Network


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

Hotspot


Dive into the research topics where Baixia Hao is active.

Publication


Featured researches published by Baixia Hao.


Journal of Biological Chemistry | 2013

Two Pore Channel 2 (TPC2) Inhibits Autophagosomal-Lysosomal Fusion by Alkalinizing Lysosomal pH

Yingying Lu; Baixia Hao; Richard Graeff; Connie W. M. Wong; Wu-Tian Wu; Jianbo Yue

Background: The role and mechanism of NAADP, an endogenous Ca2+ mobilizing nucleotide, in autophagic regulation remain to be determined. Results: Activation of NAADP/TPC2 signaling induced the accumulation of autophagosomes. Conclusion: The NAADP/TPC2 signaling inhibits autophagosomal-lysosomal fusion by alkalinizing lysosomal pH. Significance: Development of agonists or antagonists of NAADP should provide a novel approach to specifically manipulate autophagy. Autophagy is an evolutionarily conserved lysosomal degradation pathway, yet the underlying mechanisms remain poorly understood. Nicotinic acid adenine dinucleotide phosphate (NAADP), one of the most potent Ca2+ mobilizing messengers, elicits Ca2+ release from lysosomes via the two pore channel 2 (TPC2) in many cell types. Here we found that overexpression of TPC2 in HeLa or mouse embryonic stem cells inhibited autophagosomal-lysosomal fusion, thereby resulting in the accumulation of autophagosomes. Treatment of TPC2 expressing cells with a cell permeant-NAADP agonist, NAADP-AM, further induced autophagosome accumulation. On the other hand, TPC2 knockdown or treatment of cells with Ned-19, a NAADP antagonist, markedly decreased the accumulation of autophagosomes. TPC2-induced accumulation of autophagosomes was also markedly blocked by ATG5 knockdown. Interestingly, inhibiting mTOR activity failed to increase TPC2-induced autophagosome accumulation. Instead, we found that overexpression of TPC2 alkalinized lysosomal pH, and lysosomal re-acidification abolished TPC2-induced autophagosome accumulation. In addition, TPC2 overexpression had no effect on general endosomal-lysosomal degradation but prevented the recruitment of Rab-7 to autophagosomes. Taken together, our data demonstrate that TPC2/NAADP/Ca2+ signaling alkalinizes lysosomal pH to specifically inhibit the later stage of basal autophagy progression.


Journal of Biological Chemistry | 2012

A novel fluorescent cell membrane permeable caged cyclic ADP-Ribose analogue

Peilin Yu; Zhe-Hao Zhang; Baixia Hao; Yong Juan Zhao; Lihe Zhang; Hon Cheung Lee; Liangren Zhang; Jianbo Yue

Background: The available agonists for cADPR, an endogenous Ca2+-mobilizing nucleotide, are either weak or not cell-permeant. Results: We synthesized a coumarin-caged isopropylidene-protected cIDPRE (Co-i-cIDPRE), which is a potent and cell-permeant cADPR agonist. Conclusion: Uncaging of Co-i-cIDPRE activates RyRs for Ca2+ mobilization and triggers Ca2+ influx via TRPM2. Significance: Co-i-cIDPRE should provide a valuable tool to study cADPR/Ca2+ signaling. Cyclic adenosine diphosphate ribose is an endogenous Ca2+ mobilizer involved in diverse cellular processes. A cell membrane-permeable cyclic adenosine diphosphate ribose analogue, cyclic inosine diphosphoribose ether (cIDPRE), can induce Ca2+ increase in intact human Jurkat T-lymphocytes. Here we synthesized a coumarin-caged analogue of cIDPRE (Co-i-cIDPRE), aiming to have a precisely temporal and spatial control of bioactive cIDPRE release inside the cell using UV uncaging. We showed that Co-i-cIDPRE accumulated inside Jurkat cells quickly and efficiently. Uncaging of Co-i-cIDPRE evoked Ca2+ release from endoplasmic reticulum, with concomitant Ca2+ influx in Jurkat cells. Ca2+ release evoked by uncaged Co-i-cIDPRE was blocked by knockdown of ryanodine receptors (RyRs) 2 and 3 in Jurkat cells. The associated Ca2+ influx, on the other hand, was abolished by double knockdown of Stim1 and TRPM2 in Jurkat cells. Furthermore, Ca2+ release or influx evoked by uncaged Co-i-cIDPRE was recapitulated in HEK293 cells that overexpress RyRs or TRPM2, respectively, but not in wild-type cells lacking these channels. In summary, our results indicate that uncaging of Co-i-cIDPRE incites Ca2+ release from endoplasmic reticulum via RyRs and triggers Ca2+ influx via TRPM2.


Stem Cell Research | 2014

Role of STIM1 in survival and neural differentiation of mouse embryonic stem cells independent of Orai1-mediated Ca2 + entry☆

Baixia Hao; Yingying Lu; Qian Wang; Wenjing Guo; King-Ho Cheung; Jianbo Yue

Store-operated Ca(2+) entry (SOCE) is an important Ca(2+) influx pathway in non-excitable cells. STIM1, an ER Ca(2+) sensor, and Orai1, a plasma membrane Ca(2+) selective channel, are the two essential components of the Ca(2+) release activated channel (CRAC) responsible for SOCE activity. Here we explored the role of STIM1 and Orai1 in neural differentiation of mouse embryonic stem (ES) cells. We found that STIM1 and Orai1 were expressed and functionally active in ES cells, and expressions of STIM1 and Orai1 were dynamically regulated during neural differentiation of mouse ES cells. STIM1 knockdown inhibited the differentiation of mouse ES cells into neural progenitors, neurons, and astrocytes. In addition, STIM1 knockdown caused severe cell death and markedly suppressed the proliferation of neural progenitors. Surprisingly, Orai1 knockdown had little effect on neural differentiation of mouse ES cells, but the neurons derived from Orai1 knockdown ES cells, like those from STIM1 knockdown cells, had defective SOCE. Taken together, our data indicate that STIM1 is involved in both early neural differentiation of ES cells and survival of early differentiated ES cells independent of Orai1-mediated SOCE.


Communicative & Integrative Biology | 2013

NAADP/TPC2/Ca2+ Signaling Inhibits Autophagy

Yingying Lu; Baixia Hao; Richard Graeff; Jianbo Yue

Nicotinic adenine acid dinucleotide phosphate (NAADP) is one of the most potent endogenous Ca2+ mobilizing messengers. NAADP mobilizes Ca2+ from an acidic lysosome-related store, which can be subsequently amplified into global Ca2+ waves by calcium-induced calcium release (CICR) from ER/SR via Ins(1,4,5)P3 receptors or ryanodine receptors. A body of evidence indicates that 2 pore channel 2 (TPC2), a new member of the superfamily of voltage-gated ion channels containing 12 putative transmembrane segments, is the long sought after NAADP receptor. Activation of NAADP/TPC2/Ca2+ signaling inhibits the fusion between autophagosome and lysosome by alkalizing the lysosomal pH, thereby arresting autophagic flux. In addition, TPC2 is downregulated during neural differentiation of mouse embryonic stem (ES) cells, and TPC2 downregulation actually facilitates the neural lineage entry of ES cells. Here we propose the mechanism underlying how NAADP-induced Ca2+ release increases lysosomal pH and discuss the role of TPC2 in neural differentiation of mouse ES cells.


Stem Cells | 2015

CD38 Is Required for Neural Differentiation of Mouse Embryonic Stem Cells by Modulating Reactive Oxygen Species

Wenjie Wei; Yingying Lu; Baixia Hao; Kehui Zhang; Qian Wang; Andrew L. Miller; Liangren Zhang; Lihe Zhang; Jianbo Yue

CD38 is a multifunctional membrane enzyme and the main mammalian ADP‐ribosyl cyclase, which catalyzes the synthesis and hydrolysis of cADPR, a potent endogenous Ca2+ mobilizing messenger. Here, we explored the role of CD38 in the neural differentiation of mouse embryonic stem cells (ESCs). We found that the expression of CD38 was decreased during the differentiation of mouse ESCs initiated by adherent monoculture. Perturbing the CD38/cADPR signaling by either CD38 knockdown or treatment of cADPR antagonists inhibited the neural commitment of mouse ESCs, whereas overexpression of CD38 promoted it. Moreover, CD38 knockdown dampened reactive oxygen species (ROS) production during neural differentiation of ESCs by inhibiting NADPH oxidase activity, while CD38 overexpression enhanced it. Similarly, application of hydrogen peroxide mitigated the inhibitory effects of CD38 knockdown on neural differentiation of ESCs. Taken together, our data indicate that the CD38 signaling pathway is required for neural differentiation of mouse ESCs by modulating ROS production. Stem Cells 2015;33:2664–2673


Experimental Cell Research | 2013

Requirement of B-Raf, C-Raf, and A-Raf for the growth and survival of mouse embryonic stem cells

Wenjing Guo; Baixia Hao; Qian Wang; Yingying Lu; Jianbo Yue

Extracellular signal-regulated kinases (ERKs) have been implicated to be dispensable for self-renewal of mouse embryonic stem (ES) cells, and simultaneous inhibition of both ERK signaling and glycogen synthase kinase 3 (GSK3) not only allows mouse ES cells to self-renew independent of extracellular stimuli but also enables more efficient derivation of naïve ES cells from mouse and rat strains. Interestingly, some ERKs stay active in mouse ES cells which are maintained in regular medium containing leukemia inhibitory factor (LIF) and bone morphogenetic protein (BMP). Yet, the upstream signaling for ERK activation and their roles in mouse ES cells, other than promoting or priming differentiation, have not been determined. Here we found that mouse ES cells express three forms of Raf kinases, A-Raf, B-Raf, and C-Raf. Knocking-down each single Raf member failed to affect the sustained ERK activity, neither did A-Raf and B-Raf double knockdown or B-Raf and C-Raf double knockdown change it in ES cells. Interestingly, B-Raf and C-Raf double knockdown, not A-Raf and B-Raf knockdown, inhibited the maximal ERK activation induced by LIF, concomitant with the slower growth of ES cells. On the other hand, A-Raf, B-Raf, and C-Raf triple knockdown markedly inhibited both the maximal and sustained ERK activity in ES cells. Moreover, Raf triple knockdown, similar to the treatment of U-0126, an MEK inhibitor, significantly inhibited the survival and proliferation of ES cells, thereby compromising the colony propagation of mouse ES cells. In summary, our data demonstrate that all three Raf members are required for ERK activation in mouse ES cells and are involved in growth and survival of mouse ES cells.


Cell Calcium | 2016

The role of Ca2+ signaling on the self-renewal and neural differentiation of embryonic stem cells (ESCs)

Baixia Hao; Sarah Elizabeth Webb; Andrew L. Miller; Jianbo Yue


Cardiovascular Research | 2018

Integrated transcriptomic and regulatory network analyses identify microRNA-200c as a novel repressor of human pluripotent stem cell-derived cardiomyocyte differentiation and maturation

Ellen Ngar Yun Poon; Baixia Hao; Daogang Guan; Mulin Jun Li; Jun Lu; Yong Yang; Binbin Wu; Stanley Chun Ming Wu; Sarah Elizabeth Webb; Yan Liang; Andrew L. Miller; Xiaoqiang Yao; Junwen Wang; Bin Yan; Kenneth R. Boheler


Archive | 2017

The Role of Ca2+ Signalling in the Differentiation of Embryonic Stem Cells (ESCs) into Cardiomyocytes

Baixia Hao; Wenjie Wei; Sarah Webb; Andrew Miller; Jianbo Yue


Archive | 2012

A Novel Fluorescent Cell Membrane Permeable Caged cyclic ADP-Ribose Analogue. Pei-Lin Yu, Zhe-Hao Zhang, Bai-Xia Hao, Yong-Juan Zhao, Li-He Zhang, Hon-Cheung Lee, Liangren Zhang*, Jianbo Yue*

Baixia Hao; Zhe-Hao Zhang; Lihe Zhang; Yong Juan Zhao; Hon Cheung Lee; Liangren Zhang; Jianbo Yue; Peilin Yu

Collaboration


Dive into the Baixia Hao's collaboration.

Top Co-Authors

Avatar

Jianbo Yue

City University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Yingying Lu

University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Andrew L. Miller

Hong Kong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Qian Wang

City University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sarah Elizabeth Webb

Hong Kong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Wenjie Wei

City University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Wenjing Guo

University of Hong Kong

View shared research outputs
Researchain Logo
Decentralizing Knowledge