Network


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

Hotspot


Dive into the research topics where Xinbing Han is active.

Publication


Featured researches published by Xinbing Han.


The FASEB Journal | 2010

Interleukin-10 overexpression in macrophages suppresses atherosclerosis in hyperlipidemic mice

Xinbing Han; Shiro Kitamoto; Hong-Wei Wang; William A. Boisvert

In atherogenesis, macrophage foam cell formation is modulated by pathways involving both the uptake and efflux of cholesterol. We recently showed that interleukin-10 (IL-10) modulates lipid metabolism by enhancing both uptake and efflux of cholesterol in macrophages. However, the mechanistic details of these properties in vivo have been unclear. Thus, the purpose of this study was to determine whether expression of IL-10 in macrophages would alter susceptibility to atherosclerosis and whether IL-10 exerts its antiatherosclerotic properties by modulating lipid metabolism in macrophages. We utilized a macrophage-specific retroviral vector that allows long-term in vivo expression of IL-10 in macrophages through transplantation of retrovirally transduced bone marrow cells (BMCs). IL-10 expressed by macrophages derived from transduced BMCs inhibited atherosclerosis in LDLR(-/-) mice by reducing cholesteryl ester accumulation in atherosclerotic sites. Experiments with primary macrophages indicated that macrophage source of IL-10 stimulated both the uptake (by up-regulating scavenger receptors) and efflux of cholesterol (by activating the PPARgamma-LXR-ABCA1/ABCG1 pathway), thereby reducing inflammation and apoptosis in atherosclerosis. These findings indicate that BMC-transduced macrophage IL-10 production can act as a strong antiatherogenic agent, and they highlight a novel antiatherosclerotic therapy using a simple, yet effective, stem cell transduction system that facilitates long-term expression of IL-10 in macrophages.


Journal of Biological Chemistry | 2009

Interleukin-10 Facilitates Both Cholesterol Uptake and Efflux in Macrophages

Xinbing Han; Shiro Kitamoto; Qingyu Lian; William A. Boisvert

Foam cell formation is a hallmark event during atherosclerosis. The current paradigm is that lipid uptake by scavenger receptor in macrophages initiates the chronic proinflammatory cascade and necrosis core formation that characterize atherosclerosis. We report here that a cytokine considered to be anti-atherogenic, interleukin-10 (IL10), promotes cholesterol uptake from modified lipoproteins in macrophages and its transformation into foam cells by increasing the expression of scavenger receptor CD36 and scavenger receptor A. Although uptake of modified lipoproteins is considered proatherogenic, we found that IL10 also increases cholesterol efflux from macrophages to protect against toxicity of free cholesterol accumulation in the cell. This process was PPARγ-dependent and was mediated through up-regulation of ABCA1 (ATP-binding cassette transporter A1) protein expression. Importantly, expression of inflammatory molecules, such as tumor necrosis factor-α, intercellular adhesion molecule-1, and MMP9 as well as apoptosis were dramatically suppressed in lipid-laden foam cells treated with IL10. The notion that IL10 can mediate both the uptake of cholesterol from modified lipoproteins and the efflux of stored cholesterol suggests that the process of foam cell formation is not necessarily detrimental as long as mechanisms of cholesterol efflux and transfer to an exogenous acceptor are functioning robustly. Our results present a comprehensive antiatherogenic role of IL10 in macrophages, including enhanced disposal of harmful lipoproteins, inhibition of inflammatory molecules, and reduced apoptosis.


Journal of Biological Chemistry | 2012

Epigenetic Regulation of Tumor Necrosis Factor α (TNFα) Release in Human Macrophages by HIV-1 Single-stranded RNA (ssRNA) Is Dependent on TLR8 Signaling

Xinbing Han; Xin Li; Simon Yue; Asha Anandaiah; Falah Hashem; Peter S. Reinach; Henry Koziel; Souvenir D. Tachado

Background: TLR7/TLR8 recognize viral ssRNA, but in human macrophages recognition of HIV-1 ssRNA is not known. Results: In human macrophages, HIV-1 ssRNA promotes TNFα release and requires endocytosis through TLR8, MyD88, and histone modification. Conclusion: TLR8 recognizes HIV-1 ssRNA and promotes TNFα release through chromatin remodeling. Significance: Targeting macrophage TLR8 signaling may serve as a novel therapeutic strategy to modify HIV infection. Human macrophages at mucosal sites are essential targets for acute HIV infection. During the chronic phase of infection, they are persistent reservoirs for the AIDS virus. HIV virions gain entry into macrophages following ligation of surface CD4-CCR5 co-receptors, which leads to the release of two copies of HIV ssRNA. These events lead to reverse transcription and viral replication initiation. Toll-like receptors TLR7 and TLR8 recognize specific intracellular viral ssRNA sequences, but in human alveolar macrophages, their individual roles in TLR-mediated HIV ssRNA recognition are unclear. In the current study, HIV-1 ssRNA induced TNFα release in a dose-dependent manner in adherent human macrophages expressing both intracellular TLR7 and TLR8. This response was reduced by inhibiting either endocytosis (50 μm dynasore) or endosomal acidification (1 μg/ml chloroquine). Either MYD88 or TLR8 gene knockdown with relevant siRNA reduced HIV-1 ssRNA-mediated TNFα release, but silencing TLR7 had no effect on this response. Furthermore, HIV-1 ssRNA induced histone 4 acetylation at the TNFα promoter as well as trimethylation of histone 3 at lysine 4, whereas TLR8 gene knockdown reduced these effects. Taken together in human macrophages, TLR8 binds and internalizes HIV ssRNA, leading to endosomal acidification, chromatin remodeling, and increases in TNFα release. Drugs targeting macrophage TLR8-linked signaling pathways may modulate the innate immune response to acute HIV infection by reducing viral replication.


Infection and Immunity | 2013

Vitamin D Rescues Impaired Mycobacterium tuberculosis-Mediated Tumor Necrosis Factor Release in Macrophages of HIV-Seropositive Individuals through an Enhanced Toll-Like Receptor Signaling Pathway In Vitro

Asha Anandaiah; Sanjeev Sinha; Medhavi Bole; Surendra Sharma; Narendra Kumar; Kalpana Luthra; Xin Li; Xiuqin Zhou; Benjamin Nelson; Xinbing Han; Souvenir D. Tachado; Naimish R. Patel; Henry Koziel

ABSTRACT Mycobacterium tuberculosis disease represents an enormous global health problem, with exceptionally high morbidity and mortality in HIV-seropositive (HIV+) persons. Alveolar macrophages from HIV+ persons demonstrate specific and targeted impairment of critical host cell responses, including impaired M. tuberculosis-mediated tumor necrosis factor (TNF) release and macrophage apoptosis. Vitamin D may promote anti-M. tuberculosis responses through upregulation of macrophage NO, NADPH oxidase, cathelicidin, and autophagy mechanisms, but whether vitamin D promotes anti-M. tuberculosis mechanisms in HIV+ macrophages is not known. In the current study, human macrophages exposed to M. tuberculosis demonstrated robust release of TNF, IκB degradation, and NF-κB nuclear translocation, and these responses were independent of vitamin D pretreatment. In marked contrast, HIV+ U1 human macrophages exposed to M. tuberculosis demonstrated very low TNF release and no significant IκB degradation or NF-κB nuclear translocation, whereas vitamin D pretreatment restored these critical responses. The vitamin D-mediated restored responses were dependent in part on macrophage CD14 expression. Importantly, similar response patterns were observed with clinically relevant human alveolar macrophages from healthy individuals and asymptomatic HIV+ persons at high clinical risk of M. tuberculosis infection. Taken together with the observation that local bronchoalveolar lavage fluid (BALF) levels of vitamin D are severely deficient in HIV+ persons, the data from this study demonstrate that exogenous vitamin D can selectively rescue impaired critical innate immune responses in vitro in alveolar macrophages from HIV+ persons at risk for M. tuberculosis disease, supporting a potential role for exogenous vitamin D as a therapeutic adjuvant in M. tuberculosis infection in HIV+ persons.


American Journal of Pathology | 2013

Chitinase Inhibition Promotes Atherosclerosis in Hyperlipidemic Mice

Shiro Kitamoto; Kensuke Egashira; Toshihiro Ichiki; Xinbing Han; Sara McCurdy; Shohei Sakuda; Kenji Sunagawa; William A. Boisvert

Chitinase 1 (CHIT1) is secreted by activated macrophages. Chitinase activity is raised in atherosclerotic patient sera and is present in atherosclerotic plaque. However, the role of CHIT1 in atherosclerosis is unknown. Preliminary studies of atherosclerosis in cynomolgous monkeys revealed CHIT1 to be closely correlated with areas of macrophage infiltration. Thus, we investigated the effects of a chitinase inhibitor, allosamidin, on macrophage function in vitro and on atherosclerotic development in vivo. In RAW264.7 cells, allosamidin elevated monocyte chemoattractant protein 1 and tumor necrosis factor alpha expression, and increased activator protein 1 and nuclear factor-κB transcriptional activity. Although inducible nitric oxide synthase, IL-6, and IL-1β expression were increased, Arg1 expression was decreased by chitinase inhibition, suggesting that suppression of CHIT1 activity polarizes macrophages into a M1 phenotype. Allosamidin decreased scavenger receptor AI, CD36, ABCA1, and ABCG1 expression which led to suppression of cholesterol uptake and apolipoprotein AI-mediated cholesterol efflux in macrophages. These effects were confirmed with CHIT1 siRNA transfection and CHIT1 plasmid transfection experiments in primary macrophages. Apolipoprotein E-deficient hyperlipidemic mice treated for 6 weeks with constant administration of allosamidin and fed an atherogenic diet showed aggravated atherosclerotic lesion formation. These data suggest that CHIT1 exerts protective effects against atherosclerosis by suppressing inflammatory responses and polarizing macrophages toward an M2 phenotype, and promoting lipid uptake and cholesterol efflux in macrophages.


Journal of Immunology | 2011

Mammalian Target of Rapamycin Inhibition in Macrophages of Asymptomatic HIV+ Persons Reverses the Decrease in TLR-4–Mediated TNF-α Release through Prolongation of MAPK Pathway Activation

Xin Li; Xinbing Han; Juliana Llano; Medhavi Bole; Xiuqin Zhou; Katharine Swan; Asha Anandaiah; Benjamin Nelson; Naimish R. Patel; Peter S. Reinach; Henry Koziel; Souvenir D. Tachado

TLR-4–mediated signaling is significantly impaired in macrophages from HIV+ persons, predominantly owing to altered MyD88-dependent pathway signaling caused in part by constitutive activation of PI3K. In this study we assessed in these macrophages if the blunted increase in TLR-4–mediated TNF-α release induced by lipid A (LA) is associated with PI3K-induced upregulation of mammalian target of rapamycin (mTOR) activity. mTOR inhibition with rapamycin enhanced TLR-4–mediated TNF-α release, but suppressed anti-inflammatory IL-10 release. Targeted gene silencing of mTOR in macrophages resulted in LA-induced TNF-α and IL-10 release patterns similar to those induced by rapamycin. Rapamycin restored MyD88/IL-1R–associated kinase interaction in a dose-dependent manner. Targeted gene silencing of MyD88 (short hairpin RNA) and mTOR (RNA interference) inhibition resulted in TLR-4–mediated 70-kDa ribosomal protein S6 kinase activation and enhanced TNF-α release, whereas IL-10 release was inhibited in both silenced and nonsilenced HIV+ macrophages. Furthermore, mTOR inhibition augmented LA-induced TNF-α release through enhanced and prolonged phosphorylation of ERK1/2 and JNK1/2 MAPK, which was associated with time-dependent MKP-1 destabilization. Taken together, impaired TLR-4–mediated TNF-α release in HIV+ macrophages is attributable in part to mTOR activation by constitutive PI3K expression in a MyD88-dependent signaling pathway. These changes result in MAPK phosphatase 1 stabilization, which shortens and blunts MAPK activation. mTOR inhibition may serve as a potential therapeutic target to upregulate macrophage innate immune host defense responsiveness in HIV+ persons.


PLOS ONE | 2014

HIV-derived ssRNA binds to TLR8 to induce inflammation-driven macrophage foam cell formation.

Mark A. Bernard; Xinbing Han; Sonya Inderbitzin; Ifunanya Agbim; Hui Zhao; Henry Koziel; Souvenir D. Tachado

Even though combined anti-retroviral therapy (cART) dramatically improves patient survival, they remain at a higher risk of being afflicted with non-infectious complications such as cardiovascular disease (CVD). This increased risk is linked to persistent inflammation and chronic immune activation. In this study, we assessed whether this complication is related to HIV-derived ssRNAs inducing in macrophages increases in TNFα release through TLR8 activation leading to foam cell formation. HIV ssRNAs induced foam cell formation in monocyte-derived macrophages (MDMs) in a dose-dependent manner. This response was reduced when either endocytosis or endosomal acidification was inhibited by dynasore or chloroquine, respectively. Using a flow cytometry FRET assay, we demonstrated that ssRNAs bind to TLR8 in HEK cells. In MDMs, ssRNAs triggered a TLR8-mediated inflammatory response that ultimately lead to foam cell formation. Targeted silencing of the TLR8 and MYD88 genes reduced foam cell formation. Furthermore, foam cell formation induced by these ssRNAs was blocked by an anti-TNFα neutralizing antibody. Taken together in MDMs, HIV ssRNAs are internalized; bind TLR8 in the endosome followed by endosomal acidification. TLR8 signaling then triggers TNFα release and ultimately leads to foam cell formation. As this response was inhibited by a blocking anti-TNFα antibody, drug targeting HIV ssRNA-driven TLR8 activation may serve as a potential therapeutic target to reduce chronic immune activation and inflammation leading to CVD in HIV+ patients.


PLOS ONE | 2012

\(Leu128^{3.43}\) (L128) and \(Val247^{6.40}\) (V247) of CXCR1 Are Critical Amino Acid Residues for G Protein Coupling and Receptor Activation

Xinbing Han; Souvenir D. Tachado; Henryk Koziel; William A. Boisvert

CXCR1, a classic GPCR that binds IL-8, plays a key role in neutrophil activation and migration by activating phospholipase C (PLC)β through Gα15 and Gαi which generates diacylglycerol and inositol phosphates (IPs). In this study, two conserved amino acid residues of CXCR1 on the transmembrane domain (TM) 3 and TM6, Leu1283.43 (L128) and Val2476.40 (V247), respectively, were selectively substituted with other amino acids to investigate the role of these conserved residues in CXCR1 activation. Although two selective mutants on Leu128, Leu128Ala (L128A) and Leu128Arg (L128R), demonstrated high binding affinity to IL-8, they were not capable of coupling to G proteins and consequently lost the functional response of the receptors. By contrast, among the four mutants at residue Val247 (TM6.40), replacing Val247 with Ala (V247A) and Asn (V247N) led to constitutive activation of mutant receptors when cotransfected with Gα15. The V247N mutant also constitutively activated the Gαi protein. These results indicate that L128 on TM3.43 is involved in G protein coupling and receptor activation but is unimportant for ligand binding. On the other hand, V247 on TM6.40 plays a critical role in maintaining the receptor in the inactive state, and the substitution of V247 impaired the receptor constraint and stabilized an active conformation. Functionally, there was an increase in chemotaxis in response to IL-8 in cells expressing V247A and V247N. Our findings indicate that Leu1283.43 and Val2476.40 are critical for G protein coupling and activation of signaling effectors, providing a valuable insight into the mechanism of CXCR1 activation.


Archive | 2012

The Role of IL-10 in Atherosclerosis

Xinbing Han; William A. Boisvert

Cardiovascular diseases, including coronary artery disease (CAD), ischemic gangrene, abdominal aortic aneurysms, and many cases of heart failure and stroke currently account for the most number of deaths in the Western world (Hansson et al., 2006). The root cause of these diseases is atherosclerosis, which is widely accepted these days to be a chronic inflammatory disease in addition to the more recognized disorder of lipid metabolism. Although it was established long ago that high levels of low-density lipoprotein (LDL) cholesterol is a major risk factor for atherosclerosis, more recently both innate and adaptive immune systems have been accepted as major participants in the initiation and progression of atherosclerosis. Besides monocytes/macrophages, T cells and dendritic cells (DCs) can be detected within atherosclerotic lesions and have been implicated in the pathogenesis of atherosclerosis (Hansson and Libby, 2006) (Weber et al., 2008). Atherosclerotic lesion progression has been shown to depend on ongoing, chronic inflammation in the artery wall. Following hyperlipidemia, a rapid influx of circulating monocytes into the atherosclerosis-prone areas of the arterial intima occurs. These recruited inflammatory monocytes differentiate into macrophages and take up modified atherogenic cholesteryl ester (CE)rich lipoproteins in the intima of the vessel wall (Lusis, 2000) (Ross, 1999) (Wang and Tall, 2003). The accumulation of cholesterol-loaded macrophages in the arterial wall called “foam cells” is a key feature of early atherosclerotic lesions (Brown and Goldstein, 1983). Upon lipid uptake within the artery wall, macrophage foam cells activate a compensatory pathway for cholesterol efflux, mediated by the ATP-binding cassette (ABC) transporters ABCA1 and ABCG1 (Wang et al., 2007). During systemic hypercholesterolemia, however, this homeostatic mechanism is overwhelmed, leading to the accumulation of foam cells and the initiation of fatty streak lesions. The importance of these transporters is illustrated by the fact that a combined deficiency of ABCA1 and ABCG1 accelerates foam cell accumulation and atherosclerotic development in mice (Yvan-Charvet et al., 2007). Cholesterol loading of macrophages also stimulates the production of inflammatory mediators, which recruit other cell types and contribute to the development of a complex lesion (Hansson et al., 2002). Thus, processes that interfere with the intracellular cholesterol balance would be expected to exacerbate lesion formation.


Archive | 2011

Mammalian Target of Rapamycin Inhibition in Macrophages of Asymptomatic HIV + Persons Reverses the Decrease in TLR-4-Mediated TNF-a Release through Prolongation of MAPK Pathway Activation

Xin Li; Xinbing Han; Juliana Llano; Medhavi Bole; Xiuqin Zhou; Katharine Swan; Asha Anandaiah; Benjamin Nelson; Naimish R. Patel; Peter S. Reinach; Henry Koziel; Souvenir D. Tachado

Collaboration


Dive into the Xinbing Han's collaboration.

Top Co-Authors

Avatar

Souvenir D. Tachado

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Henry Koziel

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xin Li

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Medhavi Bole

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Naimish R. Patel

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Asha Anandaiah

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xiuqin Zhou

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge