Network


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

Hotspot


Dive into the research topics where Hanjun Guan is active.

Publication


Featured researches published by Hanjun Guan.


Journal of Molecular Neuroscience | 2004

Neprilysin regulates amyloid Beta peptide levels.

Robert A. Marr; Hanjun Guan; Edward Rockenstein; Mark S. Kindy; Fred H. Gage; Inder M. Verma; Eliezer Masliah; Louis B. Hersh

That neprilysin (NEP) is a major Aβ peptide-degrading enzyme in vivo is shown by higher Aβ peptide levels in the brain of an NEP knockout mouse. In addition, we show that infusion of an NEP inhibitor, but not inhibitors of other peptidases, into the brains of an APP transgenic mouse elevates Aβ levels. We have investigated the use of NEP as a potential therapeutic agent to prevent the accumulation of Aβ peptides in the brain. Lentivirus expressing NEP was initially used to demonstrate the ability of the enzyme to reduce Aβ levels in a model CHO cell line and to make primary hippocampal neurons resistant to Aβ-mediated neurotoxicity. Injection of NEP-expressing lentivirus, but not inactive NEP-expressing lentivirus, GFP-expressing lentivirus, or vehicle, into the hippocampus of 12–20-mo-old hAPP transgenic mice led to an approx 50% reduction in the number of amyloid plaques. These studies provide the impetus for further investigating of the use of NEP in a gene transfer therapy paradigm to prevent the accumulation of Aβ and prevent or delay the onset of Alzheimer’s disease.


Journal of Neuroscience Research | 2009

Peripherally Expressed Neprilysin Reduces Brain Amyloid Burden : A Novel Approach for Treating Alzheimer's Disease

Hanjun Guan; Yinxing Liu; Abigail Daily; Myung Hee Kim; Salvatore Oddo; Frank M. LaFerla; James R. Pauly; M. Paul Murphy; Louis B. Hersh

A number of therapeutic strategies for treating Alzheimers disease have focused on reducing amyloid burden in the brain. Among these approaches, the expression of amyloid β peptide (Aβ)‐degrading enzymes in the brain has been shown to be effective but to date not practical for treating patients. We report here a novel strategy for lowering amyloid burden in the brain by peripherally expressing the Aβ‐degrading enzyme neprilysin on leukocytes in the 3xTg‐AD mouse model of Alzheimers disease. Through transplantation of lentivirus‐transduced bone marrow cells, the Aβ‐degrading protease neprilysin was expressed on the surface of leukocytes. This peripheral neprilysin reduced soluble brain Aβ peptide levels by ∼30% and lowered the accumulation of amyloid β peptides by 50–60% when transplantation was performed at both young and early adult age. In addition, peripheral neprilysin expression reduced amyloid‐dependent performance deficits as measured by the Morris water maze. Unlike other methods designed to lower Aβ levels in blood, which cause a net increase in peptide, neprilysin expression results in the catabolism of Aβ to small, innocuous peptide fragments. These findings demonstrate that peripherally expressed neprilysin, and likely other Aβ‐degrading enzymes, has the potential to be utilized as a therapeutic approach to prevent and treat Alzheimers disease and suggest that this approach should be explored further.


Molecular Therapy | 2009

Expression of neprilysin in skeletal muscle reduces amyloid burden in a transgenic mouse model of Alzheimer disease.

Yinxing Liu; Christa M. Studzinski; Tina L. Beckett; Hanjun Guan; Matthew Hersh; M. Paul Murphy; Ronald L. Klein; Louis B. Hersh

Neprilysin (NEP) is a zinc metallopeptidase that efficiently degrades the amyloid beta (Abeta) peptides believed to be involved in the etiology of Alzheimer disease (AD). The focus of this study was to develop a new and tractable therapeutic approach for treating AD using NEP gene therapy. We have introduced adeno-associated virus (AAV) expressing the mouse NEP gene into the hindlimb muscle of 6-month-old human amyloid precursor protein (hAPP) (3X-Tg-AD) mice, an age which correlates with early stage AD. Overexpression of NEP in muscle decreased brain soluble Abeta peptide levels by approximately 60% and decreased amyloid deposits by approximately 50%, with no apparent adverse effects. Expression of NEP on muscle did not affect the levels of a number of other physiological peptides known to be in vitro substrates. These findings demonstrate that peripheral expression of NEP and likely other peptidases represents an alternative to direct administration into brain and illustrates the potential for using NEP expression in muscle for the prevention and treatment of AD.


Neuroscience Letters | 2004

Estrogen regulates neprilysin activity in rat brain.

Jian Huang; Hanjun Guan; Rosemarie M. Booze; Christopher B. Eckman; Louis B. Hersh

Neprilysin is a zinc metalloendopeptidase that regulates the activity of a number of physiological peptides through hydrolytic inactivation. Most recently, evidence has accumulated that neprilysin is involved in the clearance of amyloid beta peptides in the brain. Previous studies have shown that the neprilysin gene responds to progesterone, androgen, and glucocorticoids. We now show that estrogen regulates neprilysin activity in rat brain. Ovariectomy leads to a 30% decrease in neprilysin activity at 45 or 85 days, but not 21 days, post surgery. Estrogen replacement restores neprilysin levels back to control values. These changes in neprilysin activity suggest that in women estrogen is required to maintain basal levels of neprilysin.


Peptides | 2007

In vitro and in vivo degradation of Aβ peptide by peptidases coupled to erythrocytes

Yinxing Liu; Hanjun Guan; Tina L. Beckett; Maria A. Juliano; Luiz Juliano; Eun Suk Song; K. Martin Chow; M. Paul Murphy; Louis B. Hersh

It is generally believed that amyloid beta peptides (Abeta) are the key mediators of Alzheimers disease. Therapeutic interventions have been directed toward impairing the synthesis or accelerating the clearance of Abeta. An equilibrium between blood and brain Abeta exists mediated by carriers that transport Abeta across the blood-brain barrier. Passive immunotherapy has been shown to be effective in mouse models of AD, where the plasma borne antibody binds plasma Abeta causing an efflux of Abeta from the brain. As an alternative to passive immunotherapy we have considered the use of Abeta-degrading peptidases to lower plasma Abeta levels. Here we compare the ability of three Abeta-degrading peptidases to degrade Abeta. Biotinylated peptidases were coupled to the surface of biotinylated erythrocytes via streptavidin. These erythrocyte-bound peptidases degrade Abeta peptide in plasma. Thus, peptidases bound to or expressed on the surface of erythroid cells represent an alternative to passive immunotherapy.


Molecular Neurodegeneration | 2010

Aminopeptidases do not directly degrade tau protein.

K. Martin Chow; Hanjun Guan; Louis B. Hersh

BackgroundTau hyperphosphorylation and aggregation to form intracellular neurofibrillar tangles is prevalent in a number of tauopathies. Thus there is current interest in the mechanisms involved in Tau clearance. It was recently reported that Tau can be degraded by an aminopeptidase known as the puromycin sensitive aminopeptidase (PSA). Until now PSA has been reported to only cleave peptides, with the largest reported substrates having 30-50 amino acids. We have studied this unique PSA cleavage reaction using a number of different PSA preparations.ResultsAn N-terminally His tagged-PSA was expressed and purified from Sf9 insect cells. Although this PSA preparation cleaved Tau, product analysis with N and C terminal Tau antibodies coupled with mass spectrometry showed an endoproteolytic cleavage atypical for an aminopeptidase. Furthermore, the reaction was not blocked by the general aminopeptidase inhibitor bestatin or the specific PSA inhibitor puromycin. In order to test whether Tau hydrolysis might be caused by a protease contaminant the enzyme was expressed in E. coli as glutathione S-transferase and maltose binding protein fusion proteins or in Sf9 cells as a C-terminally His-tagged protein. After purification to near homogeneity none of these other recombinant forms of PSA cleaved Tau. Further, Tau-cleaving activity and aminopeptidase activities derived from the Sf9 cell expression system were separable by molecular sieve chromatography. When tested in a cellular context we again failed to see a PSA dependent cleavage of Tau. A commercial preparation of a related aminopeptidase, aminopeptidase N, also exhibited Tau cleaving activity, but this activity could also be separated from aminopeptidase activity.ConclusionIt is concluded that PSA does not directly cleave Tau.


Prostate Cancer and Prostatic Diseases | 2008

Neutral endopeptidase inhibits prostate cancer tumorigenesis by reducing FGF-2-mediated angiogenesis.

David Y.T. Chen; Oscar B. Goodman; Rong Zheng; Ruoqian Shen; Hanjun Guan; Louis B. Hersh; David M. Nanus

Neutral endopeptidase (NEP) is a cell surface peptidase that catalytically inactivates a variety of physiologically active peptides including basic fibroblast growth factor (FGF-2). We investigated the effect of using lentivirus to overexpress NEP in NEP-deficient DU145 prostate cancer cells. Third-generation lentiviral vectors encoding wild-type NEP (L-NEP), catalytically inactive mutant NEP (L-NEPmu), and green fluorescent protein (L-GFP) were stably introduced into DU145 cells. FGF-2 levels in cell culture supernatants decreased by 80% in L-NEP-infected DU145 cells compared to cells infected with L-NEPmu or L-GFP (P<0.05) while levels of other angiogenic factors were not altered. In vitro tubulogenesis of human vascular endothelial cells induced by conditioned media from DU145 cells infected with L-NEP was significantly reduced compared with that from DU145 cells infected with L-GFP (P<0.05). Tumor xenografts from L-NEP-infected DU145 cells were significantly smaller compared to control cell xenografts and vascularity within these tumors was decreased (P<0.05). Our data suggest that stable expression of NEP in DU145 cells inhibits prostate cancer tumorigenicity by inhibiting angiogenesis, with a probable mechanism being proteolytic inactivation of FGF-2.


Diabetologia | 2012

Degradation of islet amyloid polypeptide by neprilysin.

Hanjun Guan; K. M. Chow; R. Shah; Christopher J. Rhodes; Louis B. Hersh

Aims/hypothesisA progressive loss of pancreatic beta cell function, a decrease in beta cell mass and accumulation of islet amyloid is characteristic of type 2 diabetes mellitus. The main constituent of islet amyloid is islet amyloid polypeptide (IAPP). In this study, we examined the ability of the peptidase neprilysin to cleave IAPP and prevent human IAPP-induced pancreatic beta cell toxicity.MethodsNeprilysin and a catalytically compromised neprilysin mutant were tested for their ability to inhibit human IAPP fibrillisation and human IAPP-induced pancreatic beta cell cytotoxicity. Degradation of human IAPP by neprilysin was followed by HPLC, and the degradation products were identified by MS.ResultsNeprilysin prevented IAPP fibrillisation by cleaving IAPP at Arg11-Leu12, Leu12-Ala13, Asn14-Phe15, Phe15-Leu16, Asn22-Phe23 and Ala25-Ile26. It also appears to prevent human IAPP fibrillisation through a non-catalytic interaction. Neprilysin protected against beta cell cytotoxicity induced by exogenously added or endogenously produced human IAPP.Conclusions/interpretationThe data presented support a potential therapeutic role for neprilysin in preventing type 2 diabetes mellitus. This study supports the hypothesis that extracellular human IAPP contributes to human IAPP-induced beta cell cytotoxicity. Whether human IAPP exerts its cytotoxic effect through a totally extracellular mechanism or through a cellular reuptake mechanism is unclear at this time.


Cancer Gene Therapy | 2007

Lentiviral vector neutral endopeptidase gene transfer suppresses prostate cancer tumor growth

Rong Zheng; Oscar B. Goodman; Ruoqian Shen; Hanjun Guan; Louis B. Hersh; David M. Nanus

Neprilysin (neutral endopeptidase, NEP) is a cell surface peptidase whose expression is lost in approximately 50% of prostate cancers (PC). NEP normally functions to inactivate peptides such as bombesin and endothelin-1, and potentiates the effects of the PTEN tumor suppressor via a direct protein–protein interaction. NEP loss contributes to PC progression. We investigated the therapeutic efficacy of using a lentiviral vector system to restore NEP expression in PC cells. Third-generation lentiviral vectors encoding wild-type NEP (L-NEP) or green fluorescent protein (L-GFP) were introduced into NEP-deficient 22RV1 PC cells. Cells infected with L-NEP or L-GFP at a multiplicity of infection of 10 demonstrated NEP enzyme activity of 1171.2±4.9 and 17.2±5.3 pmol/μg/min (P<0.0001), respectively. Cell viability, proliferation and invasion were each significantly inhibited in 22RV1 cells expressing NEP compared with control cells infected with L-GFP (P<0.01). Analysis of known downstream effects of NEP showed NEP-expressing cells exhibiting decreased Akt and focal adhesion kinase phosphorylation and increased PTEN protein expression. Finally, injection of L-NEP into established 22RV1 xenograft tumors significantly inhibited tumor growth (P<0.01). These experiments demonstrate that lentiviral NEP gene transfer is a novel targeted strategy for the treatment of NEP-deficient PC.


PLOS ONE | 2015

The Mitochondrial Peptidase Pitrilysin Degrades Islet Amyloid Polypeptide in Beta-Cells

Hanjun Guan; K. Martin Chow; Eun-Suk Song; Nirmal Verma; Florin Despa; Louis B. Hersh

Amyloid formation and mitochondrial dysfunction are characteristics of type 2 diabetes. The major peptide constituent of the amyloid deposits in type 2 diabetes is islet amyloid polypeptide (IAPP). In this study, we found that pitrilysin, a zinc metallopeptidase of the inverzincin family, degrades monomeric, but not oligomeric, islet amyloid polypeptide in vitro. In insulinoma cells when pitrilysin expression was decreased to 5% of normal levels, there was a 60% increase in islet amyloid polypeptide-induced apoptosis. In contrast, overexpression of pitrilysin protects insulinoma cells from human islet amyloid polypeptide-induced apoptosis. Since pitrilysin is a mitochondrial protein, we used immunofluorescence staining of pancreases from human IAPP transgenic mice and Western blot analysis of IAPP in isolated mitochondria from insulinoma cells to provide evidence for a putative intramitochondrial pool of IAPP. These results suggest that pitrilysin regulates islet amyloid polypeptide in beta cells and suggest the presence of an intramitochondrial pool of islet amyloid polypeptide involved in beta-cell apoptosis.

Collaboration


Dive into the Hanjun Guan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yinxing Liu

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge