Network


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

Hotspot


Dive into the research topics where Huali Chen is active.

Publication


Featured researches published by Huali Chen.


Journal of Drug Targeting | 2012

Comparison of four different peptides to enhance accumulation of liposomes into the brain

Yao Qin; Qianyu Zhang; Huali Chen; Wenmin Yuan; Rui Kuai; Fulan Xie; Li Zhang; Xiaoxiao Wang; Zhirong Zhang; Ji Liu; Qin He

The cell penetrating peptide TAT, which appears to enter cells with alacrity, can pass through the BBB efficiently. It has been indentified to enhance the brain delivery of the liposome. However, little was known about its mechanism. TAT contains a basic region consisting of six arginine and two lysine residues. These eight basic amino acids seem to be the key to its highly efficient membrane translocation and brain delivery. In this study, four selected peptides are synthesized. (1) TAT peptide with terminal Cysteine (Cys-AYGRKKRRQRRR). (2) TAT peptide with disordered sequence (Cys-RKARYRGRKRQR). (3) Glycine and glutamic acid substituted TAT peptide (Cys-AYGGQQGGQGGG). (4) R8 (Cys-RRRRRRRR). Liposomes were chosen as the delivery vehicle. The peptide was covalently bonded with the liposome. We compare four peptides for their brain targeting potential, and investigate their ability to target liposomes to the brain in vitro and in vivo. The cellular uptake of these four liposomes by brain capillary endothelial cells (BCECs) of rats and C6s and the mechanism of the pathway of endocytosis were explored. Biodistribution in vivo was also investigated qualitatively and quantitatively. The results showed that the charge of the peptide played an important role in enhancing its brain delivery. The sequence had little to do with its membrane translocation and brain delivery indicated there might be no specific receptor or transporter for the Tat peptide.


International Journal of Pharmaceutics | 2011

Liposome formulated with TAT-modified cholesterol for improving brain delivery and therapeutic efficacy on brain glioma in animals

Yao Qin; Huali Chen; Qianyu Zhang; Xiaoxiao Wang; Wenmin Yuan; Rui Kuai; Jie Tang; Li Zhang; Zhirong Zhang; Qiang Zhang; Ji Liu; Qin He

The treatment of central nervous system diseases such as brain glioma is a major challenge due to the presence of the blood-brain barrier (BBB). A cell-penetrating peptide TAT (AYGRKKRRQRRR), which appears to enter cells with alacrity, was employed to enhance the delivery efficiency of normal drug formulation to the brain. Targeting liposomal formulations often apply modified phospholipids as anchors. However, cholesterol, another liposomal component more stable and cheaper, has not been fully investigated as an alternative anchor. In our study, TAT was covalently conjugated with cholesterol for preparing doxorubicin-loaded liposome for brain glioma therapy. Cellular uptake by brain capillary endothelial cells (BCECs) and C6 glioma cells was explored. The anti-proliferative activity against C6s confirmed strong inhibitory effect of the liposomes modified with doxorubicin-loaded TAT. The bio-distribution findings in brains and hearts were evident of higher efficiency of brain delivery and lower cardiotoxic risk. The results on survival of the brain glioma-bearing animals indicate that survival time of the glioma-bearing rats treated with TAT-modified liposome was much longer than in the other groups. In conclusion, the potency of the TAT-modified liposome to enter the BBB appears to be related with the TAT on the liposomes surface. The TAT-modified liposome may improve the therapeutic efficacy on brain glioma in vitro and in vivo.


International Journal of Pharmaceutics | 2011

Liposome formulated with TAT-modified cholesterol for enhancing the brain delivery.

Yao Qin; Huali Chen; Wenmin Yuan; Rui Kuai; Qianyu Zhang; Fulan Xie; Li Zhang; Zhirong Zhang; Ji Liu; Qin He

Delivery of drugs to the brain is a major challenge due to the presence of the blood-brain barrier (BBB). The cell penetrating peptide TAT, which appears to enter cells with alacrity, can pass through the BBB efficiently. With this in mind, a novel TAT-modified liposome (TAT-LIP) was developed for overcoming the ineffective delivery of normal drug formulation to the brain. Targeting liposomal formulations are always composed of modified phospholipids as an anchor. However, cholesterol, another liposomal component, which was more stable and cheaper, has not been fully investigated as an alternative anchor. In this study, TAT was covalently conjugated with the cholesterol to prepare the liposome. The cellular uptake by brain capillary endothelial cells (BCECs) of rats and the mechanism of TAT-LIP pathway of endocytosis was explored. The blood brain barrier model in vitro was established to evaluate the transendothelial ability crossing the BBB and its transport mechanism. The biodistribution of each formulation was further identified. The results showed that the positive charge of the TAT-LIP played an important role in enhancing its brain delivery. The absorptive endocytosis might be one of the mechanisms of TAT-LIP crossing the BBB. In conclusion, the experimental data in vitro and in vivo indicated that the TAT-LIP was a promising brain drug delivery system due to its high delivery efficiency across the BBB.


European Journal of Pharmaceutical Sciences | 2010

Lactoferrin-modified procationic liposomes as a novel drug carrier for brain delivery.

Huali Chen; Lei Tang; Yao Qin; Yujia Yin; Jie Tang; Wenwei Tang; Xun Sun; Zhirong Zhang; Ji Liu; Qin He

In this study, a new drug carrier for brain delivery, lactoferrin-modified procationic liposome, was developed and evaluated in vitro and in vivo. The procationic liposomes (PCLs) were neutral or negatively charged at physiological pH, and when they touched brain capillary endothelial cells with the help of a brain-targeting ligand, lactoferrin (Lf), they were changed into cationic liposomes (CL). The PCLs and lactoferrin-modified procationic liposomes (Lf-PCLs) with different CHETA/Lf ratio were prepared and characterized. The primary brain capillary endothelial cells (BCECs) were cultured to investigate the potential cytotoxicity and uptake of liposomes in vitro. An in vitro model of the blood-brain barrier (BBB), developed by the co-culture of BCECs and astrocytes (ACs), was employed to evaluate the ability and mechanisms of liposomes to cross endothelial cells. The liposome uptake by the mouse brain in vivo was detected by HPLC-fluorescence analysis. The results indicated that compared with the conventional liposomes and CLs, PCL and Lf-PCLs showed an improved performance in the uptake efficiency and cytotoxicity. Besides the uptake mediated by clathrin-dependent endocytosis of PCL, Lf-PCL crossed the BCECs through lipid raft/caveloae-mediated endocytosis. The endocytosis involved in the transport of Lf-PCL crossing BBB was mediated by both receptor- and absorption-mediated transcytosis. Compared with the conventional liposomes, PCL and Lf-PCL-8 (CHETA/Lf ratio=1:8, w/w) were observed to show much improved characteristics of the localization in the brain. This study suggested that Lf-PCL was an available brain drug delivery carrier with potential future application.


European Journal of Pharmaceutical Sciences | 2011

Lactoferrin modified doxorubicin-loaded procationic liposomes for the treatment of gliomas

Huali Chen; Yao Qin; Qianyu Zhang; Wei Jiang; Lei Tang; Ji Liu; Qin He

In this study, a brain-targeted chemotherapeutical delivery system, doxorubicin-loaded lactoferrin-modified procationic liposome (DOX-Lf-PCL) was developed, and its therapeutic effect for glioma was evaluated. The uptake profile of various DOX formulations in vitro by primary brain capillary endothelial cells (BCECs) and glioma cell C6 were studied by laser scanning confocal microscope and flow cytometry. An intracranial tumor model of rats was employed to evaluate the therapeutic effect of DOX-Lf-PCLs for glioma. Five groups of glioma-bearing rats (total n=50) were subjected to three cycles of 2.5mg/kg body weight of doxorubicin in different formulations or normal saline (N.S.) and analyzed for survival (median survival time, Kaplan-Meier). The results indicated that compared with the DOX solution or DOX-loaded conventional liposomes (DOX-Lips), DOX-PCLs and DOX-Lf-PCLs showed an improved performance in the uptake efficiency in BCECs and C6 cells. The DOX-Lf-PCLs can inhibit the growth of C6 more efficiently in vitro than other DOX formulations. The endocytosis involved in the DOX-Lf-PCLs uptake of C6 was mediated by both receptor- and absorption-mediated transcytosis. DOX-Lf-PCLs could significantly extend the survival time compared with the N.S. control and other DOX formulations. This study showed that the therapy with DOX-Lf-PCLs offers an effective therapeutic potential for gliomas.


Biochemical Pharmacology | 2014

Ghrelin inhibits doxorubicin cardiotoxicity by inhibiting excessive autophagy through AMPK and p38-MAPK

Xue Wang; Xu-Lei Wang; Huali Chen; Dan Wu; Jia-Xiang Chen; Xiao-Xiao Wang; Ru-Li Li; Jin-Han He; Li Mo; Xiaobo Cen; Yu-Quan Wei; Wei Jiang

Doxorubicin (DOX) is a wide spectrum antitumor drug, but its clinical application is limited by the cardiotoxicity. Ghrelin, a multi-functional peptide hormone with metabolic regulation in energy homeostasis, plays important roles in cardiovascular protection. Now, the underlying mechanisms of ghrelin against DOX-induced cardiomyocyte apoptosis and atrophy are still not clear. In the present study, we revealed an autophagy-dependent mechanism involved in ghrelins protection against DOX-induced cardiomyocyte death and size decrease. We observed that DOX insult induced remarkable mortality and cardiac dysfunction in mice, and increase in LDH leakage, cardiomyocyte apoptosis and decrease in cell viability and size in mouse hearts and H9c2 cell cultures, which were effectively improved by ghrelin supplement. We further observed that the strong autophagy stirred by DOX exposure was paralleling with the serious apoptosis and size decrease in cardiomyocytes. Ghrelin, like an autophagy inhibitor, 3-MA, inhibited the DOX-induced autophagy and attenuated cardiomyocyte apoptosis and size decrease. Furthermore, ghrelin significantly reduced the intercellular oxidative stress level, a strong autophagy trigger, partly by augmenting the expression and activities of the endogenous anti-oxidative enzymes. After the further investigation in the post signaling pathways of ghrelin receptors in H9c2 cells, including ERK, p38/MAPK, JNK, AMPK and Akt, we observed that ghrelin supplement only reduced the DOX-activated AMPK and augmented the DOX-down regulated p38-MAPK and mTOR phosphorylation. Our results indicated that ghrelin effectively improved the cardiomyocyte survival and size maintenance by suppressing the excessive autophagy through both ROS inhibition and mTOR induction through suppressing AMPK activity and stimulating p38-MAPK activity.


PLOS ONE | 2013

Intermedin Suppresses Pressure Overload Cardiac Hypertrophy through Activation of Autophagy

Huali Chen; Xue Wang; MingMing Tong; Dan Wu; Sisi Wu; Jia-Xiang Chen; Xiao-Xiao Wang; Xu-Lei Wang; Yu Kang; Hong Tang; Chaoshu Tang; Wei Jiang

Left ventricular hypertrophy is a maladaptive response to pressure overload and an important risk factor for heart failure. Intermedin (IMD), a multi-functional peptide, plays important roles in cardiovascular protection. In this study, we revealed an autophagy-dependent mechanism involved in IMD’s protection against cardiac remodeling and cardiomyocyte death in heart hypertrophy. We observed that transverse aortic contraction (TAC) induction, Ang II or ISO exposure induced remarkable increase in the expression of endogenous IMD and its receptor components, CRLR, RAMP1 and RAMP3, in mouse hearts and H9c2 cell cultures, respectively. Furthermore, the heart size, heart weight/body weight ratios, cardiomyocyte size and apoptosis, interstitial collagen, hypertrophic markers including ANP and BNP expression were also significantly increased, which were effectively suppressed by IMD supplementation. In addition, IMD induced capillary angiogenesis and improved functions in hypertrophic hearts. We further observed that IMD induced strong autophagy in hypertrophic hearts and cultured cells, which was paralleling with the decrease in cardiomyocyte size and apoptosis. Furthermore, an autophagy inhibitor, 3-MA, was used to block the IMD-augmented autophagy level, and then the protection of IMD on cardiomyocyte hypertrophy and apoptosis was almost abrogated. We also observed that IMD supplementation stirred intracellular cAMP production, and augmented the ERK1/2 phosphorylation induced by Ang II/ISO exposure in H9c2 cells. In addition, we inhibited PI3K, PKA and MAPK/ERK1/2 signaling pathways by using wortamannin, H89 and PD98059, respectively, in H9c2 cells co-incubating with both IMD and Ang II or ISO, and observed that these inhibitors effectively reduced IMD-augmented autophagy level, but only H89 and PD98059 pre-incubation abrogated the anti-apoptotic action of IMD. These results indicate that the endogenous IMD and its receptor complexes are induced in hypertrophic cardiomyocytes and proposed to play an important role in the pathogenesis of cardiac hypertrophy, and the autophagy stirred by IMD supplementation is involved in its protection against cardiomyocyte hypertrophy and apoptosis through the activation of both cAMP/PKA and MAPK/ERK1/2 pathways.


DNA and Cell Biology | 2011

Salidroside protects against hydrogen peroxide-induced injury in cardiac H9c2 cells via PI3K-Akt dependent pathway.

Ye Zhu; Ya-Ping Shi; Dan Wu; Ya-Jing Ji; Xue Wang; Huali Chen; Sisi Wu; De-Jia Huang; Wei Jiang

Oxidative stress induces serious tissue injury in cardiovascular diseases. Salidroside, with its strong antioxidative and cytoprotective actions, is of particular interest in the development of antioxidative therapies for oxidative injury in cardiac diseases. We examined the pharmacological effects of salidroside on H9c2 rat cardiomyoblast cells under conditions of oxidative stress induced by hydrogen peroxide (H2O2) challenge. Salidroside attenuated H2O2-impaired cell viability in a concentration-dependent manner, and effectively inhibited cellular malondialdehyde production, lethal sarcolemmal disruption, cell necrosis, and apoptosis induced by H2O2 insult. Salidroside significantly augmented Akt phosphorylation at Serine 473 in the absence or presence of H2O2 stimulation; wortmannin, a specific inhibitor of PI3K, abrogated salidroside protection. Salidroside increased the intracellular mRNA expression and activities of catalase and Mn-superoxide dismutases in a PI3K-dependent manner. Our results indicated that salidroside protected cardiomyocytes against oxidative injury through activating the PI3K/Akt pathway and increasing the expression and activities of endogenous PI3K dependent antioxidant enzymes.


Journal of Drug Targeting | 2010

In vitro and in vivo investigation of glucose-mediated brain-targeting liposomes

Yao Qin; Wei Fan; Huali Chen; Nian Yao; Wenwei Tang; Jie Tang; Wenmin Yuan; Rui Kuai; Zhirong Zhang; Yong Wu; Qin He

New glycosyl derivative of cholesterol was synthesized as a material for preparing novel liposome to overcome the ineffective delivery of normal drug formulations to brain by targeting the (glucose transporters) GLUTs on the BBB. Coumarin-6 was used as fluorescent probe. The results have shown that the cytotoxicity for the brain capillary endothelial cells (BCECs) of the glucose-mediated brain targeting liposome containing coumarin-6 was less than that of conventional liposome. The BBB model in vitro was established by coculturing of BCECs and astrocytes (ACs) of rat to test the transendothelial ability crossing the BBB. The transendothelial ability was confirmed strengthen alone with the amount of the new glycosyl derivative of cholesterol used in liposome. After i.v. administration of LIP, control liposome (CLP), and GLP-4, the AUC0–t of coumarin-6 for GLP-4 was 2.85 times higher than that of LIP, and 3.33 times higher than that of CLP. The Cmax of CLP-4 was 1.43 times higher than that of LIP, and 3.10 times higher than that of CLP. Both pharmacokinetics and distribution in mice were also investigated to show that this novel brain targeting drug delivery system was promising.


Peptides | 2012

Ghrelin protects against cobalt chloride-induced hypoxic injury in cardiac H9c2 cells by inhibiting oxidative stress and inducing autophagy.

Xin-Xin Tong; Dan Wu; Xue Wang; Huali Chen; Jia-Xiang Chen; Xiao-Xiao Wang; Xu-Lei Wang; Lu Gan; Zhi-Yun Guo; Gui-Xiu Shi; Yi-Zheng Zhang; Wei Jiang

Ghrelin is a multifunctional peptide that actively protects against cardiovascular ischemic diseases, but the underlying mechanisms are unclear. We used CoCl(2) to mimic hypoxic conditions in cardiac H9c2 cells in order to study the mechanism by which ghrelin protects cardiac myocytes against hypoxic injury by regulating the content of intracellular ROS and autophagy levels. Cell apoptosis and necrosis were evaluated by the flow cytometry assay, Hoechst staining, and LDH activity. Cell viability was detected by the WST-1 assay; ROS levels were assessed using DCFH2-DA; and Nox1, catalase and Mn-SOD were assayed by real-time PCR and activity assays. LC3II was measured by Western blot analysis. We observed that CoCl(2) induced apoptosis and death of H9c2 cells in a dose- and time-dependent manner. This was characterized by an increase in cell apoptosis, LDH activity, ROS content, Nox1 expression, and autophagy levels and a decrease in cell viability, catalase, and Mn-SOD activities. Ghrelin treatment significantly attenuated CoCl(2)-induced hypoxic injury by decreasing cell apoptosis, LDH activity, ROS content, and Nox1 expression and increasing cell viability, autophagy levels, catalase, and Mn-SOD mRNA levels and activities. Further experiments revealed that inhibiting autophagy using 3-MA or AMPK pathway with compound C almost abrogated the induction of ghrelin in autophagy. This was associated with a decrease in cell viability and an increase in LDH activity. Our results indicate that ghrelin protected cardiac myocytes against CoCl(2)-induced hypoxic injury by decreasing Nox1 expression, increasing the expression and activity of endogenous antioxidant enzymes, and inducing protective autophagy in an AMPK-dependent manner.

Collaboration


Dive into the Huali Chen'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

Rui Kuai

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Wenmin Yuan

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge