Network


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

Hotspot


Dive into the research topics where Guangjin Zhou is active.

Publication


Featured researches published by Guangjin Zhou.


Journal of Clinical Investigation | 2011

Krüppel-like factor 4 regulates macrophage polarization

Xudong Liao; Nikunj Sharma; Fehmida Kapadia; Guangjin Zhou; Yuan Lu; Hong Hong; Kaavya Paruchuri; Ganapati H. Mahabeleshwar; Elise Dalmas; Nicolas Venteclef; Chris A. Flask; Julian Kim; Bryan W. Doreian; Kurt Q. Lu; Klaus H. Kaestner; Anne Hamik; Karine Clément; Mukesh K. Jain

Current paradigms suggest that two macrophage subsets, termed M1 and M2, are involved in inflammation and host defense. While the distinct functions of M1 and M2 macrophages have been intensively studied - the former are considered proinflammatory and the latter antiinflammatory - the determinants of their speciation are incompletely understood. Here we report our studies that identify Krüppel-like factor 4 (KLF4) as a critical regulator of macrophage polarization. Macrophage KLF4 expression was robustly induced in M2 macrophages and strongly reduced in M1 macrophages, observations that were recapitulated in human inflammatory paradigms in vivo. Mechanistically, KLF4 was found to cooperate with Stat6 to induce an M2 genetic program and inhibit M1 targets via sequestration of coactivators required for NF-κB activation. KLF4-deficient macrophages demonstrated increased proinflammatory gene expression, enhanced bactericidal activity, and altered metabolism. Furthermore, mice bearing myeloid-specific deletion of KLF4 exhibited delayed wound healing and were predisposed to developing diet-induced obesity, glucose intolerance, and insulin resistance. Collectively, these data identify KLF4 as what we believe to be a novel regulator of macrophage polarization.


Journal of Clinical Investigation | 2012

Endothelial Kruppel-like factor 4 protects against atherothrombosis in mice.

Guangjin Zhou; Anne Hamik; Lalitha Nayak; Hongmei Tian; Hong Shi; Yuan Lu; Nikunj Sharma; Xudong Liao; Andrew T. Hale; Lauren M Boerboom; Ryan E. Feaver; Huiyun Gao; Amar Desai; Alvin H. Schmaier; Stanton L. Gerson; Yunmei Wang; G. Brandon Atkins; Brett R. Blackman; Daniel I. Simon; Mukesh K. Jain

The endothelium regulates vascular homeostasis, and endothelial dysfunction is a proximate event in the pathogenesis of atherothrombosis. Stimulation of the endothelium with proinflammatory cytokines or exposure to hemodynamic-induced disturbed flow leads to a proadhesive and prothrombotic phenotype that promotes atherothrombosis. In contrast, exposure to arterial laminar flow induces a gene program that confers a largely antiadhesive, antithrombotic effect. The molecular basis for this differential effect on endothelial function remains poorly understood. While recent insights implicate Kruppel-like factors (KLFs) as important regulators of vascular homeostasis, the in vivo role of these factors in endothelial biology remains unproven. Here, we show that endothelial KLF4 is an essential determinant of atherogenesis and thrombosis. Using in vivo EC-specific KLF4 overexpression and knockdown murine models, we found that KLF4 induced an antiadhesive, antithrombotic state. Mechanistically, we demonstrated that KLF4 differentially regulated pertinent endothelial targets via competition for the coactivator p300. These observations provide cogent evidence implicating endothelial KLFs as essential in vivo regulators of vascular function in the adult animal.


Journal of Biological Chemistry | 2010

Krüppel-like Factor 4 Inhibits Epithelial-to-Mesenchymal Transition through Regulation of E-cadherin Gene Expression

Jennifer L. Yori; Emhonta Johnson; Guangjin Zhou; Mukesh K. Jain; Ruth A. Keri

The Krüppel-like factor 4 (KLF4) is a transcriptional regulator of proliferation and differentiation in epithelial cells, both during development and tumorigenesis. Although KLF4 functions as a tumor suppressor in several tissues, including the colon, the role of KLF4 in breast cancer is less clear. Here, we show that KLF4 is necessary for maintenance of the epithelial phenotype in non-transformed MCF-10A mammary epithelial cells. KLF4 silencing led to alterations in epithelial cell morphology and migration, indicative of an epithelial-to-mesenchymal transition. Consistent with these changes, decreased levels of KLF4 also resulted in the loss of E-cadherin protein and mRNA. Promoter/reporter analyses revealed decreased E-cadherin promoter activity with KLF4 silencing, while chromatin immunoprecipitation identified endogenous KLF4 binding to the GC-rich/E-box region of this promoter. Furthermore, forced expression of KLF4 in the highly metastatic MDA-MB-231 breast tumor cell line was sufficient to restore E-cadherin expression and suppress migration and invasion. These findings identify E-cadherin as a novel transcriptional target of KLF4. The clear requirement for KLF4 to maintain E-cadherin expression and prevent epithelial-to-mesenchymal transition in mammary epithelial cells supports a metastasis suppressive role for KLF4 in breast cancer.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

Myeloid Krüppel-Like Factor 4 Deficiency Augments Atherogenesis in ApoE−/− Mice—Brief Report

Nikunj Sharma; Yuan Lu; Guangjin Zhou; Xudong Liao; Parul Kapil; Puneet Anand; Ganapati H. Mahabeleshwar; Jonathan S. Stamler; Mukesh K. Jain

Objective—To investigate the role of Krüppel-like factor 4 (KLF4), an essential transcriptional regulator of macrophage polarization (M1/M2), in the pathogenesis of atherosclerosis. Methods and Results—Despite the acknowledged importance of macrophages in atherosclerosis, the role of M1 (classically activated or proinflammatory) versus M2 (alternatively activated or anti-inflammatory) macrophages in this process remains incompletely understood. We recently identified KLF4 as a regulator of macrophage subset specification; that is, KLF4 promotes M2 and inhibits M1 phenotype. Here, we provide evidence that KLF4-deficient macrophages exhibit enhanced proinflammatory activation and foam cell formation in response to oxidized lipids. In vivo, myeloid KLF4-deficient mice (ApoE−/− background) develop significantly more vascular inflammation and atherosclerotic lesion formation. Conclusion—Our findings identify myeloid KLF4 as an essential regulator of vascular inflammation and experimental atherogenesis.


Journal of Clinical Investigation | 2013

Kruppel-like factor 15 is critical for vascular inflammation

Yuan Lu; Lisheng Zhang; Xudong Liao; Panjamaporn Sangwung; Domenick A. Prosdocimo; Guangjin Zhou; Alexander R. Votruba; Leigh Brian; Yuh Jung Han; Huiyun Gao; Yunmei Wang; Koichi Shimizu; Kaitlyn Weinert-Stein; Maria Khrestian; Daniel I. Simon; Neil J. Freedman; Mukesh K. Jain

Activation of cells intrinsic to the vessel wall is central to the initiation and progression of vascular inflammation. As the dominant cellular constituent of the vessel wall, vascular smooth muscle cells (VSMCs) and their functions are critical determinants of vascular disease. While factors that regulate VSMC proliferation and migration have been identified, the endogenous regulators of VSMC proinflammatory activation remain incompletely defined. The Kruppel-like family of transcription factors (KLFs) are important regulators of inflammation. In this study, we identified Kruppel-like factor 15 (KLF15) as an essential regulator of VSMC proinflammatory activation. KLF15 levels were markedly reduced in human atherosclerotic tissues. Mice with systemic and smooth muscle-specific deficiency of KLF15 exhibited an aggressive inflammatory vasculopathy in two distinct models of vascular disease: orthotopic carotid artery transplantation and diet-induced atherosclerosis. We demonstrated that KLF15 alters the acetylation status and activity of the proinflammatory factor NF-κB through direct interaction with the histone acetyltransferase p300. These studies identify a previously unrecognized KLF15-dependent pathway that regulates VSMC proinflammatory activation.


Journal of Clinical Investigation | 2015

Kruppel-like factor 4 is critical for transcriptional control of cardiac mitochondrial homeostasis

Xudong Liao; Rongli Zhang; Yuan Lu; Domenick A. Prosdocimo; Panjamaporn Sangwung; Lilei Zhang; Guangjin Zhou; Puneet Anand; Ling Lai; Teresa C. Leone; Hisashi Fujioka; Fang Ye; Mariana G. Rosca; Charles L. Hoppel; P. Christian Schulze; E. Dale Abel; Jonathan S. Stamler; Daniel P. Kelly; Mukesh K. Jain

Mitochondrial homeostasis is critical for tissue health, and mitochondrial dysfunction contributes to numerous diseases, including heart failure. Here, we have shown that the transcription factor Kruppel-like factor 4 (KLF4) governs mitochondrial biogenesis, metabolic function, dynamics, and autophagic clearance. Adult mice with cardiac-specific Klf4 deficiency developed cardiac dysfunction with aging or in response to pressure overload that was characterized by reduced myocardial ATP levels, elevated ROS, and marked alterations in mitochondrial shape, size, ultrastructure, and alignment. Evaluation of mitochondria isolated from KLF4-deficient hearts revealed a reduced respiration rate that is likely due to defects in electron transport chain complex I. Further, cardiac-specific, embryonic Klf4 deletion resulted in postnatal premature mortality, impaired mitochondrial biogenesis, and altered mitochondrial maturation. We determined that KLF4 binds to, cooperates with, and is requisite for optimal function of the estrogen-related receptor/PPARγ coactivator 1 (ERR/PGC-1) transcriptional regulatory module on metabolic and mitochondrial targets. Finally, we found that KLF4 regulates autophagy flux through transcriptional regulation of a broad array of autophagy genes in cardiomyocytes. Collectively, these findings identify KLF4 as a nodal transcriptional regulator of mitochondrial homeostasis.


Nature Communications | 2015

Circadian control of bile acid synthesis by a KLF15- Fgf15 axis

Shuxin Han; Rongli Zhang; Rajan Jain; Hong Shi; Lilei Zhang; Guangjin Zhou; Panjamaporn Sangwung; Derin Tugal; G. Brandon Atkins; Domenick A. Prosdocimo; Yuan Lu; Xiaonan Han; Patrick Tso; Xudong Liao; Jonathan A. Epstein; Mukesh K. Jain

Recent studies have shown that starburst dwarf galaxies have steeply rising rotation curves in their inner parts, pointing to a close link between the intense star formation and a centrally concentrated mass distribution (baryons and dark matter). More quiescent dwarf irregulars typically have slowly rising rotation curves, although some “compact” irregulars with steep, inner rotation curves exist. We analyze archival Hubble Space Telescope images of two nearby “compact” irregular galaxies (NGC 4190 and NGC 5204), which were selected solely on the basis of their dynamical properties and their proximity. We derive their recent star-formation histories by fitting colormagnitude diagrams of resolved stellar populations, and find that the star-formation properties of both galaxies are consistent with those of known starburst dwarfs. Despite the small sample, this strongly reinforces the notion that the starburst activity is closely related to the inner shape of the potential well.Circadian control of nutrient availability is critical to efficiently meet the energetic demands of an organism. Production of bile acids (BAs), which facilitate digestion and absorption of nutrients, is a major regulator of this process. Here we identify a KLF15-Fgf15 signalling axis that regulates circadian BA production. Systemic Klf15 deficiency disrupted circadian expression of key BA synthetic enzymes, tissue BA levels and triglyceride/cholesterol absorption. Studies in liver-specific Klf15-knockout mice suggested a non-hepatic basis for regulation of BA production. Ileal Fgf15 is a potent inhibitor of BA synthesis. Using a combination of biochemical, molecular and functional assays (including ileectomy and bile duct catheterization), we identify KLF15 as the first endogenous negative regulator of circadian Fgf15 expression. Elucidation of this novel pathway controlling circadian BA production has important implications for physiologic control of nutrient availability and metabolic homeostasis.


JCI insight | 2017

KLF2 and KLF4 control endothelial identity and vascular integrity

Panjamaporn Sangwung; Guangjin Zhou; Lalitha Nayak; E. Ricky Chan; Sandeep Kumar; Dong-Won Kang; Rongli Zhang; Xudong Liao; Yuan Lu; Keiki Sugi; Hisashi Fujioka; Hong Shi; Stephanie Lapping; Chandra C. Ghosh; Sarah J. Higgins; Samir M. Parikh; Hanjoong Jo; Mukesh K. Jain

Maintenance of vascular integrity in the adult animal is needed for survival, and it is critically dependent on the endothelial lining, which controls barrier function, blood fluidity, and flow dynamics. However, nodal regulators that coordinate endothelial identity and function in the adult animal remain poorly characterized. Here, we show that endothelial KLF2 and KLF4 control a large segment of the endothelial transcriptome, thereby affecting virtually all key endothelial functions. Inducible endothelial-specific deletion of Klf2 and/or Klf4 reveals that a single allele of either gene is sufficient for survival, but absence of both (EC-DKO) results in acute death from myocardial infarction, heart failure, and stroke. EC-DKO animals exhibit profound compromise in vascular integrity and profound dysregulation of the coagulation system. Collectively, these studies establish an absolute requirement for KLF2/4 for maintenance of endothelial and vascular integrity in the adult animal.


American Journal of Respiratory Cell and Molecular Biology | 2014

Endothelial Krüppel-Like Factor 4 Modulates Pulmonary Arterial Hypertension

Mohammad Shatat; Hongmei Tian; Rongli Zhang; Gaurav Tandon; Andrew T. Hale; Jason S. Fritz; Guangjin Zhou; José Martínez-González; Cristina Rodríguez; Hunter C. Champion; Mukesh K. Jain; Anne Hamik

Krüppel-like factor 4 (KLF4) is a transcription factor expressed in the vascular endothelium, where it promotes anti-inflammatory and anticoagulant states, and increases endothelial nitric oxide synthase expression. We examined the role of endothelial KLF4 in pulmonary arterial (PA) hypertension (PAH). Mice with endothelial KLF4 knockdown were exposed to hypoxia for 3 weeks, followed by measurement of right ventricular and PA pressures, pulmonary vascular muscularization, and right ventricular hypertrophy. The effect of KLF4 on target gene expression was assessed in lungs from these mice, verified in vitro by small interfering RNA (siRNA) knockdown of KLF4, and further studied at the promoter level with cotransfection experiments. KLF4 expression was measured in lung tissue from patients with PAH and normal control subjects. We found that, after hypoxia, right ventricular and PA pressures were significantly higher in KLF4 knockdown animals than controls. Knockdown animals also had more severe pulmonary vascular muscularization and right ventricular hypertrophy. KLF4 knockdown resulted in increased pulmonary expression of endothelin-1 and decreased expression of endothelial nitric oxide synthase, endothelin receptor subtype B, and prostacyclin synthase. Concordant findings were observed in vitro, both with siRNA knockdown of KLF4 and promoter activity assays. Finally, KLF4 expression was reduced in lungs from patients with PAH. In conclusion, endothelial KLF4 regulates the transcription of genes involved in key pathways implicated in PAH, and its loss exacerbates pulmonary hypertension in response to chronic hypoxia in mice. These results introduce a novel transcriptional modulator of PAH, with the potential of becoming a new therapeutic target.


Circulation | 2017

Krüppel-Like Factor 4 Regulation of Cholesterol-25-Hydroxylase and Liver X Receptor Mitigates Atherosclerosis Susceptibility

Zhao Li; Marcy Martin; Jin Zhang; Hsi Yuan Huang; Liang Bai; Jiao Zhang; Jian Kang; Ming He; Jie Li; Mano Ram Maurya; Shakti Gupta; Guangjin Zhou; Panjamaporn Sangwung; Yong Jiang Xu; Ting Lei; Hsien-Da Huang; Mohit Jain; Mukesh K. Jain; Shankar Subramaniam; John Y.-J. Shyy

Background: Atherosclerosis is a multifaceted inflammatory disease involving cells in the vascular wall (eg, endothelial cells [ECs]), as well as circulating and resident immunogenic cells (eg, monocytes/macrophages). Acting as a ligand for liver X receptor (LXR), but an inhibitor of SREBP2 (sterol regulatory element-binding protein 2), 25-hydroxycholesterol, and its catalyzing enzyme cholesterol-25-hydroxylase (Ch25h) are important in regulating cellular inflammatory status and cholesterol biosynthesis in both ECs and monocytes/macrophages. Methods: Bioinformatic analyses were used to investigate RNA-sequencing data to identify cholesterol oxidation and efflux genes regulated by Krüppel-like factor 4 (KLF4). In vitro experiments involving cultured ECs and macrophages and in vivo methods involving mice with Ch25h ablation were then used to explore the atheroprotective role of KLF4-Ch25h/LXR. Results: Vasoprotective stimuli increased the expression of Ch25h and LXR via KLF4. The KLF4-Ch25h/LXR homeostatic axis functions through suppressing inflammation, evidenced by the reduction of inflammasome activity in ECs and the promotion of M1 to M2 phenotypic transition in macrophages. The increased atherosclerosis in apolipoprotein E–/–/Ch25h–/– mice further demonstrates the beneficial role of the KLF4-Ch25h/LXR axis in vascular function and disease. Conclusions: KLF4 transactivates Ch25h and LXR, thereby promoting the synergistic effects between ECs and macrophages to protect against atherosclerosis susceptibility.

Collaboration


Dive into the Guangjin Zhou's collaboration.

Top Co-Authors

Avatar

Mukesh K. Jain

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Xudong Liao

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Yuan Lu

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Panjamaporn Sangwung

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Hong Shi

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Rongli Zhang

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Domenick A. Prosdocimo

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

G. Brandon Atkins

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Nikunj Sharma

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Anne Hamik

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge