Network


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

Hotspot


Dive into the research topics where Anling Liu is active.

Publication


Featured researches published by Anling Liu.


Breast Cancer Research and Treatment | 2012

Targeting of mTORC2 prevents cell migration and promotes apoptosis in breast cancer

Haiyan Li; Jun Lin; Xiaokai Wang; Guangyu Yao; Liping Wang; Hang Zheng; Cuilan Yang; Chunhong Jia; Anling Liu; Xiaochun Bai

Most of breast cancers are resistant to mammalian target of rapamycin complex 1 (mTORC1) inhibitors rapamycin and rapalogs. Recent studies indicate mTORC2 is emerging as a promising cancer therapeutic target. In this study, we compared the inhibitory effects of targeting mTORC1 with mTORC2 on a variety of breast cancer cell lines and xenograft. We demonstrated that inhibition of mTORC1/2 by mTOR kinase inhibitors PP242 and OSI-027 effectively suppress phosphorylation of Akt (S473) and breast cancer cell proliferation. Targeting of mTORC2 either by kinase inhibitors or rictor knockdown, but not inhibition of mTORC1 either by rapamycin or raptor knockdown promotes serum starvation- or cisplatin-induced apoptosis. Furthermore, targeting of mTORC2 but not mTORC1 efficiently prevent breast cancer cell migration. Most importantly, in vivo administration of PP242 but not rapamycin as single agent effectively prevents breast tumor growth and induces apoptosis in xenograft. Our data suggest that agents that inhibit mTORC2 may have advantages over selective mTORC1 inhibitors in the treatment of breast cancers. Given that mTOR kinase inhibitors are in clinical trials, this study provides a strong rationale for testing the use of mTOR kinase inhibitors or combination of mTOR kinase inhibitors and cisplatin in the clinic.


Cellular Signalling | 2010

Multi-mechanisms are involved in reactive oxygen species regulation of mTORC1 signaling

Ming Li; Li Zhao; Jun Liu; Anling Liu; Chunhong Jia; Dongzhu Ma; Yu Jiang; Xiaochun Bai

The mammalian target of rapamycin complex 1(mTORC1) integrates diverse signals to control cell growth, proliferation, survival, and metabolism. Role of reactive oxygen species (ROS) on mTORC1 signaling remains obscure and mechanisms through which ROS modulate mTORC1 are not known.We demonstrate that low doses ROS exposure stimulate mTORC1 while high concentrations or long-term ROS treatment decrease mTORC1 activity in vivo and in a variety of cell lines. The dose/time needed for inhibition or activation are cell type dependent. In HEK293 cells hydrogen peroxide (H(2)O(2)) stimulates phosphorylation of AMP-activated kinase (AMPK) (T172) and Raptor (S792), enhances association of activated AMPK with Raptor. Furthermore, AMPK inhibitor compound c inhibits H(2)O(2)-induced Raptor (S792) phosphorylation and reverses H(2)O(2)-induced dephosphorylation of mTORC1 downstream targets p70-S6K1 (T389), S6 (S235/236) and 4E-BP1 (T37/46). H(2)O(2) also stimulates association of endogenous protein phosphatase 2A catalytic subunit (PP2Ac) with p70-S6K1. Like compound c, inhibitor of PP2A, okadaic acid partially reverses inactivation of mTORC1 substrates induced by H(2)O(2). Moreover, inhibition of PP2A and AMPK partially rescued cells from H(2)O(2)-induced cell death. High doses of H(2)O(2) inhibit while low doses of H(2)O(2) activate mTORC1 both in TSC2(-/-) P53(-/-) and TSC2(+/+) P53(-/-) MEFs. These data suggest that PP2A and AMPK-mediated phosphorylation of Raptor mediate H(2)O(2)-induced inhibition of mTORC1 signaling.


Nature Communications | 2016

mTORC1 regulates PTHrP to coordinate chondrocyte growth, proliferation and differentiation

Bo Yan; Zhongmin Zhang; Dadi Jin; Chen Cai; Chunhong Jia; Wen Liu; Ting Wang; Sheng-fa Li; Haiyan Zhang; Bin Huang; Pinglin Lai; Hua Wang; Anling Liu; Chun Zeng; Daozhang Cai; Yu Jiang; Xiaochun Bai

Precise coordination of cell growth, proliferation and differentiation is essential for the development of multicellular organisms. Here, we report that although the mechanistic target of rapamycin complex 1 (mTORC1) activity is required for chondrocyte growth and proliferation, its inactivation is essential for chondrocyte differentiation. Hyperactivation of mTORC1 via TSC1 gene deletion in chondrocytes causes uncoupling of the normal proliferation and differentiation programme within the growth plate, resulting in uncontrolled cell proliferation, and blockage of differentiation and chondrodysplasia in mice. Rapamycin promotes chondrocyte differentiation and restores these defects in mutant mice. Mechanistically, mTORC1 downstream kinase S6K1 interacts with and phosphorylates Gli2, and releases Gli2 from SuFu binding, resulting in nuclear translocation of Gli2 and transcription of parathyroid hormone-related peptide (PTHrP), a key regulator of bone development. Our findings demonstrate that dynamically controlled mTORC1 activity is crucial to coordinate chondrocyte proliferation and differentiation partially through regulating Gli2/PTHrP during endochondral bone development.


Breast Cancer Research and Treatment | 2012

mTORC1 is a target of nordihydroguaiaretic acid to prevent breast tumor growth in vitro and in vivo

Yue Zhang; Song Xu; Jun Lin; Guangyu Yao; Zelong Han; Bo Liang; Zhenhong Zou; Zhenguo Chen; Qiancheng Song; Yifan Dai; Tianming Gao; Anling Liu; Xiaochun Bai

Nordihydroguaiaretic acid (NDGA) is a natural phenolic compound isolated from the creosote bush Larrea divaricata, which has anti-tumor activities both in vitro and in vivo. Its analogs are in clinical development for use in refractory solid tumors. But the mechanisms underlying the anti-cancer effect of NDGA are not fully understood. In this study, we identified mammalian target of rapamycin complex 1 (mTORC1) as a target of NDGA both in cultured breast cancer cells and in xenograft models. NDGA effectively inhibited basal level of mTORC1 but not mTORC2 activity in breast cancer cell lines. NDGA also suppressed mTORC1 downstream signaling such as expression of cyclin D1, hypoxia-inducible factor-α and VEGF, and prevented proliferation in breast cancer cells. Although NDGA stimulated AMP-activated protein kinase (AMPK)/tuberous sclerosis complex 2 (TSC2) signaling, which negatively regulates mTORC1, AMPK and TSC2 deletion could not diminish the inhibition of mTORC1 by NDGA. Subsequent studies revealed that NDGA may also direct target mTORC1 complex because NDGA suppressed amino acids- and insulin-stimulated mTORC1 and acted like rapamycin to disrupt mTOR–Raptor interaction. Most importantly, NDGA repressed breast tumor growth and targeted mTORC1 and its downstream signaling in xenograft models. Together our data provide a novel mechanism for NDGA activity which could help explain its anti-cancer activity. Disruption of mTOR–Raptor complex and activation of AMPK/TSC signaling may contribute to inhibitory effects of NDGA against mTORC1. Our data also raise the possibility that NDGA, as an mTORC1 inhibitor, may have a broad spectrum of action on breast cancers.


Journal of Bone and Mineral Research | 2017

mTORC1 Inhibits NF-κB/NFATc1 Signaling and Prevents Osteoclast Precursor Differentiation, In Vitro and In Mice.

Yue Zhang; Song Xu; Kai Li; Kang Tan; Kangyan Liang; Jian Wang; Junhui Shen; Wenchong Zou; Le Hu; Daozhang Cai; Changhai Ding; Mangmang Li; Guozhi Xiao; Bin Liu; Anling Liu; Xiaochun Bai

The mechanistic target of rapamycin complex 1 (mTORC1) is a critical sensor for bone homeostasis and bone formation; however, the role of mTORC1 in osteoclast development and the underlying mechanisms have not yet been fully established. Here, we found that mTORC1 activity declined during osteoclast precursors differentiation in vitro and in vivo. We further targeted deletion of Raptor (mTORC1 key component) or Tsc1 (mTORC1 negative regulator) to constitutively inhibit or activate mTORC1 in osteoclast precursors (monocytes/macrophages), using LyzM‐cre mice. Osteoclastic formation was drastically increased in cultures of Raptor deficient bone marrow monocytes/macrophages (BMMs), and Raptor‐deficient mice displayed osteopenia with enhanced osteoclastogenesis. Conversely, BMMs lacking Tsc1 exhibited a severe defect in osteoclast‐like differentiation and absorptive function, both of which were restored following rapamycin treatment. Importantly, expression of nuclear factor κ‐light‐chain‐enhancer of activated B cells (NF‐κB) and nuclear factor of activated T cells, cytoplasmic 1 (NFATc1), transcription factors that are essential for osteoclast differentiation was negatively regulated by mTORC1 in osteoclast lineages. These results provide evidence that mTORC1 plays as a critical role as an osteoclastic differentiation‐limiting signal and suggest a potential drawback in treating bone loss–related diseases with mTOR inhibitors clinically.


Cell Death and Disease | 2016

Loss of Rictor with aging in osteoblasts promotes age-related bone loss

Pinling Lai; Qiancheng Song; Cheng Yang; Zhen Li; Sichi Liu; Bin Liu; Mangmang Li; Hongwen Deng; Daozhang Cai; Dadi Jin; Anling Liu; Xiaochun Bai

Osteoblast dysfunction is a major cause of age-related bone loss, but the mechanisms underlying changes in osteoblast function with aging are poorly understood. This study demonstrates that osteoblasts in aged mice exhibit markedly impaired adhesion to the bone formation surface and reduced mineralization in vivo and in vitro. Rictor, a specific component of the mechanistic target of rapamycin complex 2 (mTORC2) that controls cytoskeletal organization and cell survival, is downregulated with aging in osteoblasts. Mechanistically, we found that an increased level of reactive oxygen species with aging stimulates the expression of miR-218, which directly targets Rictor and reduces osteoblast bone surface adhesion and survival, resulting in a decreased number of functional osteoblasts and accelerated bone loss in aged mice. Our findings reveal a novel functional pathway important for age-related bone loss and support for miR-218 and Rictor as potential targets for therapeutic intervention for age-related osteoporosis treatment.


Blood | 2017

Osteoblasts support megakaryopoiesis through production of interleukin 9

Min Xiao; Yongkui Wang; Chen Tao; Zhenyu Wang; Jun Yang; Zhenguo Chen; Zhipeng Zou; Mangmang Li; Anling Liu; Chunhong Jia; Bin Huang; Bo Yan; Pinglin Lai; Changhai Ding; Daozhang Cai; Guozhi Xiao; Yu Jiang; Xiaochun Bai

Severe thrombocytopenia is a significant challenge in patients undergoing myelosuppressive chemotherapy for malignancies. Understanding the biology of platelet-producing megakaryocytes development in the bone marrow microenvironment may facilitate the development of novel therapies to stimulate platelet production and prevent thrombocytopenia. We report here that osteoblasts supported megakaryopoiesis by secreting interleukin-9 (IL-9), which stimulated IL-9 receptor (IL-9R)/Stat3 signaling in promoting megakaryopoiesis. IL-9 production in osteoblasts was negatively regulated by the mechanistic target of rapamycin complex 1 (mTORC1) signaling in a NF-κB-dependent manner. Constitutive activation of mTORC1 inhibited IL-9 production in osteoblasts and suppressed megakaryocytic cells expansion, whereas mTORC1 inactivation increased IL-9 production and enhanced megakaryocyte and platelet numbers in mice. In mouse models, we showed that IL-9 administration stimulated megakaryopoiesis, whereas neutralizing endogenous IL-9 or IL-9R depletion inhibited the process. Importantly, we found that low doses of IL-9 efficiently prevented chemotherapy-induced thrombocytopenia (CIT) and accelerated platelet recovery after CIT. These data indicate that IL-9 is an essential regulator of megakaryopoiesis and a promising therapeutic agent for treatment of thrombocytopenia such as CIT.


Annals of the Rheumatic Diseases | 2018

Tyrosine kinase Fyn promotes osteoarthritis by activating the β-catenin pathway

Kai Li; Yue Zhang; Yuwei Zhang; Wenqing Jiang; Junhui Shen; Song Xu; Daozhang Cai; Jie Shen; Bin Huang; Mangmang Li; Qiancheng Song; Yu Jiang; Anling Liu; Xiaochun Bai

Objectives To investigate the role of tyrosine kinase Fyn in the development of osteoarthritis (OA) and the underlying mechanisms, and to define whether targeting Fyn could prevent OA in mice. Methods Cartilage samples from normal and aged mice were analysed with proteome-wide screening. Fyn expression was examined with immunofluorescence in human and age-dependent or experimental mouse OA cartilage samples. Experimental OA in Fyn-knockout mice was induced by destabilisation of the medial meniscus. Primary cultured mouse chondrocytes were treated with proinflammatory cytokine interleukin-1β. The inhibitor of Src kinase family, AZD0530 (saracatinib), and inhibitor of Fyn, PP1, were used to treat experimental OA in mice. Results Fyn expression was markedly upregulated in human OA cartilage and in cartilage from aged mice and those with post-traumatic OA. Fyn accumulates in articular chondrocytes and interacts directly with and phosphorylates β-catenin at Tyr142, which stabilises β-catenin and promotes its nuclear translocation. The deletion of Fyn effectively delayed the development of post-traumatic and age-dependent OA in mice. Fyn inhibitors AZD0530 and PP1 significantly attenuated OA progression by blocking the β-catenin pathway and reducing the levels of extracellular matrix catabolic enzymes in the articular cartilage. Conclusions Fyn accumulates and activates β-catenin signalling in chondrocytes, accelerating the degradation of the articular cartilage and OA development. Targeting Fyn is a novel and potentially therapeutic approach to the treatment of OA.


Journal of Bone and Mineral Research | 2018

Inactivation of mTORC1 Signaling in Osterix-Expressing Cells Impairs B-cell Differentiation: mTORC1 IN OSX-EXPRESSING CELLS REGULATES B-CELL DEVELOPMENT

Yongkui Wang; Min Xiao; Chen Tao; Jing Chen; Zhenyu Wang; Jun Yang; Zhenguo Chen; Zhipeng Zou; Anling Liu; Daozhang Cai; Yu Jiang; Changhai Ding; Mangmang Li; Xiaochun Bai

Osteoblasts provide a microenvironmental niche for B‐cell commitment and maturation in the bone marrow (BM). Any abnormity of osteoblasts function may result in the defect of B lymphopoiesis. Signaling from mechanistic target of rapamycin complex 1 (mTORC1) has been implicated in regulating the expansion and differentiation of osteoblasts. Thus, we raise a hypothesis that mTORC1 signaling in osteoblasts plays a vital role in B‐cell development. Inactivation of mTORC1 in osterix‐expressing cells (mainly osteoblast lineage) through Osx‐Cre‐directed deletion of Raptor (an mTORC1‐specific component) resulted in a reduction in the total B‐cell population in the BM, which was due to a block in early B‐cell development from the pro‐B to pre‐B cell stage. Further mechanistic studies revealed that this defect was the result of reduction of interleukin‐7 (IL‐7) expression in osterix‐expressing immature osteoblasts, which caused the abnormality of IL‐7/Stat5 signaling in early B lymphocytes, leading to an increased apoptosis of pre‐B plus immature B cells. In vitro and in vivo studies demonstrated that the addition of exogenous IL‐7 partially restored B lymphopoiesis in the BM of Raptor mutant mice. Furthermore, total BM cells cultured in conditioned media from Raptor null immature osteoblasts or media with anti‐IL‐7 neutralizing antibody failed to differentiate into pre‐B and immature B cells, indicating that inactivation of mTORC1 in immature osteoblast cannot fully support normal B‐cell development. Taken together, these findings demonstrate a novel role for mTORC1 in the regulation of bone marrow environments that support B‐cell differentiation via regulating IL‐7 expression.


Bone | 2018

Bone and plasma citrate is reduced in osteoporosis

Hongdong Chen; Yeyang Wang; Huaiqian Dai; Xinggui Tian; Zhong-Kai Cui; Zhenguo Chen; Le Hu; Qiancheng Song; Anling Liu; Zhiyong Zhang; Guozhi Xiao; Jian Yang; Yu Jiang; Xiaochun Bai

High concentration of citrate exists in bone of humans and all osteo-vertebrates, and citrate incorporation imparts important biomechanical and other functional properties to bone. However, which cells are responsible for citrate production in bone remains unclear and whether the citrate component changes with bone loss during osteoporosis is also not known. Here, we show that the citrate content is markedly reduced in the bone of mice or rats with age-related, ovariectomy-induced or retinoic acid-induced bone loss. Plasmic citrate is also downregulated in osteoporotic animals. Importantly, the plasmic citrate level of aged osteoporotic males is significantly lower than that of young healthy males and positively correlates with human lumbar spine bone mineral density (BMD) and total hip BMD. Furthermore, citrate production increases with in vitro osteoblastic differentiation, accompanied by upregulation of proteins involved in citrate secretion, suggesting that osteoblasts are highly specialized cells that produce citrate in bone. Our findings establish a novel relationship between citrate content and bone loss-related diseases such as osteoporosis, suggesting a critical role of bone citrate in the maintenance of the citrate balance in the circulation. Serum citrate level may thus represent a novel marker for osteoporosis.

Collaboration


Dive into the Anling Liu's collaboration.

Top Co-Authors

Avatar

Xiaochun Bai

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Daozhang Cai

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Yu Jiang

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Zhenguo Chen

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Chunhong Jia

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Mangmang Li

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Qiancheng Song

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Changhai Ding

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Song Xu

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Yue Zhang

Southern Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge