Network


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

Hotspot


Dive into the research topics where Zhao Jun Liu is active.

Publication


Featured researches published by Zhao Jun Liu.


Journal of Clinical Investigation | 2007

Diabetic impairments in NO-mediated endothelial progenitor cell mobilization and homing are reversed by hyperoxia and SDF-1α

Katherine A. Gallagher; Zhao Jun Liu; Min Xiao; Haiying Chen; Lee J. Goldstein; Donald G. Buerk; April Nedeau; Stephen R. Thom; Omaida C. Velazquez

Endothelial progenitor cells (EPCs) are essential in vasculogenesis and wound healing, but their circulating and wound level numbers are decreased in diabetes. This study aimed to determine mechanisms responsible for the diabetic defect in circulating and wound EPCs. Since mobilization of BM EPCs occurs via eNOS activation, we hypothesized that eNOS activation is impaired in diabetes, which results in reduced EPC mobilization. Since hyperoxia activates NOS in other tissues, we investigated whether hyperoxia restores EPC mobilization in diabetic mice through BM NOS activation. Additionally, we studied the hypothesis that impaired EPC homing in diabetes is due to decreased wound level stromal cell-derived factor-1alpha (SDF-1alpha), a chemokine that mediates EPC recruitment in ischemia. Diabetic mice showed impaired phosphorylation of BM eNOS, decreased circulating EPCs, and diminished SDF-1alpha expression in cutaneous wounds. Hyperoxia increased BM NO and circulating EPCs, effects inhibited by the NOS inhibitor N-nitro-L-arginine-methyl ester. Administration of SDF-1alpha into wounds reversed the EPC homing impairment and, with hyperoxia, synergistically enhanced EPC mobilization, homing, and wound healing. Thus, hyperoxia reversed the diabetic defect in EPC mobilization, and SDF-1alpha reversed the diabetic defect in EPC homing. The targets identified, which we believe to be novel, can significantly advance the field of diabetic wound healing.


Molecular and Cellular Biology | 2003

Regulation of Notch1 and Dll4 by Vascular Endothelial Growth Factor in Arterial Endothelial Cells: Implications for Modulating Arteriogenesis and Angiogenesis

Zhao Jun Liu; Takashi Shirakawa; Yan Li; Akinobu Soma; Masahiro Oka; G. Paolo Dotto; Ronald M. Fairman; Omaida C. Velazquez; Meenhard Herlyn

ABSTRACT Notch and its ligands play critical roles in cell fate determination. Expression of Notch and ligand in vascular endothelium and defects in vascular phenotypes of targeted mutants in the Notch pathway have suggested a critical role for Notch signaling in vasculogenesis and angiogenesis. However, the angiogenic signaling that controls Notch and ligand gene expression is unknown. We show here that vascular endothelial growth factor (VEGF) but not basic fibroblast growth factor can induce gene expression of Notch1 and its ligand, Delta-like 4 (Dll4), in human arterial endothelial cells. The VEGF-induced specific signaling is mediated through VEGF receptors 1 and 2 and is transmitted via the phosphatidylinositol 3-kinase/Akt pathway but is independent of mitogen-activated protein kinase and Src tyrosine kinase. Constitutive activation of Notch signaling stabilizes network formation of endothelial cells on Matrigel and enhances formation of vessel-like structures in a three-dimensional angiogenesis model, whereas blocking Notch signaling can partially inhibit network formation. This study provides the first evidence for regulation of Notch/Delta gene expression by an angiogenic growth factor and insight into the critical role of Notch signaling in arteriogenesis and angiogenesis.


Journal of Cellular Biochemistry | 2009

Trafficking and differentiation of mesenchymal stem cells

Zhao Jun Liu; Ying Zhuge; Omaida C. Velazquez

Mesenchymal stem cells (MSCs) are a heterogeneous population of stem/progenitor cells with pluripotent capacity to differentiate into mesodermal and non‐mesodermal cell lineages, including osteocytes, adipocytes, chondrocytes, myocytes, cardiomyocytes, fibroblasts, myofibroblasts, epithelial cells, and neurons. MSCs reside primarily in the bone marrow, but also exist in other sites such as adipose tissue, peripheral blood, cord blood, liver, and fetal tissues. When stimulated by specific signals, these cells can be released from their niche in the bone marrow into circulation and recruited to the target tissues where they undergo in situ differentiation and contribute to tissue regeneration and homeostasis. Several characteristics of MSCs, such as the potential to differentiate into multiple lineages and the ability to be expanded ex vivo while retaining their original lineage differentiation commitment, make these cells very interesting targets for potential therapeutic use in regenerative medicine and tissue engineering. The feasibility for transplantation of primary or engineered MSCs as cell‐based therapy has been demonstrated. In this review, we summarize the current knowledge on the signals that control trafficking and differentiation of MSCs. J. Cell. Biochem. 106: 984–991, 2009.


Antioxidants & Redox Signaling | 2008

Hyperoxia, endothelial progenitor cell mobilization, and diabetic wound healing.

Zhao Jun Liu; Omaida C. Velazquez

Diabetic foot disease is a major health problem, which affects 15% of the 200 million patients with diabetes worldwide. Diminished peripheral blood flow and decreased local neovascularization are critical factors that contribute to the delayed or nonhealing wounds in these patients. The correction of impaired local angiogenesis may be a key component in developing therapeutic protocols for treating chronic wounds of the lower extremity and diabetic foot ulcers. Endothelial progenitor cells (EPCs) are the key cellular effectors of postnatal neovascularization and play a central role in wound healing, but their circulating and wound-level numbers are decreased in diabetes, implicating an abnormality in EPC mobilization and homing mechanisms. The deficiency in EPC mobilization is presumably due to impairment of eNOS-NO cascade in bone marrow (BM). Hyperoxia, induced by a clinically relevant hyperbaric oxygen therapy (HBO) protocol, can significantly enhance the mobilization of EPCs from the BM into peripheral blood. However, increased circulating EPCs failed to reach to wound tissues. This is partly a result of downregulated production of SDF-1alpha in local wound lesions with diabetes. Administration of exogenous SDF-1alpha into wounds reversed the EPC homing impairment and, with hyperoxia, synergistically enhanced EPC mobilization, homing, neovascularization, and wound healing.


Biochemical Pharmacology | 2010

Notch signaling: Emerging molecular targets for cancer therapy

Ling Yin; Omaida C. Velazquez; Zhao Jun Liu

The Notch signaling pathway is a highly conserved developmental pathway, which plays a critical role in cell-fate decision, tissue patterning and morphogenesis. There is increasing evidence that this pathway is dysregulated in a variety of malignancies, and can behave as either an oncogene or a tumor suppressor depending upon cell context. This review highlights the current evidence for aberration of the Notch signaling pathway in a wide range of tumors from hematological cancers, such as leukemia and lymphoma through to skin, breast, lung, pancreas, colon and brain tumors. It proposes that the Notch signaling pathway may represent novel therapeutic targets and will be a welcome asset to the cancer therapeutic arena.


The FASEB Journal | 2002

Fibroblast-dependent differentiation of human microvascular endothelial cells into capillary-like 3-dimensional networks

Omaida C. Velazquez; Ruthanne Snyder; Zhao Jun Liu; Ronald M. Fairman; Meenhard Herlyn

An in vitro model has been developed to study migration, survival, proliferation, and capillary‐like differentiation of human microvascular endothelial cells (HMVECs) in an environment that avoids tumor promoters and complex matrices. HMVEC monolayers were plated, then induced to form three‐dimensional, capillary‐like networks by overlaying with human type I collagen followed by a second overlay of collagen with embedded fibroblasts. Detachment and migration of endothelial cells into the matrix was triggered within hours by the overlaying collagen, and the fibroblasts stimulated survival and formation of cords, vacuoles, tubes, and, after 4 to 5 days, capillary networks. The differentiation into branching capillary‐like structures was dependent on direct fibroblast‐endothelial cell contact and was not achieved when fibroblasts were replaced by seven types of melanoma cells, which included radial and vertical growth phase primary and metastatic stages. Vascular endothelial growth factor (VEGF), when overexpressed in fibroblasts, stimulated endothelial cell proliferation and migration, whereas angiopoietin‐1 (Ang1) had only motogenic effects. Neutralizing antibodies against VEGF and blocking antibodies for VEGF‐receptor 2 (VEGFR2) significantly inhibited but not completely obliterated capillary network formation, suggesting that the VEGF signaling pathway is important but not exclusive and that other fibroblast‐derived soluble factors and fibroblast‐endothelial cell contact are essential for endothelial cell survival and differentiation.


Experimental Cell Research | 2008

A CXCL5- and bFGF-dependent effect of PDGF-B-activated fibroblasts in promoting trafficking and differentiation of bone marrow-derived mesenchymal stem cells.

April Nedeau; Richard J. Bauer; Katherine A. Gallagher; Haiying Chen; Zhao Jun Liu; Omaida C. Velazquez

Adult bone marrow-derived mesenchymal stem cells (MSCs) are able to differentiate into myofibroblasts and be recruited into wound lesions and contribute to wound healing. The cellular and molecular mechanisms responsible for MSC trafficking and differentiation, however, are poorly understood. Local resting resident fibroblasts are activated after injury and play a critical role in recruiting MSCs. We investigated the role of platelet-derived growth factor-B-activated fibroblasts (PDGF-B-aFBs) in regulating recruitment, migration and differentiation of MSCs from GFP transgenic mice in an in vitro wound healing assay and a novel three-dimensional (3D) model. PDGF-B-aFBs caused significant increases in MSC migration velocity compared to control as demonstrated by time-lapse photography in an in vitro wound healing assay. Consistently, invasion/migration of MSCs into 3D collagen gels was enhanced in the presence of PDGF-B-aFBs. In addition, PDGF-B-aFBs induced differentiation of MSCs into myofibroblast. The regulatory effects of PDGF-B-aFBs are likely to be mediated by basic fibroblast growth factor (bFGF) and epithelial neutrophil activating peptide-78 (ENA-78 or CXCL5) as protein array analysis indicated elevated levels of these two soluble factors in culture supernatant of PDGF-B-aFBs. Blocking antibodies against bFGF and CXCL5 were able to inhibit both trafficking and differentiation of MSCs into 3D collagen gels while supplement of exogenous bFGF and/or CXCL5 promoted invasion/migration of MSCs into 3D collagen gels. Our results reveal that PDGF-B-aFBs play a key role in the recruitment/migration and differentiation of MSCs and implicate a bFGF- and CXCL5-dependent mechanism in mediating these effects.


Atherosclerosis | 2012

Notch activation induces endothelial cell senescence and pro-inflammatory response: implication of Notch signaling in atherosclerosis.

Zhao Jun Liu; Yurong Tan; Gary W. Beecham; David Seo; Runxia Tian; Yan Li; Roberto I. Vazquez-Padron; Margaret A. Pericak-Vance; Jeffery M. Vance; Pascal J. Goldschmidt-Clermont; Alan S. Livingstone; Omaida C. Velazquez

OBJECTIVE Notch signaling plays pivotal roles in the pathogenesis of vascular disease. However, little is known about its role in atherosclerosis. We sought to investigate the potential involvement of the Notch signaling in atherosclerosis. METHODS Expression of Notch pathway components in mouse and human aorta with or without atherosclerosis plaque was examined by immunohistochemistry. Expression of Notch target genes in young versus aged human endothelial cells (EC) was examined by PCRArray and immunoblot. In vitro loss- and gain-of-function approaches were utilized to evaluate the role of Notch signaling in inducing EC senescence and secretion of pro-inflammatory cytokines by ProteinArray. Notch gene profile was studied in 1054 blood samples of patients with coronary artery disease (CAD). Genotyping was performed using the Genome-Wide Single Nucleotide Polymorphism (SNP) Array. RESULTS Notch pathway components were upregulated in luminal EC at atherosclerotic lesions from mouse and human aortas. In addition, the Notch pathway was activated in aged but not young human EC. Enforced Notch activation resulted in EC senescence and significantly upregulated expression of several molecules implicated in the inflammatory response (IL-6/IL-8/IL-1α/RANTES/ICAM-1). The upregulated IL-6 was partially responsible for mediating leukocyte transendothelial migration. Genetic association analysis detected, of 82 SNPs across 6 Notch pathway genes analyzed, 4 SNPs with nominal association with CAD burden. CONCLUSION Notch pathway is activated in luminal EC at atherosclerotic plaques and results in pro-inflammatory response and senescence of EC. Notch signaling may be linked to human CAD risk. These findings implicate a potential involvement of Notch signaling in atherosclerosis.


The FASEB Journal | 2003

VEGF-A and αVβ3 integrin synergistically rescue angiogenesis via N-Ras and PI3-K signaling in human microvascular endothelial cells

Zhao Jun Liu; Ruthanne Snyder; Akinobu Soma; Takashi Shirakawa; Barry L. Ziober; Ronald M. Fairman; Meenhard Herlyn; Omaida C. Velazquez

We recently showed that normal fibroblasts mediate capillary‐like differentiation of human microvascular endothelial cells (HMVEC) in a 3‐D angiogenesis model. Here, we show that a collaborative effect of VEGF‐A and αVβ3 integrin is critical in fibroblast‐mediated angiogenesis because enhancement of both VEGF production by fibroblasts and β3 integrin expression in HMVEC can rescue capillary‐like endothelial differentiation under reduced serum conditions. To investigate the downstream signaling mechanisms, we compared N‐Ras and Rho/Rac/Cdc42, as well as phosphatidylinositol 3‐kinase (PI3‐K) and Akt, for their involvement in the capillary‐like network formation. The dominant‐negative mutant of N‐Ras (N‐RasN17), but not the mutants of Rho/Rac/Cdc42, suppressed network formation. Overexpression of a constitutively active form of PI3‐K rescued the network formation, which was inhibited by a dominant‐negative β3 integrin; however, an active form of Akt failed to rescue the inhibition but induced a phenotypic change in HMVEC. Moreover, PI3‐K is a downstream target of N‐Ras because it could be co‐immunoprecipitated with N‐Ras, and its active form could rescue the inhibitory effect of N‐RasN17. Thus, our data indicate the existence of N‐Ras‐ and PI3‐K‐dependent but Rho/Rac/Cdc42‐and Akt‐independent signaling mechanisms for the synergistic effect of VEGF‐A and αVβ3 on fibroblast‐mediated microvascular network formation.


Advances in pharmacology | 2012

Targeting Notch Signaling for Cancer Therapeutic Intervention

Hongwei Shao; Qinghua Huang; Zhao Jun Liu

The Notch signaling pathway is an evolutionarily conserved, intercellular signaling cascade. The Notch proteins are single-pass receptors that are activated upon interaction with the Delta (or Delta-like) and Jagged/Serrate families of membrane-bound ligands. Association of ligand-receptor leads to proteolytic cleavages that liberate the Notch intracellular domain (NICD) from the plasma membrane. The NICD translocates to the nucleus, where it forms a complex with the DNA-binding protein CSL, displacing a histone deacetylase (HDAc)-corepressor (CoR) complex from CSL. Components of a transcriptional complex, such as MAML1 and histone acetyltransferases (HATs), are recruited to the NICD-CSL complex, leading to the transcriptional activation of Notch target genes. The Notch signaling pathway plays a critical role in cell fate decision, tissue patterning, morphogenesis, and is hence regarded as a developmental pathway. However, if this pathway goes awry, it contributes to cellular transformation and tumorigenesis. There is mounting evidence that this pathway is dysregulated in a variety of malignancies, and can behave as either an oncogene or a tumor suppressor depending upon cell context. This chapter highlights the current evidence for aberration of the Notch signaling pathway in a wide range of tumors from hematological cancers, such as leukemia and lymphoma, through to lung, skin, breast, pancreas, colon, prostate, ovarian, brain, and liver tumors. It proposes that the Notch signaling pathway may represent novel target for cancer therapeutic intervention.

Collaboration


Dive into the Zhao Jun Liu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yan Li

University of Miami

View shared research outputs
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