Network


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

Hotspot


Dive into the research topics where Hui-fang Zhou is active.

Publication


Featured researches published by Hui-fang Zhou.


Circulation | 2009

Complement-Dependent Neutrophil Recruitment Is Critical for the Development of Elastase-Induced Abdominal Aortic Aneurysm

Monica B. Pagano; Hui-fang Zhou; Terri L. Ennis; Xiaobo Wu; John D. Lambris; John P. Atkinson; Robert W. Thompson; Dennis E. Hourcade; Christine T. N. Pham

Background— We previously established that neutrophils play a critical role in the development of experimental abdominal aortic aneurysm (AAA). The signal that initiates the influx of neutrophils to the aortic wall, however, remains unknown. In this study, we tested the hypothesis that complement participates in the development of AAA by providing the necessary chemotactic signal that recruits neutrophils to the aortic wall. Methods and Results— Using an elastase-induced model of AAA, we showed that pretreatment of C57BL/6 mice with cobra venom factor, which depleted serum of complement activity, protected mice from AAA development. Whereas control mice exhibited a mean aortic diameter of 156±2% on day 14 after elastase perfusion, mice treated with cobra venom factor exhibited a mean aortic diameter of 90±4% (P<0.001). Examination of mice deficient in factor B further indicated that the alternative pathway of complement played a major role in this process (mean aortic diameter of 105±4% in factor B–deficient mice, P<0.001 compared with controls). Activation of the alternative pathway led to generation of the anaphylatoxins C3a and C5a, which recruited neutrophils to the aortic wall. Moreover, antagonism of both C3a and C5a activity was required to block AAA, which suggests that each can independently promote the aneurysmal phenotype. In addition, we demonstrated that complement alternative-pathway involvement was not restricted to this experimental model but was also evident in human AAAs. Conclusions— The identification of involvement of the complement system in the pathophysiology of AAA provides a new target for therapeutic intervention in this common disease.


The FASEB Journal | 2009

αvβ3-Targeted nanotherapy suppresses inflammatory arthritis in mice

Hui-fang Zhou; Happy W. Chan; Samuel A. Wickline; Gregory M. Lanza; Christine T. N. Pham

The purpose of this study was to assess whether an alternative treatment approach that targets angiogenesis, delivered through ligand‐targeted nanotherapy, would ameliorate inflammatory arthritis. Arthritis was induced using the K/BxN mouse model of inflammatory arthritis. After arthritis was clearly established, mice received three consecutive daily doses of αvβ3–targeted fumagillin nanoparticles. Control groups received no treatment or αvβ3–targeted nanoparticles without drugs. Disease score and paw thickness were measured daily. Mice that received αvβ3–targeted fumagillin nanoparticles showed a significantly lower disease activity score (mean score of 1.4±0.4;P<0.001) and change in ankle thickness (mean increase of 0.17±0.05 mm;P<0.001) 7 d after arthritis induction, whereas the group that received αvβ3–targeted nanoparticles without drugs exhibited a mean arthritic score of 9.0 ± 0.3 and mean change in ankle thickness of 1.01 ± 0.09 mm. Meanwhile, the group that received no treatment showed a mean arthritic score of 9.8 ± 0.5 and mean change in ankle thickness of 1.05 ± 0.10 mm. Synovial tissues from animals treated with targeted fumagillin nanoparticles also showed significant decrease in inflammation and angiogenesis and preserved proteoglycan integrity. Ligand‐targeted nanotherapy to deliver antiangiogenic agents may represent an effective way to treat inflammatory arthritis.—Zhou, H.‐F., Chan, H. W., Wickline, S. A., Lanza, G. M., Pham, C. T. N. αvβ3–Targeted nanotherapy suppresses inflammatory arthritis in mice. FASEB J. 23, 2978–2985 (2009). www.fasebj.org


Proceedings of the National Academy of Sciences of the United States of America | 2012

Antibody directs properdin-dependent activation of the complement alternative pathway in a mouse model of abdominal aortic aneurysm.

Hui-fang Zhou; Huimin Yan; Cordula M. Stover; Fernandez Tm; Rodriguez de Cordoba S; Wen-Chao Song; Xiaobo Wu; Robert W. Thompson; Wilhelm J. Schwaeble; John P. Atkinson; Dennis E. Hourcade; Christine T. N. Pham

Abdominal aortic aneurysm (AAA) is a complex inflammatory vascular disease. There are currently limited treatment options for AAA when surgery is inapplicable. Therefore, insights into molecular mechanisms underlying AAA pathogenesis may reveal therapeutic targets that could be manipulated pharmacologically or biologically to halt disease progression. Using an elastase-induced AAA mouse model, we previously established that the complement alternative pathway (AP) plays a critical role in the development of AAA. However, the mechanism by which complement AP is initiated remains undefined. The complement protein properdin, traditionally viewed as a positive regulator of the AP, may also initiate complement activation by binding directly to target surfaces. In this study, we sought to determine whether properdin serves as a focal point for the initiation of the AP complement activation in AAA. Using a properdin loss of function mutation in mice and a mutant form of the complement factor B protein that produces a stable, properdin-free AP C3 convertase, we show that properdin is required for the development of elastase-induced AAA in its primary role as a convertase stabilizer. Unexpectedly, we find that, in AAA, natural IgG antibodies direct AP-mediated complement activation. The absence of IgG abrogates C3 deposition in elastase-perfused aortic wall and protects animals from AAA development. We also determine that blockade of properdin activity prevents aneurysm formation. These results indicate that an innate immune response to self-antigens activates the complement system and initiates the inflammatory cascade in AAA. Moreover, the study suggests that properdin-targeting strategies may halt aneurysmal growth.


Journal of Clinical Investigation | 2014

Peptide-siRNA nanocomplexes targeting NF-κB subunit p65 suppress nascent experimental arthritis

Hui-fang Zhou; Huimin Yan; Hua Pan; Kirk K. Hou; Antonina Akk; Luke E. Springer; Ying Hu; J. Stacy Allen; Samuel A. Wickline; Christine T. N. Pham

The NF-κB signaling pathway is implicated in various inflammatory diseases, including rheumatoid arthritis (RA); therefore, inhibition of this pathway has the potential to ameliorate an array of inflammatory diseases. Given that NF-κB signaling is critical for many immune cell functions, systemic blockade of this pathway may lead to detrimental side effects. siRNAs coupled with a safe and effective delivery nanoplatform may afford the specificity lacking in systemic administration of small-molecule inhibitors. Here we demonstrated that a melittin-derived cationic amphipathic peptide combined with siRNA targeting the p65 subunit of NF-κB (p5RHH-p65) noncovalently self-assemble into stable nanocomplexes that home to the inflamed joints in a murine model of RA. Specifically, administration of p5RHH-p65 siRNA nanocomplexes abrogated inflammatory cytokine expression and cellular influx into the joints, protected against bone erosions, and preserved cartilage integrity. The p5RHH-p65 siRNA nanocomplexes potently suppressed early inflammatory arthritis without affecting p65 expression in off-target organs or eliciting a humoral response after serial injections. These data suggest that this self-assembling, largely nontoxic platform may have broad utility for the specific delivery of siRNA to target and limit inflammatory processes for the treatment of a variety of diseases.


Nanomedicine: Nanotechnology, Biology and Medicine | 2010

Synergistic effect of antiangiogenic nanotherapy combined with methotrexate in the treatment of experimental inflammatory arthritis

Hui-fang Zhou; Grace Hu; Samuel A. Wickline; Gregory M. Lanza; Christine T. N. Pham

AIM This study examines the effect of combining the antiangiogenic effect of αvß₃-targeted fumagillin nanoparticles with the conventional antirheumatic drug methotrexate for the treatment of inflammatory arthritis. METHOD Arthritis was induced in mice by K/BxN serum transfer, and disease activity was monitored by clinical score and change in ankle thickness. Groups of mice received nanoparticles or methotrexate as single therapy or nanoparticles and methotrexate as combination therapy. RESULTS We found that animals treated with a pulse dose of fumagillin nanoparticles followed by methotrexate had significantly improved and sustained clinical response compared with those treated with either agent alone. Histological analysis confirmed a significant decrease in inflammatory cell influx, bone erosions, cartilage damage and angiogenesis with the combination therapy. CONCLUSION Analysis of plasma cytokine levels suggests that fumagillin nanoparticles enhanced the systemic anti-inflammatory effects of methotrexate. Antiangiogenic nanotherapy may represent a promising approach for the treatment of inflammatory arthritis when combined with a conventional antirheumatic drug.


Journal of Immunology | 2013

CD43-Mediated IFN-γ Production by CD8+ T Cells Promotes Abdominal Aortic Aneurysm in Mice

Hui-fang Zhou; Huimin Yan; Luke E. Springer; Jonathan M. Green; Christine T. N. Pham

CD43 is a glycosylated surface protein abundantly expressed on lymphocytes. Its role in immune responses has been difficult to clearly establish, with evidence supporting both costimulatory and inhibitory functions. In addition, its contribution to disease pathogenesis remains elusive. Using a well-characterized murine model of elastase-induced abdominal aortic aneurysm (AAA) that recapitulates many key features of the human disease, we established that the presence of CD43 on T cells is required for AAA formation. Moreover, we found that IFN-γ–producing CD8+ T cells, but not CD4+ T cells, promote the development of aneurysm by enhancing cellular apoptosis and matrix metalloprotease activity. Reconstitution with IFN-γ–producing CD8+ T cells or recombinant IFN-γ promotes the aneurysm phenotype in CD43−/− mice, whereas IFN-γ antagonism abrogates disease in wild-type animals. Furthermore, we showed that the presence of CD43 with an intact cytoplasmic domain capable of binding to ezrin-radixin-moesin cytoskeletal proteins is essential for optimal in vivo IFN-γ production by T cells and aneurysm formation. We have thus identified a robust physiologic role for CD43 in a relevant animal model and established an important in vivo function for CD43-dependent regulation of IFN-γ production. These results further suggest that IFN-γ antagonism or selective blockade of CD43+CD8+ T cell activities merits further investigation for immunotherapy in AAA.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Fibrinogen-specific antibody induces abdominal aortic aneurysm in mice through complement lectin pathway activation.

Hui-fang Zhou; Huimin Yan; Paula Bertram; Ying Hu; Luke E. Springer; Robert W. Thompson; John A. Curci; Dennis E. Hourcade; Christine T. N. Pham

Significance Abdominal aortic aneurysm (AAA) is a common and potentially fatal vascular disease. Although surgery is recommended for large aneurysms, surgical repair offers no clear survival advantage for small AAAs. A deeper understanding of the mechanisms that underlie disease pathogenesis could suggest strategies for medical intervention. Herein we identify a natural mouse IgG antibody that recognizes fibrinogen deposited in human AAA tissues and induces aneurysm in a mouse model through activation of the complement lectin pathway (LP). Conversely, absence of antibody or complement proteins abrogates AAA development. These results support the concept that antibody directed against aortic wall epitopes initiates LP activation that culminates in AAA formation. The antibody response and complement LP may provide therapeutic targets for halting disease progression. Abdominal aortic aneurysm (AAA) is a common vascular disease associated with high mortality rate due to progressive enlargement and eventual rupture. There is currently no established therapy known to alter the rate of aneurysmal expansion. Thus, understanding the processes that initiate and sustain aneurysmal growth is pivotal for the development of medical therapies aimed at halting disease progression. Using an elastase-induced AAA mouse model that recapitulates key features of human AAA, we previously reported that a natural IgG antibody directs alternative pathway complement activation and initiates the inflammatory process that culminates in aneurysmal development. The target of this natural antibody, however, was unknown. Herein we identify a natural IgG that binds to fibrinogen deposited in elastase-perfused aortic tissues, activates the complement lectin pathway (LP), and induces AAA. Moreover, we establish that alterations in the glycosylation patterns of this antibody critically affect its ability to activate the LP in vivo. We find that LP activation precedes the alternative pathway and absence of the LP complement protein mannan-binding lectin abrogates elastase-induced AAA. In human AAA tissues the mouse anti-fibrinogen antibody recognizes epitopes that localize to the same areas that stain positively for mannan-binding lectin, which suggests that the complement LP is engaged in humans as well. Lastly, we demonstrate that circulating antibodies in a subset of AAA patients react against fibrinogen or fibrinogen-associated epitopes in human aneurysmal tissues. Our findings support the concept that an autoimmune process directed at aortic wall self-antigens may play a central role in the immunopathogenesis of AAA.


Molecular Immunology | 2016

Anti-complement activity of the Ixodes scapularis salivary protein Salp20.

Dennis E. Hourcade; Antonina Akk; Lynne M. Mitchell; Hui-fang Zhou; Richard E. Hauhart; Christine T. N. Pham

Complement, a major component of innate immunity, presents a rapid and robust defense of the intravascular space. While regulatory proteins protect host cells from complement attack, when these measures fail, unrestrained complement activation may trigger self-tissue injury, leading to pathologic conditions. Of the three complement activation pathways, the alternative pathway (AP) in particular has been implicated in numerous disease and injury states. Consequently, the AP components represent attractive targets for therapeutic intervention. The common hard-bodied ticks from the family Ixodidae derive nourishment from the blood of their mammalian hosts. During its blood meal the tick is exposed to host immune effectors, including the complement system. In defense, the tick produces salivary proteins that can inhibit host immune functions. The Salp20 salivary protein of Ixodes scapularis inhibits the host AP pathway by binding properdin and dissociating C3bBbP, the active C3 convertase. In these studies we examined Salp20 activity in various complement-mediated pathologies. Our results indicate that Salp20 can inhibit AP-dependent pathogenesis in the mouse. Its efficacy may be part in due to synergic effects it provides with the endogenous AP regulator, factor H. While Salp20 itself would be expected to be highly immunogenic and therefore inappropriate for therapeutic use, its emergence speaks for the potential development of a non-immunogenic Salp20 mimic that replicates its anti-properdin activity.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2016

Neutrophil Proteases Promote Experimental Abdominal Aortic Aneurysm via Extracellular Trap Release and Plasmacytoid Dendritic Cell Activation

Huimin Yan; Hui-fang Zhou; Antonina Akk; Ying Hu; Luke E. Springer; Terri L. Ennis; Christine T. N. Pham

Objective—We previously established that neutrophil-derived dipeptidyl peptidase I (DPPI) is essential for experimental abdominal aortic aneurysm (AAA) development. Because DPPI activates several neutrophil serine proteases, it remains to be determined whether the AAA-promoting effect of DPPI is mediated by neutrophil serine proteases. Approach and Results—Using an elastase-induced AAA model, we demonstrate that the absence of 2 neutrophil serine proteases, neutrophil elastase and proteinase-3, recapitulates the AAA-resistant phenotype of DPPI-deficient mice. DPPI and neutrophil serine proteases direct the in vitro and in vivo release of extracellular structures termed neutrophil extracellular traps (NETs). Administration of DNase1, which dismantles NETs, suppresses elastase-induced AAA in wild-type animals and in DPPI-deficient mice reconstituted with wild-type neutrophils. NETs also contain the cathelicidin-related antimicrobial peptide that complexes with self-DNA in recruiting plasmacytoid dendritic cells (pDCs), inducing type I interferons (IFNs) and promoting AAA in DPPI-deficient mice. Conversely, depletion of pDCs or blockade of type I IFNs suppresses experimental AAA. Moreover, we find an abundance of human cathelicidin peptide, a 37 amino acid sequence starting with 2 leucines and the human orthologue of cathelicidin-related antimicrobial peptide, in the vicinity of pDCs in human AAA tissues. Increased type I IFN mRNA expression is observed in human AAA tissues and circulating IFN-&agr; is detected in ≈50% of the AAA sera examined. Conclusions—These results suggest that neutrophil protease–mediated NET release contributes to elastase-induced AAA through pDC activation and type I IFN production. These findings increase our understanding of the pathways underlying AAA inflammatory responses and suggest that limiting NET, pDC, and type I IFN activities may suppress aneurysm progression.


Journal of Rheumatic Diseases and Treatment | 2015

Suppression of Experimental Arthritis through AMP-Activated Protein Kinase Activation and Autophagy Modulation

Huimin Yan; Hui-fang Zhou; Ying Hu; Christine T. N. Pham

Autophagy plays a central role in various disease processes. However, its contribution to inflammatory arthritides such as rheumatoid arthritis (RA) is unclear. We observed that autophagy is engaged in the K/BxN serum transfer model of RA but autophagic flux is severely impaired. Metformin is an anti-diabetic drug that has been shown to stimulate autophagy. Induction of autophagic flux, through metformin-mediated AMP-activated protein kinase (AMPK) activation and interruption of mammalian target of rapamycin (mTOR) signaling mitigated the inflammation in experimental arthritis. Further investigation into the effects of metformin suggest that the drug directly activates AMPK and dose-dependently suppressed the release of TNF-α, IL-6, and MCP-1 by macrophages while enhancing the release of IL-10 in vitro. In vivo, metformin treatment significantly suppressed clinical arthritis and inflammatory cytokine production. Mechanistic studies suggest that metformin exerts its anti-inflammatory effects by correcting the impaired autophagic flux observed in the K/BxN arthritis model and suppressing NF-κB-mediated signaling through selective degradation of IκB kinase (IKK). These findings establish a central role for autophagy in inflammatory arthritis and argue that autophagy modulators such as metformin may represent potential therapeutic agents for the treatment of RA.

Collaboration


Dive into the Hui-fang Zhou's collaboration.

Top Co-Authors

Avatar

Christine T. N. Pham

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Huimin Yan

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Dennis E. Hourcade

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Antonina Akk

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Luke E. Springer

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Ying Hu

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Robert W. Thompson

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Samuel A. Wickline

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Gregory M. Lanza

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

John P. Atkinson

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge