Network


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

Hotspot


Dive into the research topics where Zhaomin Li is active.

Publication


Featured researches published by Zhaomin Li.


Blood | 2014

Loss of Asxl1 leads to myelodysplastic syndrome-like disease in mice.

Jiapeng Wang; Zhaomin Li; Yongzheng He; Feng Pan; Shi Chen; Steven D. Rhodes; Lihn Nguyen; Jin Yuan; Li Jiang; Xianlin Yang; Ophelia Weeks; Ziyue Liu; Jiehao Zhou; Hongyu Ni; Chen-Leng Cai; Mingjiang Xu; Feng Chun Yang

ASXL1 is mutated/deleted with high frequencies in multiple forms of myeloid malignancies, and its alterations are associated with poor prognosis. De novo ASXL1 mutations cause Bohring-Opitz syndrome characterized by multiple congenital malformations. We show that Asxl1 deletion in mice led to developmental abnormalities including dwarfism, anophthalmia, and 80% embryonic lethality. Surviving Asxl1(-/-) mice lived for up to 42 days and developed features of myelodysplastic syndrome (MDS), including dysplastic neutrophils and multiple lineage cytopenia. Asxl1(-/-) mice had a reduced hematopoietic stem cell (HSC) pool, and Asxl1(-/-) HSCs exhibited decreased hematopoietic repopulating capacity, with skewed cell differentiation favoring granulocytic lineage. Asxl1(+/-) mice also developed mild MDS-like disease, which could progress to MDS/myeloproliferative neoplasm, demonstrating a haploinsufficient effect of Asxl1 in the pathogenesis of myeloid malignancies. Asxl1 loss led to an increased apoptosis and mitosis in Lineage(-)c-Kit(+) (Lin(-)c-Kit(+)) cells, consistent with human MDS. Furthermore, Asxl1(-/-) Lin(-)c-Kit(+) cells exhibited decreased global levels of H3K27me3 and H3K4me3 and altered expression of genes regulating apoptosis (Bcl2, Bcl2l12, Bcl2l13). Collectively, we report a novel ASXL1 murine model that recapitulates human myeloid malignancies, implying that Asxl1 functions as a tumor suppressor to maintain hematopoietic cell homeostasis. Future work is necessary to clarify the contribution of microenvironment to the hematopoietic phenotypes observed in the constitutional Asxl1(-/-) mice.


Science Advances | 2017

ASXL1 interacts with the cohesin complex to maintain chromatid separation and gene expression for normal hematopoiesis.

Zhaomin Li; Peng Zhang; Aimin Yan; Zhengyu Guo; Yuguang Ban; Jin Li; Shi Chen; Hui Yang; Yongzheng He; Jianping Li; Ying Guo; Wen Zhang; Ehsan Hajiramezanali; Huangda An; Darlene Fajardo; J. William Harbour; Yijun Ruan; Stephen D. Nimer; Peng Yu; Xi Chen; Mingjiang Xu; Feng Chun Yang

Asxl1 loss–induced myeloid malignancies are mediated partially through impaired ASXL1 cohesin interaction and cohesin functions. ASXL1 is frequently mutated in a spectrum of myeloid malignancies with poor prognosis. Loss of Asxl1 leads to myelodysplastic syndrome–like disease in mice; however, the underlying molecular mechanisms remain unclear. We report that ASXL1 interacts with the cohesin complex, which has been shown to guide sister chromatid segregation and regulate gene expression. Loss of Asxl1 impairs the cohesin function, as reflected by an impaired telophase chromatid disjunction in hematopoietic cells. Chromatin immunoprecipitation followed by DNA sequencing data revealed that ASXL1, RAD21, and SMC1A share 93% of genomic binding sites at promoter regions in Lin−cKit+ (LK) cells. We have shown that loss of Asxl1 reduces the genome binding of RAD21 and SMC1A and alters the expression of ASXL1/cohesin target genes in LK cells. Our study underscores the ASXL1-cohesin interaction as a novel means to maintain normal sister chromatid separation and regulate gene expression in hematopoietic cells.


Blood | 2017

Gain-of-function of ASXL1 truncating protein in the pathogenesis of myeloid malignancies

Hui Yang; Stefan Kurtenbach; Ying Guo; Ines Lohse; Michael A. Durante; Jianping Li; Zhaomin Li; Hassan Al-Ali; Lingxiao Li; Zizhen Chen; Matthew G. Field; Peng Zhang; Shi Chen; Shohei Yamamoto; Zhuo Li; Yuan Zhou; Stephen D. Nimer; J. William Harbour; Claes Wahlestedt; Mingjiang Xu; Feng Chun Yang

Additional Sex Combs-Like 1 (ASXL1) is mutated at a high frequency in all forms of myeloid malignancies associated with poor prognosis. We generated a Vav1 promoter-driven Flag-Asxl1Y588X transgenic mouse model, Asxl1Y588X Tg, to express a truncated FLAG-ASXL1aa1-587 protein in the hematopoietic system. The Asxl1Y588X Tg mice had an enlarged hematopoietic stem cell (HSC) pool, shortened survival, and predisposition to a spectrum of myeloid malignancies, thereby recapitulating the characteristics of myeloid malignancy patients with ASXL1 mutations. ATAC- and RNA-sequencing analyses revealed that the ASXL1aa1-587 truncating protein expression results in more open chromatin in cKit+ cells compared with wild-type cells, accompanied by dysregulated expression of genes critical for HSC self-renewal and differentiation. Liquid chromatography-tandem mass spectrometry and coimmunoprecipitation experiments showed that ASXL1aa1-587 acquired an interaction with BRD4. An epigenetic drug screening demonstrated a hypersensitivity of Asxl1Y588X Tg bone marrow cells to BET bromodomain inhibitors. This study demonstrates that ASXL1aa1-587 plays a gain-of-function role in promoting myeloid malignancies. Our model provides a powerful platform to test therapeutic approaches of targeting the ASXL1 truncation mutations in myeloid malignancies.


Stem cell reports | 2016

Loss of Asxl1 Alters Self-Renewal and Cell Fate of Bone Marrow Stromal Cell, Leading to Bohring-Opitz-like Syndrome in Mice

Peng Zhang; Xing C; Steven D. Rhodes; Yongzheng He; Kai Deng; Zhaomin Li; Fuhong He; Caiying Zhu; Lihn Nguyen; Yuan Zhou; Shi Chen; Khalid S. Mohammad; Theresa A. Guise; Omar Abdel-Wahab; Mingjiang Xu; Qianfei Wang; Feng Chun Yang

Summary De novo ASXL1 mutations are found in patients with Bohring-Opitz syndrome, a disease with severe developmental defects and early childhood mortality. The underlying pathologic mechanisms remain largely unknown. Using Asxl1-targeted murine models, we found that Asxl1 global loss as well as conditional deletion in osteoblasts and their progenitors led to significant bone loss and a markedly decreased number of bone marrow stromal cells (BMSCs) compared with wild-type littermates. Asxl1−/− BMSCs displayed impaired self-renewal and skewed differentiation, away from osteoblasts and favoring adipocytes. RNA-sequencing analysis revealed altered expression of genes involved in cell proliferation, skeletal development, and morphogenesis. Furthermore, gene set enrichment analysis showed decreased expression of stem cell self-renewal gene signature, suggesting a role of Asxl1 in regulating the stemness of BMSCs. Importantly, re-introduction of Asxl1 normalized NANOG and OCT4 expression and restored the self-renewal capacity of Asxl1−/− BMSCs. Our study unveils a pivotal role of ASXL1 in the maintenance of BMSC functions and skeletal development.


Cell discovery | 2018

Loss of ASXL1 in the bone marrow niche dysregulates hematopoietic stem and progenitor cell fates

Peng Zhang; Zizhen Chen; Rong Li; Ying Guo; Hui Shi; Jie Bai; Hui Yang; Mengyao Sheng; Zhaomin Li; Zhuo Li; Jianping Li; Shi Chen; Weiping Yuan; Tao Cheng; Mingjiang Xu; Yuan Zhou; Feng Chun Yang

Somatic or de novo mutations of Additional sex combs-like 1 (ASXL1) frequently occur in patients with myeloid malignancies or Bohring-Opitz syndrome, respectively. We have reported that global loss of Asxl1 leads to the development of myeloid malignancies and impairs bone marrow stromal cell (BMSC) fates in mice. However, the impact of Asxl1 deletion in the BM niche on hematopoiesis remains unclear. Here, we showed that BMSCs derived from chronic myelomonocytic leukemia patients had reduced expression of ASXL1, which impaired the maintaining cord blood CD34+ cell colony-forming capacity with a myeloid differentiation bias. Furthermore, Asxl1 deletion in the mouse BMSCs altered hematopoietic stem and progenitor cell (HSC/HPC) pool and a preferential myeloid lineage increment. Immunoprecipitation and ChIP-seq analyses demonstrated a novel interaction of ASXL1 with the core subunits of RNA polymerase II (RNAPII) complex. Convergent analyses of RNA-seq and ChIP-seq data revealed that loss of Asxl1 deregulated RNAPII transcriptional function and altered the expression of genes critical for HSC/HPC maintenance, such as Vcam1. Altogether, our study provides a mechanistic insight into the function of ASXL1 in the niche to maintain normal hematopoiesis; and ASXL1 alteration in, at least, a subset of the niche cells induces myeloid differentiation bias, thus, contributes the progression of myeloid malignancies.


Nature Communications | 2017

Loss of Asxl2 leads to myeloid malignancies in mice.

Jianping Li; Fuhong He; Peng Zhang; Shi Chen; Hui Shi; Yanling Sun; Ying Guo; Hui Yang; Na Man; Sarah M. Greenblatt; Zhaomin Li; Zhengyu Guo; Yuan Zhou; Lan Wang; Lluis Morey; Sion L. Williams; Xi Chen; Qun Tian Wang; Stephen D. Nimer; Peng Yu; Qianfei Wang; Mingjiang Xu; Feng Chun Yang

ASXL2 is frequently mutated in acute myeloid leukaemia patients with t(8;21). However, the roles of ASXL2 in normal haematopoiesis and the pathogenesis of myeloid malignancies remain unknown. Here we show that deletion of Asxl2 in mice leads to the development of myelodysplastic syndrome (MDS)-like disease. Asxl2−/− mice have an increased bone marrow (BM) long-term haematopoietic stem cells (HSCs) and granulocyte–macrophage progenitors compared with wild-type controls. Recipients transplanted with Asxl2−/− and Asxl2+/− BM cells have shortened lifespan due to the development of MDS-like disease or myeloid leukaemia. Paired daughter cell assays demonstrate that Asxl2 loss enhances the self-renewal of HSCs. Deletion of Asxl2 alters the expression of genes critical for HSC self-renewal, differentiation and apoptosis in Lin−cKit+ cells. The altered gene expression is associated with dysregulated H3K27ac and H3K4me1/2. Our study demonstrates that ASXL2 functions as a tumour suppressor to maintain normal HSC function.


Journal of Bone and Mineral Research | 2015

Nf1 Haploinsufficiency Alters Myeloid Lineage Commitment and Function, Leading to Deranged Skeletal Homeostasis

Steven D. Rhodes; Hao Yang; Ruizhi Dong; Keshav Menon; Yongzheng He; Zhaomin Li; Shi Chen; Karl Staser; Li Jiang; Xiaohua Wu; Xianlin Yang; Xianghong Peng; Khalid S. Mohammad; Theresa A. Guise; Mingjiang Xu; Feng Chun Yang

Although nullizygous loss of NF1 leads to myeloid malignancies, haploinsufficient loss of NF1 (Nf1) has been shown to contribute to osteopenia and osteoporosis which occurs in approximately 50% of neurofibromatosis type 1 (NF1) patients. Bone marrow mononuclear cells of haploinsufficient NF1 patients and Nf1+/– mice exhibit increased osteoclastogenesis and accelerated bone turnover; however, the culprit hematopoietic lineages responsible for perpetuating these osteolytic manifestations have yet to be elucidated. Here we demonstrate that conditional inactivation of a single Nf1 allele within the myeloid progenitor cell population (Nf1‐LysM) is necessary and sufficient to promote multiple osteoclast gains‐in‐function, resulting in enhanced osteoclastogenesis and accelerated osteoclast bone lytic activity in response to proresorptive challenge in vivo. Surprisingly, mice conditionally Nf1 heterozygous in mature, terminally differentiated osteoclasts (Nf1‐Ctsk) do not exhibit any of these skeletal phenotypes, indicating a critical requirement for Nf1 haploinsufficiency at a more primitive/progenitor stage of myeloid development in perpetuating osteolytic activity. We further identified p21Ras‐dependent hyperphosphorylation of Pu.1 within the nucleus of Nf1 haploinsufficient myelomonocytic osteoclast precursors, providing a novel therapeutic target for the potential treatment of NF1 associated osteolytic manifestations.


PMC | 2017

ASXL1 interacts with the cohesin complex to maintain chromatid separation and gene expression for normal hematopoiesis

Zhaomin Li; Peng Zhang; Aimin Yan; Zhengyu Guo; Yuguang Ban; Jin Li; Shi Chen; Hui Yang; Yongzheng He; Jianping Li; Ying Guo; Wen Zhang; Ehsan Hajiramezanali; Huangda An; Darlene Fajardo; J. William Harbour; Yijun Ruan; Stephen D. Nimer; Peng Yu; Xi Chen; Mingjiang Xu; Feng Chun Yang


PMC | 2015

Nf1 haploinsufficiency alters myeloid lineage commitment and function, leading to deranged skeletal homeostasis

Steven D. Rhodes; Hao Yang; Ruizhi Dong; Keshav Menon; Yongzheng He; Zhaomin Li; Shi Chen; Karl Staser; Li Jiang; Xiaohua Wu; Xianlin Yang; Xianghong Peng; Khalid S. Mohammad; Theresa A. Guise; Mingjiang Xu; Feng Chun Yang


PMC | 2014

Loss of Asxl1 leads to myelodysplastic syndrome-like disease in mice

Jiapeng Wang; Zhaomin Li; Yongzheng He; Feng Pan; Shi Chen; Steven D. Rhodes; Lihn Nguyen; Jin Yuan; Li Jiang; Xianlin Yang; Ophelia Weeks; Ziyue Liu; Jiehao Zhou; Hongyu Ni; Chen-Leng Cai; Mingjiang Xu; Feng Chun Yang

Collaboration


Dive into the Zhaomin 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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge