Network


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

Hotspot


Dive into the research topics where Hongkuan Fan is active.

Publication


Featured researches published by Hongkuan Fan.


Journal of Endotoxin Research | 2004

Molecular mechanisms of endotoxin tolerance

Hongkuan Fan; James A. Cook

The phenomenon of endotoxin tolerance has been widely investigated, but to date, the molecular mechanisms of endotoxin tolerance remain to be resolved clearly. The discovery of the Toll-like receptor (TLR) family as the major receptors for lipopolysaccharide (LPS) and other bacterial products has prompted a resurgence of interest in endotoxin tolerance mechanisms. Changes of cell surface molecules, signaling proteins, pro-inflammatory and anti-inflammatory cytokines and other mediators have been examined. During tolerance expression of LPS-binding protein (LBP), CD14, myeloid differentiation protein-2 (MD-2) and TLR2 are unchanged or up-regulated, whereas TLR4 is transiently suppressed or unchanged. Proximal post-receptor signaling proteins that are altered in tolerance include augmented degradation of interleukin-1 receptor-associated kinase (IRAK), and decreased TLR4-myeloid differentiation factor 88 (MyD88) and IRAK-MyD88 association. Tolerance has also been shown to be associated with decreased Gi protein content and activity, decreased protein kinase C (PKC) activity, reduction in mitogen-activated protein kinase (MAP kinase) activity, and reduced activator protein-1 (AP-1) and nuclear factor kappa B (NF-kappaB) induced gene transactivation. However, not all signaling proteins and pathways are suppressed in tolerance and induction of specific anti-inflammatory proteins and signaling pathways may serve important counter inflammatory functions. The latter include induction of IRAK-M and suppressor of cytokine-signaling-1 (SOCS-1), phosphoinositide-3-kinase (PI3K) signaling, and increased or maintained expression of inhibitor-kappaB (IkappaB) isoforms. Also at the nuclear level, increase in the NF-kappaB subunit p50 homodimer expression and increased activation of peroxisome-proliferator-activated receptors-gamma (PPARgamma) have been linked to tolerance phenotype. Although there are species and cellular variations in manifestation of the LPS tolerant phenotype, it is clear that the tolerance phenomena have evolved as a complex orchestrated counter regulatory response to inflammation.


Journal of Endotoxin Research | 2004

Review: Molecular mechanisms of endotoxin tolerance:

Hongkuan Fan; James A. Cook

The phenomenon of endotoxin tolerance has been widely investigated, but to date, the molecular mechanisms of endotoxin tolerance remain to be resolved clearly. The discovery of the Toll-like receptor (TLR) family as the major receptors for lipopolysaccharide (LPS) and other bacterial products has prompted a resurgence of interest in endotoxin tolerance mechanisms. Changes of cell surface molecules, signaling proteins, pro-inflammatory and anti -inflammatory cytokines and other mediators have been examined. During tolerance expression of LPS-binding protein (LBP), CD14, myeloid differentiation protein-2 (MD-2) and TLR2 are unchanged or up-regulated, whereas TLR4 is transiently suppressed or unchanged. Proximal post-receptor signaling proteins that are altered in tolerance include augmented degradation of interleukin-1 receptor-associated kinase (IRAK), and decreased TLR4-myeloid differentiation factor 88 (MyD88) and IRAK-MyD88 association. Tolerance has also been shown to be associated with decreased Gi protein content and activity, decreased protein kinase C (PKC) activity, reduction in mitogen-activated protein kinase (MAP kinase) activity, and reduced activator protein-1 (AP-1) and nuclear factor kappa B (NF-κB) induced gene transactivation. However, not all signaling proteins and pathways are suppressed in tolerance and induction of specific anti-inflammatory proteins and signaling pathways may serve important counter inflammatory functions. The latter include induction of IRAK-M and suppressor of cytokine-signaling-1 (SOCS-1), phosphoinositide-3-kinase (PI3K) signaling, and increased or maintained expression of inhibitor-κB (IκB) isoforms. Also at the nuclear level, increase in the NFκB subunit p50 homodimer expression and increased activation of peroxisome-proliferatoractivated receptors-γ (PPARγ) have been linked to tolerance phenotype. Although there are species and cellular variations in manifestation of the LPS tolerant phenotype, it is clear that the tolerance phenomena have evolved as a complex orchestrated counter regulatory response to inflammation.


Shock | 2004

Toll-like receptor 4 coupled GI protein signaling pathways regulate extracellular signal-regulated kinase phosphorylation and AP-1 activation independent of NFκB activation

Hongkuan Fan; Octavia M. Peck; George E. Tempel; Perry V. Halushka; James A. Cook

Previous studies have implicated heterotrimeric Gi proteins in signaling leading to inflammatory mediator production induced by lipopolysaccharide (LPS). TLR4 has recently been shown to play a central role in response to LPS activation. We hypothesized that Gi proteins are coupled to TLR4 activation of signaling pathways. To inhibit Gi protein function, human embryonic kidney (HEK) 293 cells or RAW 264.7 cells were pretreated with pertussis toxin (PTx), an inhibitor of receptor–Gαi interaction, or transfected with dominant negative Gαi3 (Gαi3dn) or Gαi2 minigene (an inhibitory carboxyl terminus of Gαi2) plasmid. The cells were subsequently transfected with constitutively active TLR4 (TLR4ca) plasmid or TLR4ca together with an NFκB or AP-1 reporter construct. TLR4ca transfection induced ERK 1/2 activation (157 ± 14%, P < 0.01), AP-1 activation (4.0 ± 0.2-fold, P < 0.01), and NFκB activation (8.1 ± 0.4-fold, P < 0.01) compared with empty vector controls. Pretreatment with PTx inhibited TLR4ca-induced ERK 1/2 phosphorylation (30 ± 7%, P < 0.05) and AP-1 activation (36 ± 3%, P < 0.05) but did not inhibit NFκB activation. Cotransfection of TLR4ca with Gαi3dn or Gαi2 minigene also reduced TLR4ca-induced ERK 1/2 phosphorylation (34 ± 10% and 33 ± 5%, respectively, P < 0.05). Constitutively active Gαi2 and Gαi3 plasmids potentiated TLR4ca-induced ERK 1/2 phosphorylation (27 ± 3% and 41 ± 6%, respectively, P < 0.05). βARK-ct plasmid, which inhibits the function of βγ subunit of G protein, has no effect on TLR4ca-induced ERK 1/2 phosphorylation. These data support our hypothesis and provide the first evidence that Gαi-coupled signaling pathways are activated by TLR4. The TLR4-activated Gαi signaling pathway activates ERK 1/2 phosphorylation and AP-1 activation independently of TLR4-mediated signaling to NFκB activation.


Immunology | 2010

Beta-arrestin 2 negatively regulates sepsis-induced inflammation

Hongkuan Fan; Alessandra Bitto; Basilia Zingarelli; Louis M. Luttrell; Keith T. Borg; Perry V. Halushka; James A. Cook

β‐arrestins 1 and 2 are ubiquitously expressed proteins that alter signalling by G‐protein‐coupled receptors. β‐arrestin 2 plays an important role as a signalling adaptor and scaffold in regulating cellular inflammatory responses. We hypothesized that β‐arrestin 2 is a critical modulator of inflammatory response in experimental sepsis. β‐arrestin 2(−/−) and wild‐type (WT) mice were subjected to caecal ligation and puncture (CLP). The survival rate was significantly decreased (P < 0·05) in β‐arrestin 2(−/−) mice (13% survival) compared with WT mice (53% survival). A second group of mice were killed 18 hr after CLP for blood, peritoneal lavage and tissue sample collection. CLP‐induced plasma interleukin (IL)‐6 was significantly increased 25 ± 12 fold and caecal myeloperoxidase (MPO) activity was increased 2·4 ± 0·3 fold in β‐arrestin 2(−/−) compared with WT mice. β‐arrestin 2(−/−) mice exhibited more severe lung damage and higher bacterial loads compared with WT mice post CLP challenge as measured by histopathology and colony‐forming unit count. In subsequent experiments, splenocytes, peritoneal macrophages and bone marrow‐derived macrophages (BMDMs) were isolated and cultured from β‐arrestin 2(−/−) and WT mice and stimulated in vitro with lipopolysaccharide (LPS). Tumour necrosis factor (TNF)‐α, IL‐6 and IL‐10 production induced by LPS was significantly augmented (2·2 ± 0·2 fold, 1·8 ± 0·1 fold, and 2·2 ± 0·4 fold, respectively; P < 0·05) in splenocytes from β‐arrestin 2(−/−) mice compared with WT mice. The splenocyte response was different from that of peritoneal macrophages or BMDMs, which exhibited no difference in TNF‐α and IL‐6 production upon LPS stimulation between WT and β‐arrestin 2(−/−) mice. Our data demonstrate that β‐arrestin 2 functions to negatively regulate the inflammatory response in polymicrobial sepsis.


American Journal of Pathology | 2010

Peroxisome Proliferator-Activated Receptor δ Regulates Inflammation via NF-κB Signaling in Polymicrobial Sepsis

Basilia Zingarelli; Giovanna Piraino; Paul W. Hake; Michael O'Connor; Alvin Denenberg; Hongkuan Fan; James A. Cook

The nuclear peroxisome proliferator-activated receptor δ (PPARδ) is an important regulator of lipid metabolism. In contrast to its known effects on energy homeostasis, its biological role on inflammation is not well understood. We investigated the role of PPARδ in the modulation of the nuclear factor-κB (NF-κB)-driven inflammatory response to polymicrobial sepsis in vivo and in macrophages in vitro. We demonstrated that administration of GW0742, a specific PPARδ ligand, provided beneficial effects to rats subjected to cecal ligation and puncture, as shown by reduced systemic release of pro-inflammatory cytokines and neutrophil infiltration in lung, liver, and cecum, when compared with vehicle treatment. Molecular analysis revealed that treatment with GW0742 reduced NF-κB binding to DNA in lung and liver. In parallel experiments, heterozygous PPARδ-deficient mice suffered exaggerated lethality when subjected to cecal ligation and puncture and exhibited severe lung injury and higher levels of circulating tumor necrosis factor-α (TNFα) and keratinocyte-derived chemokine than wild-type mice. Furthermore, in lipopolysaccharide-stimulated J774.A1 macrophages, GW0742 reduced TNFα production by inhibiting NF-κB activation. RNA silencing of PPARδ abrogated the inhibitory effects of GW0742 on TNFα production. Chromatin immunoprecipitation assays revealed that PPARδ displaced the NF-κB p65 subunit from the κB elements of the TNFα promoter, while recruiting the co-repressor BCL6. These data suggest that PPARδ is a crucial anti-inflammatory regulator, providing a basis for novel sepsis therapies.


Molecular Medicine | 2008

Lysophosphatidic acid inhibits bacterial endotoxin-induced pro-inflammatory response: potential anti-inflammatory signaling pathways.

Hongkuan Fan; Basilia Zingarelli; Vashaunta Harris; George E. Tempel; Perry V. Halushka; James A. Cook

Previous studies have demonstrated that heterotrimeric guanine nucleotide-binding regulatory (Gi) protein-deficient mice exhibit augmented inflammatory responses to lipopolysaccharide (LPS). These findings suggest that Gi protein agonists will suppress LPS-induced inflammatory gene expression. Lysophosphatidic acid (LPA) activates G protein-coupled receptors leading to Gi protein activation. We hypothesized that LPA will inhibit LPS-induced inflammatory responses through activation of Gi-coupled anti-inflammatory signaling pathways. We examined the anti-inflammatory effect of LPA on LPS responses both in vivo and in vitro in CD-1 mice. The mice were injected intravenously with LPA (10 mg/kg) followed by intraperitoneal injection of LPS (75 mg/kg for survival and 25 mg/kg for other studies). LPA significantly increased the mice survival to endotoxemia (P < 0.05). LPA injection reduced LPS-induced plasma TNF-α production (69 ± 6%, P < 0.05) and myeloperoxidase (MPO) activity in lung (33 ± 9%, P < 0.05) as compared to vehicle injection. LPS-induced plasma IL-6 was unchanged by LPA. In vitro studies with peritoneal macrophages paralleled results from in vivo studies. LPA (1 and 10 µM) significantly inhibited LPS-induced TNFα production (61 ± 9% and 72 ± 9%, respectively, P < 0.05) but not IL-6. We further demonstrated that the anti-inflammatory effect of LPA was reversed by ERK 1/2 and phosphatase inhibitors, suggesting that ERK 1/2 pathway and serine/threonine phosphatases are involved. Inhibition of phosphatidylinositol 3 (PI3) kinase signaling pathways also partially reversed the LPA anti-inflammatory response. However, LPA did not alter NFκB and peroxisome proliferator-activated receptor γ (PPARγ) activation. Inhibitors of PPARγ did not alter LPA-induced inhibition of LPS signaling. These studies demonstrate that LPA has significant anti-inflammatory activities involving activation of ERK 1/2, serine/threonine phosphatases, and PI3 kinase signaling pathways.


Molecular Immunology | 2011

Increased expression of beta-arrestin 1 and 2 in murine models of rheumatoid arthritis: isoform specific regulation of inflammation.

Pengfei Li; James A. Cook; Gary S. Gilkeson; Louis M. Luttrell; Liping Wang; Keith T. Borg; Perry V. Halushka; Hongkuan Fan

Pro-inflammatory cytokines and chemokines play critical roles in autoimmune diseases including rheumatoid arthritis (RA). Recently, it has been reported that β-arrestin 1 and 2 are involved in the regulation of inflammation. We hypothesized that β-arrestin 1 and 2 play critical roles in murine models of RA. Using a collagen-induced arthritis (CIA) and a human TNFα transgenic (TNFtg) mouse model, we demonstrated that β-arrestin 1 and 2 expression are significantly increased in joint tissue of CIA mice and TNFtg mice. In fibroblast-like synoviocytes (FLS) isolated from hind knee joint of CIA mice, we observed an increase of β-arrestin 1 and 2 protein and mRNA levels in the early stage of arthritis. In FLS, low molecular weight hyaluronan (HA)-induced TNFα and IL-6 production was increased by overexpression of β-arrestin 1 but decreased by overexpression of β-arrestin 2 demonstrating isoform specific regulation. TNFα and HA induced an increase of β-arrestin 1 and 2 expression in FLS, while high mobility group box (HMGB)-1 only stimulated β-arrestin 1 expression. TNFα- or HA-induced β-arrestin 2 expression was blocked by a p38 inhibitor. To examine the in vivo role of β-arrestin 2 in the pathogenesis of arthritis, WT and β-arrestin 2 KO mice were subjected to collagen antibody-induced arthritis (CAIA). β-Arrestin 2 KO mice exhibited more severe arthritis in CAIA. Thus β-arrestin 2 is anti-inflammatory in CAIA. These composite observations suggest that β-arrestin 1 and 2 differentially regulate FLS inflammation and increased β-arrestin 2 may reduce experimental arthritis severity.


Immunology | 2007

Differential regulation of lipopolysaccharide and Gram-positive bacteria induced cytokine and chemokine production in macrophages by Gαi proteins

Hongkuan Fan; David L. Williams; Basilia Zingarelli; Kevin F. Breuel; Giuseppe Teti; George E. Tempel; Karsten Spicher; Guylain Boulay; Lutz Birnbaumer; Perry V. Halushka; James A. Cook

Heterotrimeric Gi proteins play a role in signalling activated by lipopolysaccharide (LPS), Staphylococcus aureus (SA) and group B streptococci (GBS), leading to production of inflammatory mediators. We hypothesized that genetic deletion of Gi proteins would alter cytokine and chemokine production induced by LPS, SA and GBS stimulation. LPS‐induced, heat‐killed SA‐induced and heat‐killed GBS‐induced cytokine and chemokine production in peritoneal macrophages from wild‐type (WT), Gαi2–/– or Gαi1/3–/– mice were investigated. LPS induced production of tumour necrosis factor‐α (TNF‐α), interleukin‐6 (IL‐6), IL‐10 and interferon‐γ‐inducible protein‐10 (IP‐10); SA induced TNF‐α, and IL‐1β production; and GBS induced TNF‐α, IL‐6, IL‐1β, macrophage inflammatory protein‐1α (MIP‐1α) and keratinocyte chemoattract (KC) production were all decreased (P < 0·05) in Gαi2–/– or Gαi1/3–/– mice compared with WT mice. In contrast to the role of Gi proteins as a positive regulator of mediators, LPS‐induced production of MIP‐1α and granulocyte–macrophage colony‐stimulating factor (GM‐CSF) were increased in macrophages from Gαi1/3–/– mice, and SA‐induced MIP‐1α production was increased in both groups of Gαi protein‐depleted mice. LPS‐induced production of KC and IL‐1β, SA‐induced production of GM‐CSF, KC and IP‐10, and GBS‐induced production of IL‐10, GM‐CSF and IP‐10 were unchanged in macrophages from Gαi2–/– or Gαi1/3–/– mice compared with WT mice. These data suggest that Gi2 and Gi1/3 proteins are both involved and differentially regulate murine inflammatory cytokine and chemokine production in response to both LPS and Gram‐positive microbial stimuli.


Immunology | 2014

Human kallistatin administration reduces organ injury and improves survival in a mouse model of polymicrobial sepsis

Pengfei Li; Grant Bledsoe; Zhi-Rong Yang; Hongkuan Fan; Lee Chao; Julie Chao

Kallistatin, a plasma protein, has been shown to exert multi‐factorial functions including inhibition of inflammation, oxidative stress and apoptosis in animal models and cultured cells. Kallistatin levels are reduced in patients with sepsis and in lipopolysaccharide (LPS)‐induced septic mice. Moreover, transgenic mice expressing kallistatin are more resistant to LPS‐induced mortality. Here, we investigated the effects of human kallistatin on organ injury and survival in a mouse model of polymicrobial sepsis. In this study, mice were injected intravenously with recombinant kallistatin (KS3, 3 mg/kg; or KS10, 10 mg/kg body weight) and then rendered septic by caecal ligation and puncture 30 min later. Kallistatin administration resulted in a > 10‐fold reduction of peritoneal bacterial counts, and significantly decreased serum tumour necrosis factor‐α, interleukin‐6 and high mobility group box‐1 (HMGB1) levels. Kallistatin also inhibited HMGB1 and toll‐like receptor‐4 gene expression in the lung and kidney. Administration of kallistatin attenuated renal damage and decreased blood urea nitrogen and serum creatinine levels, but increased endothelial nitric oxide synthase and nitric oxide levels in the kidney. In cultured endothelial cells, human kallistatin via its heparin‐binding site inhibited HMGB1‐induced nuclear factor‐κB activation and inflammatory gene expression. Moreover, kallistatin significantly reduced apoptosis and caspase‐3 activity in the spleen. Furthermore, kallistatin treatment markedly improved the survival of septic mice by 23% (KS3) and 41% (KS10). These results indicate that kallistatin is a unique protecting agent in sepsis‐induced organ damage and mortality by inhibiting inflammation and apoptosis, as well as enhancing bacterial clearance in a mouse model of polymicrobial sepsis.


Inflammation | 2012

Beneficial Effect of a CXCR4 Agonist in Murine Models of Systemic Inflammation

Hongkuan Fan; Donald Wong; Sarah Ashton; Keith T. Borg; Perry V. Halushka; James A. Cook

The chemokine CXC receptor 4 (CXCR4) is activated by stromal cell-derived factor (SDF-1α). CXCR4 may be part of a lipopolysaccharide (LPS) sensing co-clustering complex that modulates TLR4 activation and evidence suggest that SDF-1α can activate anti-inflammatory signaling pathways and suppress inflammation. In the present study we examined the hypothesis that the SDF-1α peptide analog and CXCR4 agonist CTCE-0214 is anti-inflammatory in three distinct models of murine systemic inflammation. Our findings demonstrate that CTCE-0214 in vivo significantly suppressed plasma tumor necrosis factor alpha (TNF-α) increases in acute endotoxemia and following zymosan-induced multiple organ dysfunction syndrome (MODS). In both models, CTCE-0214 did not suppress plasma increases in the anti-inflammatory cytokine interleukin (IL)-10. CTCE-0214 improved survival without antibiotics in a model of severe sepsis induced by cecal ligation and puncture (CLP). CTCE-0214 also decreased plasma increases in IL-6 but not TNF-α and IL-10 in response to CLP-induced inflammation. We demonstrated in a moderately severe model of CLP (one puncture) that IL-6 levels at 24 h were similar to sham controls. However in severe CLP (two punctures) plasma IL-6 levels were markedly elevated. Plasma SDF-1α levels varied inversely with the plasma IL-6. In addition to the beneficial effect of CTCE-0214 in these models of systemic inflammation in vivo, we also demonstrated that the analog dose dependently suppressed LPS-induced IL-6 production in bone marrow-derived macrophages. CTCE-0214 therefore may be beneficial in controlling inflammation sepsis and systemic inflammatory syndromes.

Collaboration


Dive into the Hongkuan Fan's collaboration.

Top Co-Authors

Avatar

James A. Cook

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Perry V. Halushka

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Basilia Zingarelli

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

George E. Tempel

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Octavia M. Peck

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Keith T. Borg

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Andrew J. Goodwin

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Pengfei Li

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge