Network


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

Hotspot


Dive into the research topics where Jingjun Lu is active.

Publication


Featured researches published by Jingjun Lu.


The American Journal of the Medical Sciences | 2011

Oxidized Low-Density Lipoprotein and Atherosclerosis Implications in Antioxidant Therapy

Sona Mitra; Abhishek Deshmukh; Rajesh Sachdeva; Jingjun Lu; Jawahar L. Mehta

Low-density lipoprotein (LDL)-cholesterol is important for cellular function, but in high concentrations, it can lead to atheroma formation. Over the past several decades, it has become abundantly evident that the oxidized form of LDL-cholesterol (ox-LDL) is more important in the genesis and progression of atherosclerosis than native unmodified LDL-cholesterol. Ox-LDL leads to endothelial dysfunction, an initial step in the formation of an atheroma. Ox-LDL acts via binding to a number of scavenger receptors (SR), such as SR-A1, SR-A2 and lectin-like oxidized low-density lipoprotein receptor (LOX-1). Ox-LDL can upregulate expression of its own receptor LOX-1 on endothelial cells and activate these cells. In addition, ox-LDL promotes the growth and migration of smooth muscle cells, monocytes/macrophages and fibroblasts. Ox-LDL also leads to the generation of reactive oxygen species that in physiologic concentrations combat invasion of the body by noxious agents, but when in excess, can lead to a state of oxidative stress. There is evidence for the presence of oxidative stress in a host of conditions such as atherosclerosis and aging. In this review, we discuss the role of oxidative stress, ox-LDL and LOX-1 in atherogenesis and the reasons why the traditional approaches to limit oxidant stress have not been successful.


Antioxidants & Redox Signaling | 2011

Oxidative Stress and Lectin-Like Ox-LDL-Receptor LOX-1 in Atherogenesis and Tumorigenesis

Jingjun Lu; Sona Mitra; Xianwei Wang; Magomed Khaidakov; Jawahar L. Mehta

Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) has been identified as a major receptor for oxidized low-density lipoprotein (ox-LDL) in endothelial cells, monocytes, platelets, cardiomyocytes, and vascular smooth muscle cells. Its expression is minimal under physiological conditions but can be induced under pathological conditions. The upregulation of LOX-1 by ox-LDL appears to be important for physiologic processes, such as endothelial cell proliferation, apoptosis, and endothelium remodeling. Pathophysiologic effects of ox-LDL in atherogenesis have also been firmly established, including endothelial cell dysfunction, smooth muscle cell growth and migration, monocyte transformation into macrophages, and finally platelet aggregation-seen in atherogenesis. Recent studies show a positive correlation between increased serum ox-LDL levels and an increased risk of colon, breast, and ovarian cancer. As in atherosclerosis, ox-LDL and its receptor LOX-1 activate the inflammatory pathway through nuclear factor-kappa B, leading to cell transformation. LOX-1 is important for maintaining the transformed state in developmentally diverse cancer cell lines and for tumor growth, suggesting a molecular connection between atherogenesis and tumorigenesis.


American Journal of Physiology-heart and Circulatory Physiology | 2011

Prior exposure to oxidized low-density lipoprotein limits apoptosis in subsequent generations of endothelial cells by altering promoter methylation

Sona Mitra; Magomed Khaidakov; Jingjun Lu; Srinivas Ayyadevara; Jackob Szwedo; Xian Wei Wang; Chien Chen; Said Khaidakov; Srikanth Reddy Kasula; Annjanette Stone; Igor Pogribny; Jawahar L. Mehta

Oxidized LDL (ox-LDL) plays a critical role in atherogenesis, including apoptosis. As hypercholesterolemia causes epigenetic changes resulting in long-term phenotypic consequences, we hypothesized that repeated and continuous exposure to ox-LDL may alter the pattern of apoptosis in human umbilical vein endothelial cells (HUVECs). We also analyzed global and promoter-specific methylation of apoptosis-related genes. As expected, ox-LDL evoked a dose-dependent increase in apoptosis in the first passage HUVECs that was completely abrogated by lectin-like ox-LDL receptor (LOX-1)-neutralizing antibody. Ox-LDL-induced apoptosis was associated with upregulation of proapoptotic LOX-1, ANXA5, BAX, and CASP3 and inhibition of antiapoptotic BCL2 and cIAP-1 genes accompanied with reciprocal changes in the methylation of promoter regions of these genes. Subsequent passages of cells displayed attenuated apoptotic response to repeat ox-LDL challenge with blunted gene expression and exaggerated methylation of LOX-1, BAX, ANXA5, and CASP3 genes (all P < 0.05 vs. first exposure to ox-LDL). Treatment of cells with LOX-1 antibody before initial ox-LDL treatment prevented both gene-specific promoter methylation and expression changes and reduction of apoptotic response to repeat ox-LDL challenge. Based on these data, we conclude that exposure of HUVECs to ox-LDL induces epigenetic changes leading to resistance to apoptosis in subsequent generations and that this effect may be related to the LOX-1-mediated increase in DNA methylation.


Gene Therapy | 2012

LOX-1 abrogation reduces cardiac hypertrophy and collagen accumulation following chronic ischemia in the mouse

Jingjun Lu; Xianwei Wang; Wenze Wang; Harish Muniyappa; Chang-Ping Hu; Sona Mitra; B Long; Kumuda C. Das; Jawahar L. Mehta

We hypothesized that lectin-like oxidized LDL receptor-1 (LOX-1) deletion may inhibit oxidative stress signals, reduce collagen accumulation and attenuate cardiac remodeling after chronic ischemia. Activation of LOX-1 plays a significant role in the development of inflammation, apoptosis and collagen signals during acute ischemia. Wild-type and LOX-1 knockout (KO) mice were subjected to occlusion of left coronary artery for 3 weeks. Markers of cardiac hypertrophy, fibrosis-related signals (collagen IV, collagen-1 and fibronectin) and oxidant load (nicotinamide adenine dinucleotide phosphate oxidase expression, activity of mitogen-activated protein kinases and left ventricular (LV) tissue thiobarbituric acid reactive substances) were analyzed. In in vitro experiments, HL-1 cardiomyocytes were transfected with angiotensin II (Ang II) type 1 receptor (AT1R) or type 2 receptor (AT2R) genes to determine their role in the cardiomyocyte hypertrophy. LOX-1 KO mice had 25% improvement in survival over the 3-week period of chronic ischemia. LOX-1 deletion reduced collagen deposition and cardiomyocyte hypertrophy (∼75%) in association with a decrease in oxidant load and AT1R upregulation (all P<0.05). The LOX-1 KO mice hearts exhibited a disintegrin and metalloproteinase 10 (ADAM10) and a disintegrin and metalloproteinase 17 (ADAM17) expression and matrix metalloproteinase 2 activity, and increased AT2R expression (P<0.05). Attenuation of cardiac remodeling was associated with improved cardiac hemodynamics (LV ±dp/dt and cardiac ejection fraction). In vitro studies showed that it is AT1R, and not AT2R overexpression that induces cardiomyocyte hypertrophy. We demonstrate for the first time that LOX-1 deletion reduces oxidative stress and related intracellular signaling, which leads to attenuation of the positive feedback loop involving AT1R and LOX-1. This results in reduced chronic cardiac remodeling.


Toxicology and Applied Pharmacology | 2012

Aspirin suppresses cardiac fibroblast proliferation and collagen formation through downregulation of angiotensin type 1 receptor transcription

Xianwei Wang; Jingjun Lu; Magomed Khaidakov; Sona Mitra; Zufeng Ding; Sameer Raina; Tanu Goyal; Jawahar L. Mehta

Aspirin (acetyl salicylic acid, ASA) is a common drug used for its analgesic and antipyretic effects. Recent studies show that ASA not only blocks cyclooxygenase, but also inhibits NADPH oxidase and resultant reactive oxygen species (ROS) generation, a pathway that underlies pathogenesis of several ailments, including hypertension and tissue remodeling after injury. In these disease states, angiotensin II (Ang II) activates NADPH oxidase via its type 1 receptor (AT1R) and leads to fibroblast growth and collagen synthesis. In this study, we examined if ASA would inhibit NADPH oxidase activation, upregulation of AT1R transcription, and subsequent collagen generation in mouse cardiac fibroblasts challenged with Ang II. Mouse heart fibroblasts were isolated and treated with Ang II with or without ASA. As expected, Ang II induced AT1R expression, and stimulated cardiac fibroblast growth and collagen synthesis. The AT1R blocker losartan attenuated these effects of Ang II. Similarly to losartan, ASA, and its SA moiety suppressed Ang II-mediated AT1R transcription and fibroblast proliferation as well as expression of collagens and MMPs. ASA also suppressed the expression of NADPH oxidase subunits (p22(phox), p47(phox), p67(phox), NOX2 and NOX4) and ROS generation. ASA did not affect total NF-κB p65, but inhibited its phosphorylation and activation. These observations suggest that ASA inhibits Ang II-induced NADPH oxidase expression, NF-κB activation and AT1R transcription in cardiac fibroblasts, and fibroblast proliferation and collagen expression. The critical role of NADPH oxidase activity in stimulation of AT1R transcription became apparent in experiments where ASA also inhibited AT1R transcription in cardiac fibroblasts challenged with H2O2. Since SA had similar effect as ASA on AT1R expression, we suggest that ASAs effect is mediated by its SA moiety.


Cardiovascular Drugs and Therapy | 2011

LOX-1: A Critical Player in the Genesis and Progression of Myocardial Ischemia

Jingjun Lu; Jawahar L. Mehta

Myocardial ischemia is the most common cause of mortality and morbidity in the developed countries and rapidly becoming a common malady in the developing countries. Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), encoded by the OLR1 gene, is a scavenger receptor that plays a fundamental role in the genesis and progression of atherosclerosis and its complications. LOX-1 has been identified as a major receptor for oxidized low-density lipoprotein (ox-LDL) in endothelial cells, cardiomyocytes and fibroblast. In vitro and in vivo studies show that LOX-1 is upregulated during acute myocardial ischemia, and continues to be upregulated during chronic ischemia. Further, LOX-1 inhibition reduces ischemic myocardial injury and limits cardiac remodeling. LOX-1 inhibition decreases oxidative stress and inflammatory response to injury resulting in limitation of ischemic injury. Molecular studies show that LOX-1 inhibition reduces release of pro-inflammatory cytokines and expression of angiotensin II type 1 receptor via inhibition of redox-sensitive pathways. These alterations limit cardiomyocyte hypertrophy and collagen accumulation in the ischemic regions. These alterations in molecular signaling and physical alterations can result in improved cardiac function and better survival after ischemic myocardial injury.


Hypertension | 2012

Lectin-like Oxidized Low-density Lipoprotein Receptor-1 (LOX-1) and Cardiac Fibroblast Growth

Xianwei Wang; Magomed Khaidakov; Zufeng Ding; Sona Mitra; Jingjun Lu; Yao Dai; Jawahar L. Mehta

Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) regulates growth of a variety of cells and is important in inflammation, oxidative stress, and tissue remodeling. Recent studies show that LOX-1 deletion limits cardiac remodeling after sustained hypertension. We posited that LOX-1 may affect cardiac fibroblast growth and collagen secretion. To examine this postulate, we studied growth pattern of cardiac fibroblasts from hearts of wild-type and LOX-1 knockout (KO) mice. LOX-1 KO fibroblasts exhibited dramatically reduced growth when compared with wild-type mice fibroblasts and became much larger than wild-type mice fibroblasts in serial cultures, suggesting arrest of cell division. Western blotting and immunofluorescence showed that cell division control protein 42, a key regulator for cell division, was markedly downregulated in LOX-1 KO fibroblasts. The cytoskeletal organization in these fibroblasts was significantly altered in strand orientation, and some fibroblasts were completely devoid of F-actin. Furthermore, NADPH oxidase expression and generation of reactive oxygen species, as well as cell proliferation signals serine/threonine-specific protein kinase and murine double minute 2, were significantly reduced in LOX-1 KO fibroblasts. To confirm the essential role of LOX-1 in fibroblast growth, LOX-1 KO fibroblasts were transfected with h-LOX-1 cDNA. After transfection, the altered pattern of cytoskeletal organization, as well as expression of cell division control protein 42, serine/threonine-specific protein kinase, and murine double minute 2, was normalized. In congruent with these in vitro data, we found that the cardiac fibroblast number and expression of fibronectin and procoallagen-1/collagen were significantly lower in hypertensive LOX-1 KO mice hearts than in hypertensive wild-type mice hearts subjected to sustained hypertension (angiotensin II infusion). These findings implicate LOX-1 in cytoskeletal organization and growth of cardiac fibroblasts.


Experimental Biology and Medicine | 2012

Cross-talk between inflammation and angiotensin II: studies based on direct transfection of cardiomyocytes with AT1R and AT2R cDNA.

Xianwei Wang; Magomed Khaidakov; Zufeng Ding; Sona Mitra; Jingjun Lu; Shijie Liu; Jawahar L. Mehta

Ischemic myocardium exhibits inflammation, local angiotensin II (Ang II) generation and up-regulation of LOX-1, a lectin-like ox-LDL receptor. To define the inter-active roles of Ang II and inflammation in furthering tissue injury, cultured HL-1 cardiomyocytes were treated with Ang II. Ang II treatment up-regulated the expression of Ang II type 1 (AT1R) and type 2 (AT2R) receptors as well as LOX-1. Ang II also activated p44/42MAPK, p38MAPK, c-Jun and NF-kB, and increased the expression of inflammation-related genes (interleukins-6, interleukins-10, tumor necrosis factor-a, intercellular adhesion molecule-1). To study how inflammation per se might affect expression of Ang II receptors and LOX-1, cultured, cardiomyocytes were treated with lipopolysaccharide (LPS). Like Ang II, LPS increased the expression of AT1R, AT2R and LOX-1. LPS also activated mitogen-acticated protein kinase (MAPKs), c-Jun and NF-kB, and pro-inflammatory genes. The selective inhibitors of MAPKs, c-Jun and NF-kB each blocked the transcription of LOX-1 and pro-inflammatory genes in response to Ang II as well as LPS. These observations suggested a positive feedback between Ang II and inflammation. To delineate the role of AT1R and AT2R in LOX-1 expression, another set of cardiomyocytes were transfected with AT1R or AT2R cDNA. Forced over-expression of AT1R resulted in activation of MAPKs, c-Jun and NF-kB, up-regulation of inflammatory genes and LOX-1; on the other hand forced AT2R over-expression induced up-regulation of pro-apoptotic signals (pro-IL-1b and IL-1b), and decreased LOX-1 expression. These studies show that both Ang II and inflammation mediator LPS up-regulate AT1R, AT2R and LOX-1 expression. Up-regulation of AT1R promotes inflammation and LOX-1 expression, whereas up-regulation of AT2R promotes apoptosis signals and decreases LOX-1 expression.


Kidney International | 2012

Abrogation of lectin-like oxidized LDL receptor-1 attenuates acute myocardial ischemia-induced renal dysfunction by modulating systemic and local inflammation.

Jingjun Lu; Xianwei Wang; Wenze Wang; Harish Muniyappa; Abhishek Deshmukh; Chang-Ping Hu; Kumuda C. Das; Jawahar L. Mehta


Journal of Cardiovascular Pharmacology | 2010

Antiangiogenic and antimitotic effects of aspirin in hypoxia–reoxygenation modulation of the LOX-1-NADPH oxidase axis as a potential mechanism.

Magomed Khaidakov; Jacob Szwedo; Sona Mitra; Srinivas Ayyadevara; Maxim Dobretsov; Jingjun Lu; Jawahar L. Mehta

Collaboration


Dive into the Jingjun Lu's collaboration.

Top Co-Authors

Avatar

Jawahar L. Mehta

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Sona Mitra

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Xianwei Wang

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Magomed Khaidakov

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Kumuda C. Das

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Wenze Wang

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Zufeng Ding

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Chang-Ping Hu

Central South University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Harish Muniyappa

University of Arkansas for Medical Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge