Network


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

Hotspot


Dive into the research topics where Juquan Song is active.

Publication


Featured researches published by Juquan Song.


Shock | 2008

Characterization of the inflammatory response during acute and post-acute phases after severe burn.

Gerd G. Gauglitz; Juquan Song; David N. Herndon; Celeste C. Finnerty; Darren Boehning; José M. Barral; Marc G. Jeschke

Severe burn causes a pronounced hypermetabolic response characterized by catabolism and extensive protein wasting. We recently found that this hypermetabolic state is driven by a severe inflammatory response. We characterized in detail the kinetics of serum levels of a panel of cytokines in a rat model, which may serve as reference for the development of therapeutic interventions applicable to humans. Male Sprague-Dawley rats (n = 8) received a full-thickness burn of 60% total body surface area. Serum was harvested 1, 3, 6, 12, 24, 48, 96, and 168 h after burn. Eight serum cytokines commonly used to assess the inflammatory response in humans, such as IL-1&bgr;, IL-6, IL-10, TNF, vascular endothelial growth factor, and monocyte chemotactic protein 1, and the rat-specific cytokines cytokine-induced neutrophil chemoattractant (CINC) 1, CINC-2, and CINC-3 were measured by enzyme-linked immunosorbent assay technique and were compared with controls (n = 4). Statistical analysis was conducted using the t test, with P < 0.05 considered as significantly different. Thermal injury resulted in significantly increased serum levels of IL-1&bgr;, IL-6, IL-10, monocyte chemotactic protein 1, CINC-1, CINC-2, and CINC-3 when compared with the concentrations detected in nonburned rats (P < 0.05). Serum levels of TNF-&agr; and vascular endothelial growth factor in burned rats were not found to be significantly different to controls. Burn causes a profound inflammatory response in rats. Specific cytokines known to increase in humans postburn such as IL-1 &bgr;, IL-6, IL-10, MCP-1, and IL-8 (CINC-1, CINC-2, and CINC-3 in the rat) were also observed in our rat burn model, which now allows us to study new anti-inflammatory treatment options.


Annals of Surgery | 2012

Severe injury is associated with insulin resistance, endoplasmic reticulum stress response, and unfolded protein response.

Marc G. Jeschke; Celeste C. Finnerty; David N. Herndon; Juquan Song; Darren Boehning; Ronald G. Tompkins; Henry V. Baker; Gerd G. Gauglitz

Objective: We determined whether postburn hyperglycemia and insulin resistance are associated with endoplasmic reticulum (ER) stress/unfolded protein response (UPR) activation leading to impaired insulin receptor signaling. Background: Inflammation and cellular stress, hallmarks of severely burned and critically ill patients, have been causally linked to insulin resistance in type 2 diabetes via induction of ER stress and the UPR. Methods: Twenty severely burned pediatric patients were compared with 36 nonburned children. Clinical markers, protein, and GeneChip analysis were used to identify transcriptional changes in ER stress and UPR and insulin resistance–related signaling cascades in peripheral blood leukocytes, fat, and muscle at admission and up to 466 days postburn. Results: Burn-induced inflammatory and stress responses are accompanied by profound insulin resistance and hyperglycemia. Genomic and protein analysis revealed that burn injury was associated with alterations in the signaling pathways that affect insulin resistance, ER/sarcoplasmic reticulum stress, inflammation, and cell growth/apoptosis up to 466 days postburn. Conclusion: Burn-induced insulin resistance is associated with persistent ER/sarcoplasmic reticulum stress/UPR and subsequent suppressed insulin receptor signaling over a prolonged period of time.


Journal of Cellular and Molecular Medicine | 2009

Calcium and ER stress mediate hepatic apoptosis after burn injury

Marc G. Jeschke; Gerd G. Gauglitz; Juquan Song; Gabriela A. Kulp; Celeste C. Finnerty; Robert A. Cox; José M. Barral; David N. Herndon; Darren Boehning

A hallmark of the disease state following severe burn injury is decreased liver function, which results in gross metabolic derangements that compromise patient survival. The underlying mechanisms leading to hepatocyte dysfunction after burn are essentially unknown. The aim of the present study was to determine the underlying mechanisms leading to hepatocyte dysfunction and apoptosis after burn. Rats were randomized to either control (no burn) or burn (60% total body surface area burn) and sacrificed at various time-points. Liver was either perfused to isolate primary rat hepatocytes, which were used for in vitro calcium imaging, or liver was harvested and processed for immunohistology, transmission electron microscopy, mitochondrial isolation, mass spectroscopy or Western blotting to determine the hepatic response to burn injury in vivo. We found that thermal injury leads to severely depleted endoplasmic reticulum (ER) calcium stores and consequent elevated cytosolic calcium concentrations in primary hepatocytes in vitro. Burn-induced ER calcium depletion caused depressed hepatocyte responsiveness to signalling molecules that regulate hepatic homeostasis, such as vasopressin and the purinergic agonist ATP. In vivo, thermal injury resulted in activation of the ER stress response and major alterations in mitochondrial structure and function - effects which may be mediated by increased calcium release by inositol 1,4,5-trisphosphate receptors. Our results reveal that thermal injury leads to dramatic hepatic disturbances in calcium homeostasis and resultant ER stress leading to mitochondrial abnormalities contributing to hepatic dysfunction and apoptosis after burn injury.


Molecular Medicine | 2009

Severe burn-induced endoplasmic reticulum stress and hepatic damage in mice

Juquan Song; Celeste C. Finnerty; David N. Herndon; Darren Boehning; Marc G. Jeschke

Severe burn injury results in liver dysfunction and damage, with subsequent metabolic derangements contributing to patient morbidity and mortality. On a cellular level, significant postburn hepatocyte apoptosis occurs and likely contributes to liver dysfunction. However, the underlying mechanisms of hepatocyte apoptosis are poorly understood. The endoplasmic reticulum (ER) stress response/unfolded protein response (UPR) pathway can lead to hepatocyte apoptosis under conditions of liver dysfunction. Thus, we hypothesized that ER stress/UPR may mediate hepatic dysfunction in response to burn injury. We investigated the temporal activation of hepatic ER stress in mice after a severe burn injury. Mice received a scald burn over 35% of their body surface and were killed at 1, 7, 14, and 21 d postburn. We found that severe burn induces hepatocyte apoptosis as indicated by increased caspase-3 activity (P < 0.05). Serum albumin levels decreased postburn and remained lowered for up to 21 d, indicating that constitutive secretory protein synthesis was reduced. Significantly, upregulation of the ER stress markers glucose-related protein 78 (GRP78)/BIP protein disulfide isomerase (PDI), p-protein kinase R-like endoplasmic reticulum kinase (p-PERK), and inositol-requiring enzyme 1α (IRE-1α) were found beginning 1 d postburn (P < 0.05) and persisted up to 21 d postburn (P < 0.05). Hepatic ER stress induced by burn injury was associated with compensatory upregulation of the calcium chaperone/storage proteins calnexin and calreticulin (P< 0.05), suggesting that ER calcium store depletion was the primary trigger for induction of the ER stress response. In summary, thermal injury in mice causes long-term adaptive and deleterious hepatic function alterations characterized by significant upregulation of the ER stress response.


Shock | 2015

Predictive Value of IL-8 for Sepsis and Severe Infections After Burn Injury: A Clinical Study.

Robert Kraft; David N. Herndon; Celeste C. Finnerty; Robert A. Cox; Juquan Song; Marc G. Jeschke

ABSTRACT The inflammatory response induced by burn injury contributes to increased incidence of infections, sepsis, organ failure, and mortality. Thus, monitoring postburn inflammation is of paramount importance but, so far, there are no reliable biomarkers available to monitor and/or predict infectious complications after burn. As interleukin 8 (IL-8) is a major mediator for inflammatory responses, the aim of our study was to determine whether IL-8 expression can be used to predict postburn sepsis, infections, and mortality. Plasma cytokines, acute-phase proteins, constitutive proteins, and hormones were analyzed during the first 60 days after injury from 468 pediatric burn patients. Demographics and clinical outcome variables (length of stay, infection, sepsis, multiorgan failure [MOF], and mortality) were recorded. A cutoff level for IL-8 was determined using receiver operating characteristic analysis. Statistical significance is set at P < 0.05. Receiver operating characteristic analysis identified a cutoff level of 234 pg/mL for IL-8 for survival. Patients were grouped according to their average IL-8 levels relative to this cutoff and stratified into high (H) (n = 133) and low (L) (n = 335) groups. In the L group, regression analysis revealed a significant predictive value of IL-8 to percent of total body surface area burned and incidence of MOF (P < 0.001). In the H group, IL-8 levels were able to predict sepsis (P < 0.002). In the H group, elevated IL-8 was associated with increased inflammatory and acute-phase responses compared with the L group (P < 0.05). High levels of IL-8 correlated with increased MOF, sepsis, and mortality. These data suggest that serum levels of IL-8 may be a valid biomarker for monitoring sepsis, infections, and mortality in burn patients.


Molecular Medicine | 2011

Insulin protects against hepatic damage postburn.

Marc G. Jeschke; Robert Kraft; Juquan Song; Gerd G. Gauglitz; Robert A. Cox; Natasha C. Brooks; Celeste C. Finnerty; Gabriela A. Kulp; David N. Herndon; Darren Boehning

Burn injury causes hepatic dysfunction associated with endoplasmic reticulum (ER) stress and induction of the unfolded protein response (UPR). ER stress/UPR leads to hepatic apoptosis and activation of the Jun-N-terminal kinase (JNK) signaling pathway leading to vast metabolic alterations. Insulin has been shown to attenuate hepatic damage and to improve liver function. We therefore hypothesized that insulin administration exerts its effects by attenuating postburn hepatic ER stress and subsequent apoptosis. Male Sprague Dawley rats received a 60% total body surface area (TBSA) burn injury. Animals were randomized to receive saline (controls) or insulin (2.5 IU/kg q. 24 h) and euthanized at 24 and 48 h postburn. Burn injury induced dramatic changes in liver structure and function, including induction of the ER stress response, mitochondrial dysfunction, hepatocyte apoptosis, and up-regulation of inflammatory mediators. Insulin decreased hepatocyte caspase-3 activation and apoptosis significantly at 24 and 48 h postburn. Furthermore, insulin administration decreased ER stress significantly and reversed structural and functional changes in hepatocyte mitochondria. Finally, insulin attenuated the expression of inflammatory mediators IL-6, MCP-1, and CINC-1. Insulin alleviates burn-induced ER stress, hepatocyte apoptosis, mitochondrial abnormalities, and inflammation leading to improved hepatic structure and function significantly. These results support the use of insulin therapy after traumatic injury to improve patient outcomes.


Critical Care Medicine | 2010

Insulin increases resistance to burn wound infection-associated sepsis.

Gerd G. Gauglitz; Tracy Toliver-Kinsky; Felicia N. Williams; Juquan Song; Weihua Cui; David N. Herndon; Marc G. Jeschke

Objective:This study was designed to determine the ability of insulin to improve outcome following a Pseudomonas aeruginosa wound infection in a rodent model of severe burn injury. Background:Severe burn injury predisposes patients to burn wound infections that can disseminate, lead to uncontrolled inflammation, and induce septic shock. Whereas insulin administration has been extensively discussed to improve morbidity and mortality in critically ill patients, the ability of insulin to improve outcomes of severely burned patients with infected burn wounds is not known. Design:Sprague-Dawley rats. Setting:University setting. Intervention:Burn-injured Sprague Dawley rats were randomized into treatment groups that received either saline or insulin. Burn wounds were topically inoculated with a lethal dose of Pseudomonas aeruginosa 6 days after injury. Measurements and Main Results:Survival, systemic dissemination of bacteria, systemic inflammation, and immune activation were examined. Insulin decreased the early inflammatory response to a severe burn injury. Treatment with low doses of insulin following burn injury improved the outcome of rats in response to a lethal burn wound infection. Specifically, survival was improved and systemic dissemination of bacteria from the wound was decreased. Systemic inflammation, indicated by serum interleukin-6 levels, was significantly decreased by insulin treatments after injury. Additionally, insulin treatments were associated with alterations in B and T lymphocyte responses to wound infection. Conclusions:Although the mechanisms by which insulin improves outcome following a lethal burn wound infection are not known, the data suggest that immunologic responses to infection may be altered by postburn insulin treatments.


Shock | 2008

Second hit post burn increased proximal gut mucosa epithelial cells damage.

Juquan Song; Steven Wolf; David N. Herndon; Xiao Wu Wu; Marc G. Jeschke

Secondary infections after burn are common and are a major contributor to morbidity and mortality. We previously showed that burn disrupted proximal gut mucosal homeostasis through increased epithelial cell apoptosis. In the present study, we sought to determine whether proximal gut mucosal disruption is additively affected by secondary endotoxemia after a severe burn. C57BL/6 mice received 30% total body surface area full-thickness scald burns and were randomized to receive saline or LPS 1 mg/kg body weight given intraperitoneally 72 h after burn. Proximal small bowel was harvested 12 h after LPS injection. Mucosal height and epithelial cell number were assessed on hematoxylin-eosin sections, intestinal epithelial cell apoptosis was identified by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling, and cell proliferation by immunohistochemical staining for proliferating cell nuclear antigen. Results showed that proximal gut mucosa impairment occurred 12 h after injury, including significantly decreased proximal gut wet weight, gut mucosal height, and epithelial cell number associated with increased proximal gut epithelial apoptosis (P < 0.05). This impairment diminished 72 h after burn. Second-hit endotoxemia caused additional proximal gut mucosa damage with decreased proximal gut weight, cell number, and mucosal height (P < 0.05) and significantly increased small intestinal epithelial apoptosis and mucosal atrophy, even after the first event, indicating a second detrimental effect of endotoxemia after the initial injury.


Shock | 2003

Decreased lymphocyte apoptosis by anti-tumor necrosis factor antibody in peyer's patches after severe burn

Kenneth J. Woodside; Marcus Spies; Xiao Wu Wu; Juquan Song; Shahnaz S. Quadeer; John A. Daller; Steven E. Wolf

&NA; —Severe burn results in immunosuppression, with increased lymphocyte apoptosis in both the central and peripheral immune system. As atrophy of the small intestine has been described in mouse models and intestinal lymphocytes have been implicated in the burn inflammatory response, we examined the effects of burn and tumor necrosis factor (TNF)‐&agr; on lymphocytes in intestinal Peyers patches. Anesthetized C57BL6 mice received a 30% full‐thickness scald burn on the upper back. Sham‐burned animals served as controls. Anti‐TNF or control immunoglobulin (Ig) G antibody (200 μg) was given immediately after the burn. The animals were initially resuscitated with 2 mL of normal saline, and were then sacrificed 12 h postburn. Terminal deoxyuridine nick‐end labeling (TUNEL) and proliferative cell nuclear antigen (PCNA) staining was performed. Apoptosis was quantified as apoptotic lymphocytes/high‐powered field (hpf). Results, expressed as mean ± SEM, were compared using analysis of variance (ANOVA) and the Student‐Newman‐Keuls test. All mice survived the burn. An initial time‐course experiment demonstrated maximal Peyers patch apoptosis 12 h after the burn. Sham mice had 25 ± 7 TUNEL‐stained cells/hpf in Peyers patches, whereas burned mice had 93 ± 18 cells/hpf (P < 0.05). In contrast, burned mice receiving anti‐TNF antibody had 28 ± 8 TUNEL‐stained cells/hpf (P < 0.05 vs. burn), whereas sham mice receiving anti‐TNF antibody had 20 ± 4 cells/hpf. There were no significant differences in PCNA staining between the groups. Scald burn results in lymphocyte apoptosis in Peyers patches. This apoptosis can be abrogated by the addition of anti‐TNF antibody. Apoptotic changes may lead to the failure of the intestinal immunological barrier and increased risk of sepsis.


Molecular Medicine | 2012

Propranolol Improves Impaired Hepatic Phosphatidylinositol 3-Kinase/Akt Signaling after Burn Injury

Natasha C. Brooks; Juquan Song; Darren Boehning; Robert Kraft; Celeste C. Finnerty; David N. Herndon; Marc G. Jeschke

Severe burn injury is associated with induction of the hepatic endoplasmic reticulum (ER) stress response. ER stress leads to activation of c-Jun N-terminal kinase (JNK), suppression of insulin receptor signaling via phosphorylation of insulin receptor substrate 1 and subsequent insulin resistance. Marked and sustained increases in catecholamines are prominent after a burn. Here, we show that administration of propranolol, a nonselective β1/2 adrenergic receptor antagonist, attenuates ER stress and JNK activation. Attenuation of ER stress by propranolol results in increased insulin sensitivity, as determined by activation of hepatic phosphatidylinositol 3-kinase and Akt. We conclude that catecholamine release is responsible for the ER stress response and impaired insulin receptor signaling after burn injury.

Collaboration


Dive into the Juquan Song's collaboration.

Top Co-Authors

Avatar

Steven E. Wolf

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David N. Herndon

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Celeste C. Finnerty

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Darren Boehning

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Charles E. Wade

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Lisa A. Baer

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert Kraft

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Ryan M. Huebinger

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge