11q loss, which harbor let-7g and let-7a2, respectively. We thus propose a model whereby let-7 disruption by genetic loss, LIN28B expression, or aceRNA sponging is a unifying mechanism of neuroblastoma pathogenesis. Indeed, our data show that the majority of neuroblastomas have at least one let-7 disruption event and that genetic loss in non-MYCN-amplified tumors marks decreased survival, further underscoring its importance. The inverse selective relationship between allelic loss and sponging of let-7 from highly expressed or amplified oncogenes may have broad implications for oncogenesis. Citation Format: John T. Powers, Kaloyan Tsanov, Frederik Roels, Catherine Spina, Richard Ebright, Marc Seligson, Yvanka de Soysa, Patrick Cahan, Daniel Pearson, Jessica Theisen, Ho-Chou Tu, Grace LaPier, Jihan Osborne, Samantha Ross, James Collins, Frank Berthold, George Daley. Multiple mechanisms disrupt let-7 miRNA biogenesis and function in neuroblastoma. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr LB-165.


Cancer Research | 2015

Abstract LB-290: Multiple distinct mechanisms disrupt let-7 miRNA biogenesis and function in neuroblastoma

John T. Powers; Kaloyan Tsanov; Frederik Roles; Richard Ebright; Marc T. Seligson; Yvanka de Soysa; Patrick Cahan; Jessica Theissen; Grace LaPier; Dan S. Pearson; Frank Berthold; George Q. Daley

The let-7 microRNA family are known tumor suppressors often deregulated in cancer, yet the underlying mechanisms of let-7 disruption remain poorly understood. Neuroblastoma, a neural crest derived tumor, is defined in part by poor prognosis associated with genetic amplification of MYCN, itself a let-7 target. The let-7 biogenesis inhibitor LIN28B has recently been implicated as a critical regulator of MYCN, but we have employed siRNA and CRISPR-mediated gene disruption to show that LIN28B is dispensable for both MYCN protein expression and growth of MYCN-amplified neuroblastoma cell lines despite robust de-repression of let-7, which prompted us to explore additional mechanisms for let-7 disruption. Consequently, we have found that amplified MYCN mRNA is a potent let-7 sponge that through exceptionally high expression defines a sub-class of self-sponging amplified-competing-endogenous-RNA (aceRNA), which reconciles the dispensability of LIN28B in NB cell lines. In addition, by analyzing a large cohort of tumor samples from patients, we observe frequent genomic loss of let-7 that inversely associates with MYCN-amplification, providing a functional explanation for the known MYCN-amplification-independent pattern of chromosome 3p and 11q loss, which harbor let-7g and let-7a2, respectively. We thus propose a model whereby let-7 disruption by genetic loss, LIN28B expression, or aceRNA sponging is a unifying mechanism of neuroblastoma pathogenesis. Indeed, our data show that the majority of neuroblastomas have at least one let-7 disruption event and that genetic loss in non-MYCN-amplified tumors marks decreased survival, further underscoring its importance. The inverse selective relationship between allelic loss and sponging of let-7 from highly expressed or amplified oncogenes may have broad implications for oncogenesis. Note: This abstract was not presented at the meeting. Citation Format: John T. Powers, Kaloyan M. Tsanov, Frederik Roles, Richard Ebright, Marc Seligson, Yvanka de Soysa, Patrick Cahan, Jessica Theissen, Grace S. LaPier, Dan S. Pearson, Frank Berthold, George Q. Daley. Multiple distinct mechanisms disrupt let-7 miRNA biogenesis and function in neuroblastoma. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr LB-290. doi:10.1158/1538-7445.AM2015-LB-290


Cell Stem Cell | 2016

LIN28 Regulates Stem Cell Metabolism and Conversion to Primed Pluripotency

Jin Zhang; Sutheera Ratanasirintrawoot; Sriram Chandrasekaran; Zhaoting Wu; Scott B. Ficarro; Chunxiao Yu; Christian A. Ross; Davide Cacchiarelli; Qing Xia; Marc T. Seligson; Gen Shinoda; Wen Xie; Patrick Cahan; Longfei Wang; Shyh Chang Ng; Supisara Tintara; Cole Trapnell; Tamer T. Onder; Yuin-Han Loh; Tarjei S. Mikkelsen; Piotr Sliz; Michael A. Teitell; John M. Asara; Jarrod A. Marto; Hu Li; James J. Collins; George Q. Daley

Collaboration


Dive into the Marc T. Seligson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John T. Powers

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Hao Zhu

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James J. Collins

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

John M. Asara

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge