Network


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

Hotspot


Dive into the research topics where Zhizhong Li is active.

Publication


Featured researches published by Zhizhong Li.


Developmental Cell | 2015

A long non-coding RNA, LncMyoD, regulates skeletal muscle differentiation by blocking IMP2-mediated mRNA translation.

Chenguang Gong; Zhizhong Li; Krishnan Ramanujan; Ieuan Clay; Yunyu Zhang; Sophie Lemire-Brachat; David J. Glass

Increasing evidence suggests that long non-coding RNAs (LncRNAs) represent a new class of regulators of stem cells. However, the roles of LncRNAs in stem cell maintenance and myogenesis remain largely unexamined. For this study, hundreds of intergenic LncRNAs were identified that are expressed in myoblasts and regulated during differentiation. One of these LncRNAs, termed LncMyoD, is encoded next to the Myod gene and is directly activated by MyoD during myoblast differentiation. Knockdown of LncMyoD strongly inhibits terminal muscle differentiation, largely due to a failure to exit the cell cycle. LncMyoD directly binds to IGF2-mRNA-binding protein 2 (IMP2) and negatively regulates IMP2-mediated translation of proliferation genes such as N-Ras and c-Myc. While the RNA sequence of LncMyoD is not well conserved between human and mouse, its locus, gene structure, and function are preserved. The MyoD-LncMyoD-IMP2 pathway elucidates a mechanism as to how MyoD blocks proliferation to create a permissive state for differentiation.


Developmental Cell | 2012

An HMGA2-IGF2BP2 Axis Regulates Myoblast Proliferation and Myogenesis

Zhizhong Li; Jason A. Gilbert; Yunyu Zhang; Minsi Zhang; Qiong Qiu; Krishnan Ramanujan; Tea Shavlakadze; John K. Eash; Annarita Scaramozza; Matthew M. Goddeeris; David G. Kirsch; Kevin P. Campbell; Andrew S. Brack; David J. Glass

A group of genes that are highly and specifically expressed in proliferating skeletal myoblasts during myogenesis was identified. Expression of one of these genes, Hmga2, increases coincident with satellite cell activation, and later its expression significantly declines correlating with fusion of myoblasts into myotubes. Hmga2 knockout mice exhibit impaired muscle development and reduced myoblast proliferation, while overexpression of HMGA2 promotes myoblast growth. This perturbation in proliferation can be explained by the finding that HMGA2 directly regulates the RNA-binding protein IGF2BP2. Add-back of IGF2BP2 rescues the phenotype. IGF2BP2 in turn binds to and controls the translation of a set of mRNAs, including c-myc, Sp1, and Igf1r. These data demonstrate that the HMGA2-IGF2BP2 axis functions as a key regulator of satellite cell activation and therefore skeletal muscle development.


Journal of Clinical Investigation | 2014

MicroRNA-182 drives metastasis of primary sarcomas by targeting multiple genes

Mohit Sachdeva; Jeffrey K. Mito; Chang-Lung Lee; Minsi Zhang; Zhizhong Li; Rebecca D. Dodd; David Cason; Lixia Luo; Yan Ma; David Van Mater; Rebecca A. Gladdy; Dina Lev; Diana M. Cardona; David G. Kirsch

Metastasis causes most cancer deaths, but is incompletely understood. MicroRNAs can regulate metastasis, but it is not known whether a single miRNA can regulate metastasis in primary cancer models in vivo. We compared the expression of miRNAs in metastatic and nonmetastatic primary mouse sarcomas and found that microRNA-182 (miR-182) was markedly overexpressed in some tumors that metastasized to the lungs. By utilizing genetically engineered mice with either deletion of or overexpression of miR-182 in primary sarcomas, we discovered that deletion of miR-182 substantially decreased, while overexpression of miR-182 considerably increased, the rate of lung metastasis after amputation of the tumor-bearing limb. Additionally, deletion of miR-182 decreased circulating tumor cells (CTCs), while overexpression of miR-182 increased CTCs, suggesting that miR-182 regulates intravasation of cancer cells into the circulation. We identified 4 miR-182 targets that inhibit either the migration of tumor cells or the degradation of the extracellular matrix. Notably, restoration of any of these targets in isolation did not alter the metastatic potential of sarcoma cells injected orthotopically, but the simultaneous restoration of all 4 targets together substantially decreased the number of metastases. These results demonstrate that a single miRNA can regulate metastasis of primary tumors in vivo by coordinated regulation of multiple genes.


Cell Reports | 2013

Distinct and Overlapping Sarcoma Subtypes Initiated from Muscle Stem and Progenitor Cells

Jordan M. Blum; Leonor Añó; Zhizhong Li; David Van Mater; Brian D. Bennett; Mohit Sachdeva; Irina Lagutina; Minsi Zhang; Jeffrey K. Mito; Leslie G. Dodd; Diana M. Cardona; Rebecca D. Dodd; Nerissa Williams; Yan Ma; Christoph Lepper; Corinne M. Linardic; Sayan Mukherjee; Gerard Grosveld; Chen-Ming Fan; David G. Kirsch

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children, whereas undifferentiated pleomorphic sarcoma (UPS) is one of the most common soft tissue sarcomas diagnosed in adults. To investigate the myogenic cell(s) of origin of these sarcomas, we used Pax7-CreER and MyoD-CreER mice to transform Pax7(+) and MyoD(+) myogenic progenitors by expressing oncogenic Kras(G12D) and deleting Trp53 in vivo. Pax7-CreER mice developed RMS and UPS, whereas MyoD-CreER mice developed UPS. Using gene set enrichment analysis, RMS and UPS each clustered specifically within their human counterparts. These results suggest that RMS and UPS have distinct and overlapping cells of origin within the muscle lineage. Taking them together, we have established mouse models of soft tissue sarcoma from muscle stem and progenitor cells.


Cancer Research | 2013

Oncogenic NRAS, required for pathogenesis of embryonic rhabdomyosarcoma, relies upon the HMGA2-IGF2BP2 pathway

Zhizhong Li; Yunyu Zhang; Krishnan Ramanujan; Yan Ma; David G. Kirsch; David J. Glass

Embryonic rhabdomyosarcoma (ERMS) is the most common soft-tissue tumor in children. Here, we report the identification of the minor groove DNA-binding factor high mobility group AT-hook 2 (HMGA2) as a driver of ERMS development. HMGA2 was highly expressed in normal myoblasts and ERMS cells, where its expression was essential to maintain cell proliferation, survival in vitro, and tumor outgrowth in vivo. Mechanistic investigations revealed that upregulation of the insulin-like growth factor (IGF) mRNA-binding protein IGF2BP2 was critical for HMGA2 action. In particular, IGF2BP2 was essential for mRNA and protein stability of NRAS, a frequently mutated gene in ERMS. shRNA-mediated attenuation of NRAS or pharmacologic inhibition of the MAP-ERK kinase (MEK)/extracellular signal-regulated kinase (ERK) effector pathway showed that NRAS and NRAS-mediated signaling was required for tumor maintenance. Taken together, these findings implicate the HMGA2-IGFBP2-NRAS signaling pathway as a critical oncogenic driver in ERMS.


Radiation Research | 2015

HIF-1 Alpha Regulates the Response of Primary Sarcomas to Radiation Therapy through a Cell Autonomous Mechanism

Minsi Zhang; Qiong Qiu; Zhizhong Li; Mohit Sachdeva; Hooney Min; Diana M. Cardona; Thomas F. DeLaney; Tracy Han; Yan Ma; Lixia Luo; Olga Ilkayeva; Ki Lui; Amanda G. Nichols; Christopher B. Newgard; Michael B. Kastan; Jeffrey C. Rathmell; Mark W. Dewhirst; David G. Kirsch

Hypoxia is a major cause of radiation resistance, which may predispose to local recurrence after radiation therapy. While hypoxia increases tumor cell survival after radiation exposure because there is less oxygen to oxidize damaged DNA, it remains unclear whether signaling pathways triggered by hypoxia contribute to radiation resistance. For example, intratumoral hypoxia can increase hypoxia inducible factor 1 alpha (HIF-1α), which may regulate pathways that contribute to radiation sensitization or radiation resistance. To clarify the role of HIF-1α in regulating tumor response to radiation, we generated a novel genetically engineered mouse model of soft tissue sarcoma with an intact or deleted HIF-1α. Deletion of HIF-1α sensitized primary sarcomas to radiation exposure in vivo. Moreover, cell lines derived from primary sarcomas lacking HIF-1α, or in which HIF-1α was knocked down, had decreased clonogenic survival in vitro, demonstrating that HIF-1α can promote radiation resistance in a cell autonomous manner. In HIF-1α-intact and -deleted sarcoma cells, radiation-induced reactive oxygen species, DNA damage repair and activation of autophagy were similar. However, sarcoma cells lacking HIF-1α had impaired mitochondrial biogenesis and metabolic response after irradiation, which might contribute to radiation resistance. These results show that HIF-1α promotes radiation resistance in a cell autonomous manner.


Methods of Molecular Biology | 2015

Methods to Generate Genetically Engineered Mouse Models of Soft Tissue Sarcoma

Rebecca D. Dodd; Leonor Añó; Jordan M. Blum; Zhizhong Li; David Van Mater; David G. Kirsch

We discuss the generation of primary soft tissue sarcomas in mice using the Cre-loxP system to activate conditional mutations in oncogenic Kras and the tumor suppressor p53 (LSL-Kras(G12D/+); p53(flox/flox)). Sarcomas can be generated either by adenoviral delivery of Cre recombinase, activation of transgenic Cre recombinase with tamoxifen, or through transplantation of isolated satellite cells with Cre activation in vitro. Various applications of these models are discussed, including anticancer therapies, metastasis, in vivo imaging, and genetic requirements for tumorigenesis.


Cancer Research | 2014

Abstract A58: HMGA2 controls muscle stem cell activation and rhabdomyosarcoma progression

Zhizhong Li

Rhabdomyosarcoma (RMS) is the most common soft-tissue sarcoma in children and it shares significant features with proliferating muscle progenitor (myoblast). In an effort to profile mechanistic changes that might control myogenesis and/or induce RMS, a group of genes were identified that are highly and specifically expressed in myoblasts and RMS, but not quiescent muscle stem cells or differentiated myofibers. One of these genes, HMGA2, maintains myoblast and RMS cellular proliferation and survival. Hmga2 knockout mice exhibit impaired muscle development and reduced myoblast proliferation. Similarly, targeting HMGA2 expression by shRNAs inhibited RMS proliferation and increased apoptosis. This perturbation in proliferation can be explained by the finding that HMGA2 directly regulates RNA-binding protein IGF2BP2 in both myoblast and RMS. IGF2BP2 in turn binds to and controls the translation of a set of mRNAs, including NRas, which is frequently mutated in RMS and critical for tumor progression. These data demonstrate that HMGA2-IGF2BP2 axis functions as a conserved regulator of myoblast and RMS growth and may provide a novel target for future drug development to treat this childhood cancer. References: Li Z, et.al. Oncogenic NRAS, required for pathogenesis of embryonic rhabdomyosarcoma, relies upon the HMGA2-IGF2BP2 pathway. Cancer Res. 2013 May 15;73(10):3041-50. Li Z, et,al. An HMGA2-IGF2BP2 axis regulates myoblast proliferation and myogenesis. Dev Cell. 2012 Dec 11;23(6):1176-88. Note: This abstract was not presented at the conference. Citation Format: Zhizhong Li. HMGA2 controls muscle stem cell activation and rhabdomyosarcoma progression. [abstract]. In: Proceedings of the AACR Special Conference on Pediatric Cancer at the Crossroads: Translating Discovery into Improved Outcomes; Nov 3-6, 2013; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2013;74(20 Suppl):Abstract nr A58.


Cold Spring Harbor Perspectives in Medicine | 2014

Rhabdomyosarcoma: Current Challenges and Their Implications for Developing Therapies

Simone Hettmer; Zhizhong Li; Andrew N. Billin; Frederic G. Barr; D.D.W. Cornelison; Alan R. Ehrlich; Denis C. Guttridge; Andrea Hayes-Jordan; Lee J. Helman; Peter J. Houghton; Javed Khan; David M. Langenau; Corinne M. Linardic; Ranadip Pal; Terence A. Partridge; Grace K. Pavlath; Rossella Rota; Beat W. Schäfer; Janet Shipley; Bruce Stillman; Leonard H. Wexler; Amy J. Wagers; Charles Keller


Archive | 2014

Novel lncrna polynucleotides

David J. Glass; Chenguang Gong; Zhizhong Li

Collaboration


Dive into the Zhizhong Li'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
Researchain Logo
Decentralizing Knowledge