Network


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

Hotspot


Dive into the research topics where Lemeng Zhang is active.

Publication


Featured researches published by Lemeng Zhang.


Hepatology | 2011

Endogenous histones function as alarmins in sterile inflammatory liver injury through Toll-like receptor 9 in mice.

Hai Huang; John Evankovich; Wei Yan; Gary W. Nace; Lemeng Zhang; Mark A. Ross; Xinghua Liao; Timothy R. Billiar; Jun Xu; Charles T. Esmon; Allan Tsung

Sterile inflammatory insults are known to activate innate immunity and propagate organ damage through the recognition of extracellular damage‐associated molecular pattern (DAMP) molecules. Although DAMPs such as endogenous DNA and nuclear high‐mobility group box 1 have been shown to be critical in sterile inflammation, the role of nuclear histone proteins has not yet been investigated. We report that endogenous histones function as DAMPs after ischemic injury through the pattern recognition receptor Toll‐like receptor (TLR) 9 to initiate inflammation. Using an in vivo model of hepatic ischemia/reperfusion (I/R) injury, we show that levels of circulating histones are significantly higher after I/R, and that histone neutralization significantly protects against injury. Injection of exogenous histones exacerbates I/R injury through cytotoxic effects mediated by TLR9 and MyD88. In addition, histone administration increases TLR9 activation, whereas neither TLR9 nor MyD88 mutant mice respond to exogenous histones. Furthermore, we demonstrate in vitro that extracellular histones enhance DNA‐mediated TLR9 activation in immune cells through a direct interaction. Conclusion: These novel findings reveal that histones represent a new class of DAMP molecules and serve as a crucial link between initial damage and activation of innate immunity during sterile inflammation. (HEPATOLOGY 2011; 54:999–1008)


Journal of Biological Chemistry | 2010

High Mobility Group Box 1 Release from Hepatocytes during Ischemia and Reperfusion Injury Is Mediated by Decreased Histone Deacetylase Activity

John Evankovich; Sung W. Cho; Ruilin Zhang; Jon Cardinal; Lemeng Zhang; John R. Klune; Jason Zlotnicki; Timothy R. Billiar; Allan Tsung

The mobilization and extracellular release of nuclear high mobility group box-1 (HMGB1) by ischemic cells activates inflammatory pathways following liver ischemia/reperfusion (I/R) injury. In immune cells such as macrophages, post-translational modification by acetylation appears to be critical for active HMGB1 release. Hyperacetylation shifts its equilibrium from a predominant nuclear location toward cytosolic accumulation and subsequent release. However, mechanisms governing its release by parenchymal cells such as hepatocytes are unknown. In this study, we found that serum HMGB1 released following liver I/R in vivo is acetylated, and that hepatocytes exposed to oxidative stress in vitro also released acetylated HMGB1. Histone deacetylases (HDACs) are a family of enzymes that remove acetyl groups and control the acetylation status of histones and various intracellular proteins. Levels of acetylated HMGB1 increased with a concomitant decrease in total nuclear HDAC activity, suggesting that suppression in HDAC activity contributes to the increase in acetylated HMGB1 release after oxidative stress in hepatocytes. We identified the isoforms HDAC1 and HDAC4 as critical in regulating acetylated HMGB1 release. Activation of HDAC1 was decreased in the nucleus of hepatocytes undergoing oxidative stress. In addition, HDAC1 knockdown with siRNA promoted HMGB1 translocation and release. Furthermore, we demonstrate that HDAC4 is shuttled from the nucleus to cytoplasm in response to oxidative stress, resulting in decreased HDAC activity in the nucleus. Together, these findings suggest that decreased nuclear HDAC1 and HDAC4 activities in hepatocytes following liver I/R is a mechanism that promotes the hyperacetylation and subsequent release of HMGB1.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2012

Calcium/calmodulin-dependent protein kinase IV limits organ damage in hepatic ischemia-reperfusion injury through induction of autophagy

John Evankovich; Ruilin Zhang; Jon Cardinal; Lemeng Zhang; Junda Chen; Hai Huang; Donna Beer-Stolz; Timothy R. Billiar; Matthew R. Rosengart; Allan Tsung

Sterile inflammatory insults, such as ischemia-reperfusion (I/R) injury, result from pathogenic factors, including damage-associated molecular pattern signaling, activation of innate immunity, and upregulation of proinflammatory cytokines. At the same time, a number of protective, or prosurvival, pathways are also activated, and the extent of end-organ damage is ultimately determined by the balance between these two systems. In liver I/R, members of the calcium/calmodulin-dependent protein kinase (CaMK) family are known to be activated, but their individual roles are largely unknown. In this study, we show that one CaMK member, CaMKIV, is protective in hepatic I/R by activating the prosurvival pathway of autophagy in hepatocytes. CaMKIV knockout mice experience significantly worse organ damage after I/R and are deficient in hepatocyte autophagic signaling. Restoration of autophagic signaling with rapamycin reduces organ damage in CaMKIV knockout mice to wild-type levels. In vitro, we show that CaMKIV activation induces autophagy in mouse hepatocytes, and that CaMKIV activation protects hepatocytes from oxidative stress-induced cell death. In conclusion, the protective autophagic signaling pathway serves to reduce organ damage following I/R and is regulated by activation of CaMKIV signaling in hepatocytes.


Molecular Medicine | 2012

Interferon Regulatory Factor-1 Regulates the Autophagic Response in LPS-Stimulated Macrophages through Nitric Oxide

Lemeng Zhang; Jon Cardinal; Runalia Bahar; John Evankovich; Hai Huang; Gary W. Nace; Timothy R. Billiar; Matthew R. Rosengart; Pinhua Pan; Allan Tsung

The pathogenesis of sepsis is complex and, unfortunately, poorly understood. The cellular process of autophagy is believed to play a protective role in sepsis; however, the mechanisms responsible for its regulation in this setting are ill defined. In the present study, interferon regulatory factor 1 (IRF-1) was found to regulate the autophagic response in lipopolysaccharide (LPS)-stimulated macrophages. In vivo, tissue macrophages obtained from LPS-stimulated IRF-1 knockout (KO) mice demonstrated increased autophagy and decreased apoptosis compared to those isolated from IRF-1 wild-type (WT) mice. In vitro, LPS-stimulated peritoneal macrophages obtained from IRF-1 KO mice experienced increased autophagy and decreased apoptosis. IRF-1 mediates the inhibition of autophagy by modulating the activation of the mammalian target of rapamycin (mTOR). LPS induced the activation of mTOR in WT peritoneal macrophages, but not in IRF-1 KO macrophages. In contrast, overexpression of IRF-1 alone increased the activation of mTOR and consequently decreased autophagic flux. Furthermore, the inhibitory effects of IRF-1 mTOR activity were mediated by nitric oxide (NO). Therefore, we propose a novel role for IRF-1 and NO in the regulation of macrophage autophagy during LPS stimulation in which IRF-1/NO inhibits autophagy through mTOR activation.


Shock | 2012

SPLENOCYTE APOPTOSIS AND AUTOPHAGY IS MEDIATED BY INTERFERON REGULATORY FACTOR-1 DURING MURINE ENDOTOXEMIA

Lemeng Zhang; Jon Cardinal; Pinhua Pan; Brian R. Rosborough; Ying Chang; Wei Yan; Hai Huang; Timothy R. Billiar; Matthew R. Rosengart; Allan Tsung

Sepsis-induced lymphocyte and dendritic cell apoptosis contributes to immunosuppression, which results in an inability to eradicate the primary infection as well as a propensity to acquire new, secondary infections. Another cellular process, autophagy, is also activated in immune cells and plays a protective role. In the present study, we demonstrate that interferon regulatory factor 1 (IRF-1) regulates both immune cell apoptosis and autophagy in a murine endotoxemia model. Interferon regulatory factor 1 is activated at an early phase through a Toll-like receptor 4–dependent, myeloid differentiation primary response gene 88–independent manner in splenocytes. Furthermore, IRF-1 knockout (KO) mice are protected from a lethal endotoxemia model. This protection is associated with decreased apoptosis and increased autophagy in splenocytes. Interferon regulatory factor 1 KO mice experience decreased apoptotic cell loss, especially in CD4+ T lymphocytes and myeloid antigen-presenting cells. Meanwhile, IRF-1 KO mice demonstrate increased autophagy and improved mitochondrial integrity. This increased autophagy in KO mice is attributable, at least in part, to deactivation of mammalian target of rapamycin/P70S6 signaling—a main negative regulator of autophagy. Therefore, we propose a novel role for IRF-1 in regulating both apoptosis and autophagy in splenocytes in the setting of endotoxemia with IRF-1 promoting apoptosis and inhibiting autophagy. ABBREVIATION LPS—lipopolysaccharide; TLR4—Toll-like receptor 4; MyD88—myeloid differentiation primary response gene 88; TRIF—domain-containing adaptor inducing IFN-B; IRF-1—interferon regulatory factor 1; LC3—microtubule-associated protein 1 light chain 3; mTOR—mammalian target of rapamycin; WT—wild type; KO—knockout; APCs—antigen-presenting cells


Scientific Reports | 2016

Neutrophil extracellular traps are indirectly triggered by lipopolysaccharide and contribute to acute lung injury.

Shuai Liu; Xiaoli Su; Pinhua Pan; Lemeng Zhang; Yongbin Hu; Hongyi Tan; Dongdong Wu; Ben Liu; Haitao Li; Haosi Li; Yi Li; Minhui Dai; Yuanyuan Li; Chengping Hu; Allan Tsung

Neutrophil extracellular traps (NETs) facilitate the extracellular killing of pathogens. However, excessive NETs formation and poor degradation are associated with exacerbated immune responses and tissue injury. In this study, we investigated the role of NETs in lipopolysaccharide (LPS)-mediated acute lung injury (ALI) and assessed the use of DNase I, for the treatment of ALI. Additionally, we focused on the controversial issue of whether LPS directly induces NETs release in vitro. NETs formation was detected in murine ALI tissue in vivo and was associated with increased NETs markers, citrullinated-histone H3 tissue levels and NET-DNA levels in BALF. Treatment with DNase I significantly degraded NETs and reduced citrullinated-histone H3 levels, which protected against ALI and ameliorated pulmonary oedema and total protein in BALF. In addition, DNase I significantly reduced IL-6 and TNF-α levels in plasma and BALF. In vitro, LPS-activated platelets rather than LPS alone efficiently induced NETs release. In conclusion, NETs formed during LPS-induced ALI, caused organ damage and initiated the inflammatory response. NETs degradation by DNase I promoted NET-protein clearance and protected against ALI in mice; thus, DNase I may be a new potential adjuvant for ALI therapy. Specifically, LPS induced NETs formation in an indirect manner via platelets activation.


Shock | 2016

Interferon Regulatory Factor-1 Mediates Alveolar Macrophage Pyroptosis During LPS-Induced Acute Lung Injury in Mice.

Dongdong Wu; Pinhua Pan; Xiaoli Su; Lemeng Zhang; Qingwu Qin; Hongyi Tan; Huang L; Yuanyuan Li

ABSTRACT Previously, we demonstrated that pyroptosis in alveolar macrophages (AMs) plays an essential role in lipopolysaccharide (LPS)-induced acute lung injury. However, the underlying mechanism remains largely unclear. Here, we show that the absence of interferon regulatory factor 1 (IRF-1) in genetic knock-out mice strongly abrogates pyroptosis in AMs and alleviates the LPS-induced lung injury and systemic inflammation. Our study demonstrates that IRF-1 contributes to caspase-1 activation and apoptosis-associated speck-like protein containing a caspase activation and recruitment domain pyroptosome formation in AMs and leads to downstream inflammatory cytokine release, including that of IL-1&bgr;, IL-18, and HMGB1. The nuclear translocation of IRF-1 is linked to the presence of toll-like receptor 4 (TLR4). Our findings suggest that pyroptosis and the downstream inflammatory response in AMs induced by LPS is a process that is dependent on TLR4-mediated up-regulation of IRF-1. In summary, IRF-1 plays a key role in controlling caspase-1-dependent pyroptosis and inflammation.


BioMed Research International | 2017

Neutrophil Extracellular Traps Are Pathogenic in Ventilator-Induced Lung Injury and Partially Dependent on TLR4

Haosi Li; Pinhua Pan; Xiaoli Su; Shuai Liu; Lemeng Zhang; Dongdong Wu; Haitao Li; Minhui Dai; Yi Li; Chengping Hu; Jie Chen

The pathogenesis of ventilator-induced lung injury (VILI) is associated with neutrophils. Neutrophils release neutrophil extracellular traps (NETs), which are composed of DNA and granular proteins. However, the role of NETs in VILI remains incompletely understood. Normal saline and deoxyribonuclease (DNase) were used to study the role of NETs in VILI. To further determine the role of Toll-like receptor 4 (TLR4) in NETosis, we evaluated the lung injury and NET formation in TLR4 knockout mice and wild-type mice that were mechanically ventilated. Some measures of lung injury and the NETs markers were significantly increased in the VILI group. DNase treatment markedly reduced NETs markers and lung injury. After high-tidal mechanical ventilation, the NETs markers in the TLR4 KO mice were significantly lower than in the WT mice. These data suggest that NETs are generated in VILI and pathogenic in a mouse model of VILI, and their formation is partially dependent on TLR4.


Autophagy: Cancer, Other Pathologies, Inflammation, Immunity, Infection, and Aging#R##N#Volume 12 | 2017

Chapter 8 – Role of the Transcription Factor Interferon Regulatory Factor-1 in Regulating Autophagy in Lipopolysaccharide-Stimulated Macrophages

Lemeng Zhang; Allan Tsung

Abstract The pathogenesis of sepsis is complex and unfortunately poorly understood. The cellular process of autophagy, especially in macrophages, is believed to have multiple important functions in sepsis. Our study demonstrated that, interferon regulatory factor-1 (IRF-1) could regulate the autophagic response in lipopolysaccharide (LPS)-stimulated macrophages. In vivo, tissue macrophages obtained from LPS-stimulated IRF-1 knockout (KO) mice demonstrated increased autophagy and decreased apoptosis compared to those isolated from IRF-1 wild-type (WT) mice. In vitro, LPS-stimulated peritoneal macrophages obtained from IRF-1 KO mice experienced increased autophagy and decreased apoptosis. IRF-1 mediates the inhibition of autophagy by modulating the activation of the mammalian target of rapamycin (mTOR). LPS induced the activation of mTOR in WT peritoneal macrophages, but not in IRF-1 KO macrophages. In contrast, overexpression of IRF-1 alone increased the activation of mTOR and consequently decreased autophagic flux. Furthermore, the inhibitory effects of IRF-1 mTOR activity were mediated by nitric oxide (NO). Therefore we propose a novel role for IRF-1 and NO in the regulation of macrophage autophagy during LPS stimulation, in which IRF-1/NO inhibits autophagy through mTOR activation.


Autophagy: Cancer, Other Pathologies, Inflammation, Immunity, Infection, and Aging#R##N#Volume 3 - Mitophagy | 2014

Interferon Regulatory Factor 1 Regulates both Autophagy and Apoptosis in Splenocytes during Sepsis

Lemeng Zhang; Allan Tsung

Sepsis-induced lymphocyte and dendritic cell (DC) apoptosis contributes to immunosuppression, which results in not only an inability to eradicate the primary infection, but also a propensity to acquire new, secondary infections. Another cellular process, autophagy, is also activated in immune cells, and in contrast, plays a protective role. Our previous results demonstrate that interferon regulatory factor 1 (IFR-1) regulates both splenocyte apoptosis and autophagy in a murine endotoxemia model. IRF-1 knockout (KO) mice are protected from a lethal endotoxemia model. This protection is associated with decreased apoptosis and increased autophagy in splenocytes. IRF-1 KO mice experience decreased apoptotic cell loss, especially in CD4+T lymphocytes and myeloid antigen-presenting cells. On the other hand, IRF-1 KO mice also demonstrate increased autophagy and improved mitochondrial integrity. Therefore, we proposed a novel role for IRF-1 in regulating both apoptosis and autophagy in splenocytes within the setting of endotoxemia with IRF-1 promoting apoptosis and inhibiting autophagy.

Collaboration


Dive into the Lemeng Zhang's collaboration.

Top Co-Authors

Avatar

Allan Tsung

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Hai Huang

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jon Cardinal

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pinhua Pan

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Ruilin Zhang

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Wei Yan

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Dongdong Wu

Central South University

View shared research outputs
Researchain Logo
Decentralizing Knowledge