Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
J Infect Dis ; 229(4): 1166-1177, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-37633660

ABSTRACT

Glucocorticoid (GC) therapy had been strongly recommended for pediatric sepsis (grade 1A). However, the recommendation was changed to grade 2C in 2020 due to weak evidence. About 32.8% of patients with pediatric septic develop relative adrenal insufficiency (RAI). But whether GC therapy should be determined by RAI status is controversial. This study utilized 21-day-old SF1CreSRBIfl/fl mice as the first pediatric RAI mouse model to assess the pathogenesis of RAI and evaluate GC therapy. RAI mice exhibited a substantially higher mortality rate in cecal ligation and puncture and cecal slurry-induced sepsis. These mice featured persistent inflammatory responses and were effectively rescued by GC therapy. RNA sequencing analysis revealed persistent inflammatory responses in RAI mice, caused by transcriptional dysregulation of AP-1 and NF-κB, and cytokine-induced secondary inflammatory response. Our findings support a precision medicine approach to guide GC therapy for pediatric patients based on the status of RAI.


Subject(s)
Adrenal Insufficiency , Sepsis , Humans , Child , Mice , Animals , Adrenal Insufficiency/etiology , Cytokines , NF-kappa B , Cecum , Ligation/adverse effects , Risk Factors
2.
Sci Signal ; 16(777): eadd4900, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36943922

ABSTRACT

Cytokine release syndrome (CRS) is a systemic inflammatory syndrome associated with infection- or drug-induced T cell activation and can cause multiple organ failure and even death. Because current treatments are ineffective in some patients with severe CRS, we set out to identify risk factors and mechanisms behind severe CRS that might lead to preventive therapies and better clinical outcomes in patients. In mice, we found that deficiency in the adrenal stress response-with similarities to such in patients called relative adrenal insufficiency (RAI)-conferred a high risk for lethal CRS. Mice treated with CD3 antibodies were protected against lethal CRS by the production of glucocorticoids (GC) induced by the adrenal stress response in a manner dependent on the scavenger receptor B1 (SR-BI), a receptor for high-density lipoprotein (HDL). Mice with whole-body or adrenal gland-specific SR-BI deficiency exhibited impaired GC production, more severe CRS, and increased mortality in response to CD3 antibodies. Pretreatment with a low dose of GC effectively suppressed the development of CRS and rescued survival in SR-BI-deficient mice without compromising T cell function through apoptosis. Our findings suggest that RAI may be a risk factor for therapy-induced CRS and that pretreating RAI patients with GC may prevent lethal CRS.


Subject(s)
Adrenal Glands , Glucocorticoids , Mice , Animals , Scavenger Receptors, Class B , Mice, Knockout , Glucocorticoids/pharmacology , Lipoproteins, HDL
3.
Nanomedicine (Lond) ; 18(29): 2127-2142, 2023 12.
Article in English | MEDLINE | ID: mdl-38197376

ABSTRACT

Aim: The impacts of synthetic high-density lipoprotein (sHDL) phospholipid components on anti-sepsis effects were investigated. Methods: sHDL composed with ApoA-I mimetic peptide (22A) and different phosphatidylcholines were prepared and characterized. Anti-inflammatory effects were investigated in vitro and in vivo on lipopolysaccharide (LPS)-induced inflammation models. Results: sHDLs composed with 1,2-dimyristoyl-sn-glycero-3-phosphocholine (22A-DMPC) most effectively neutralizes LPS, inhibits toll-like receptor 4 recruitment into lipid rafts, suppresses nuclear factor κB signaling and promotes activating transcription factor 3 activating. The lethal endotoxemia animal model showed the protective effects of 22A-DMPC. Conclusion: Phospholipid components affect the stability and fluidity of nanodiscs, impacting the anti-septic efficacy of sHDLs. 22A-DMPC presents the strongest LPS binding and anti-inflammatory effects in vitro and in vivo, suggesting a potential sepsis treatment.


Sepsis is triggered by endotoxins released by bacteria. These endotoxins trigger an exaggerated inflammatory response, leading to widespread inflammation and organ damage. Synthetic high-density lipoprotein (sHDL) is a potential treatment of sepsis by neutralizing endotoxins and regulating inflammatory responses. The phospholipid components of sHDL may affect the effectiveness of sHDL against sepsis. In this study, we prepared sHDLs with different phospholipids and compared their anti-septic effects on cells and in animal models. We found that sHDL made from DMPC presented the best anti-septic effects, possibly because DMPC-sHDL had the best fluidity at body temperature.


Subject(s)
Lipopolysaccharides , Phospholipids , Animals , Phospholipids/chemistry , Dimyristoylphosphatidylcholine , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
4.
Sci Signal ; 15(725): eabl9322, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35290084

ABSTRACT

Sepsis is a major health issue with mortality exceeding 30% and few treatment options. We found that high-density lipoprotein cholesterol (HDL-C) abundance was reduced by 45% in septic patients compared to that in nonseptic patients. Furthermore, HDL-C abundance in nonsurviving septic patients was substantially lower than in those patients who survived. We therefore hypothesized that replenishing HDL might be a therapeutic approach for treating sepsis and found that supplementing HDL with synthetic HDL (sHDL) provided protection against sepsis in mice. In mice subjected to cecal ligation and puncture (CLP), infusing the sHDL ETC-642 increased plasma HDL-C amounts and improved the 7-day survival rate. Septic mice treated with sHDL showed improved kidney function and reduced inflammation, as indicated by marked decreases in the plasma concentrations of blood urea nitrogen (BUN) and the cytokines interleukin-6 (IL-6) and IL-10, respectively. We found that sHDL inhibited the ability of the endotoxins LPS and LPA to activate inflammatory pathways in RAW264.7 cells and HEK-Blue cells expressing the receptors TLR4 or TLR2 and NF-κB reporters. In addition, sHDL inhibited the activation of HUVECs by LPS, LTA, and TNF-α. Together, these data indicate that sHDL treatment protects mice from sepsis in multiple ways and that it might be an effective therapy for patients with sepsis.


Subject(s)
Sepsis , Animals , Cytokines/metabolism , Humans , Interleukin-6/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Sepsis/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
5.
Mol Nutr Food Res ; 66(5): e2100615, 2022 03.
Article in English | MEDLINE | ID: mdl-34921741

ABSTRACT

SCOPE: Red meat intake is reported to be correlated with chronic diseases. A potential causal factor is N-glycolylneuraminic acid (Neu5Gc) which metabolically incorporates into diverse glycoconjugates in humans. This study aims to investigate the impact of exposure to Neu5Gc-rich red meat on healthy cytidine-5'-monophospho-N-acetylneuraminic acid hydroxylase (Cmah) knock-out mice and the underlying mechanisms. METHODS AND RESULTS: CMAH-/- mice are fed Neu5Gc-rich diet for short-term (4 months) and long-term (10 months). Health status and levels of inflammatory cytokines are assessed. Caco-2 cells are used to investigate the intestinal absorption of Neu5Gc-containing glycoprotein, and in vitro fermentation is used to investigate the Neu5Gc utilization by gut microbiota. Neu5Gc-rich diets show neither measurable abnormality in physio-biochemical and inflammatory indexes nor observable alterations of liver tissue in mice. Glycosylation of lactoferrin limits its intestinal epithelial absorption, and the absorption of Neu5Gc attach onto glycoprotein is thus limited. Neu5Gc is also simultaneously utilized by microorganisms under simulated gut conditions. CONCLUSION: The results indicate that the long-term intake of Neu5Gc-rich red meat has no adverse effect on the health of CMAH-/- mice, which may be related to the limited absorption of Neu5Gc that is regulated by protein glycosylation, and the metabolism of Neu5Gc by gut microorganisms.


Subject(s)
Gastrointestinal Microbiome , Animals , Caco-2 Cells , Glycoproteins/metabolism , Glycosylation , Humans , Mice , Mixed Function Oxygenases/metabolism
6.
Front Immunol ; 13: 1110516, 2022.
Article in English | MEDLINE | ID: mdl-36713379

ABSTRACT

Introduction: 25-60% of septic patients experience relative adrenal insufficiency (RAI) and glucocorticoid (GC) is frequently used in septic patients. However, the efficacy of GC therapy and whether GC therapy should be based on the status of RAI are highly controversial. Our poor understanding about the pathogenesis of RAI and a lack of RAI animal model present significant barriers to address these critical issues. Methods: Scavenger receptor BI (SR-BI) regulates stress-induced GC (iGC) production in response to stress. We generated SF1CreSR-BIfl/fl mice and utilized the mice as a RAI model to elucidate the pathogenesis of RAI and GC therapy in sepsis. SF1CreSR-BIfl/fl mice did not express SR-BI in adrenal gland and lacked iGC production upon ACTH stimulation, thus, they are RAI. Results and Discussion: RAI mice were susceptible to cecal ligation and puncture (CLP)-induced sepsis (6.7% survival in SF1CreSR-BIfl/fl mice versus 86.4% in SR-BIfl/fl mice; p = 0.0001). Compared to a well-controlled systemic inflammatory response in SR-BIfl/fl mice, SF1CreSR-BIfl/fl mice featured a persistent hyperinflammatory response. Supplementation of a low stress dose of GC to SF1CreSR-BIfl/fl mice kept the inflammatory response under control and rescued the mice. However, SR-BIfl/fl mice receiving GC treatment exhibited significantly less survival compared to SR-BIfl/fl mice without GC treatment. In conclusions, we demonstrated that RAI is a risk factor for death in this mouse model of sepsis. We further demonstrated that RAI is an endotype of sepsis, which features persistent hyperinflammatory response. We found that GC treatment benefits mice with RAI but harms mice without RAI. Our study provides a proof of concept to support a precision medicine approach for sepsis therapy - selectively applying GC therapy for a subgroup of patients with RAI.


Subject(s)
Adrenal Insufficiency , Sepsis , Animals , Mice , Adrenal Insufficiency/drug therapy , Adrenal Insufficiency/etiology , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Precision Medicine , Risk Factors , Sepsis/pathology
7.
Ultrasound Med Biol ; 47(3): 769-776, 2021 03.
Article in English | MEDLINE | ID: mdl-33358338

ABSTRACT

Thymus involution is characterized by a progressive regression of thymus size and contributes to immunosuppression in sepsis. High-frequency ultrasonography is a non-invasive monitoring system in multiple organs, including the thymus, in mice. However, thymus involution has not been studied using ultrasonography in septic mice. This study reports ultrasound approaches to monitoring septic thymus involution in mice. Sepsis was induced by cecum ligation and puncture (CLP). Mice were euthanized at three time points: baseline and days 3 and 10 after CLP. Thymus areas and volumes were measured using 2-D and 3-D ultrasound approaches. Thymus weights were measured ex vivo. Compared with values at baseline, both thymus area and volume decreased significantly at days 3 and 10. In addition, thymus areas and volumes correlated positively with thymus weights. In conclusion, ultrasonography provides reliable thymus measurements and is an optimal technique for monitoring thymus involution in septic mice.


Subject(s)
Lymphatic Diseases/complications , Lymphatic Diseases/diagnostic imaging , Sepsis/complications , Thymus Gland/diagnostic imaging , Animals , Male , Mice , Mice, Inbred C57BL , Ultrasonography
8.
Arterioscler Thromb Vasc Biol ; 40(8): 1830-1837, 2020 08.
Article in English | MEDLINE | ID: mdl-32522007

ABSTRACT

OBJECTIVE: Adrenal gland secretes stress-induced glucocorticoids (iGCs) to coping with stress. Previous study showed that SR-BI (scavenger receptor BI) null (SR-BI-/-) mice failed to generate iGC in stress conditions, suggesting that SR-BI-mediated cholesterol uptake from HDL (high-density lipoprotein) is a key regulator for iGC production. However, the LDL (low-density lipoprotein)/LDLr (LDL receptor) pathway can also provide cholesterol for iGC synthesis, but rodents have limited LDL levels in circulation. Here, we generated SR-BI-/-ApoBtg (apolipoprotein B transgenic) mice with normal LDL levels in circulation to determine the relative contribution of the HDL/SR-BI and LDL/LDLr pathways to iGC production in stress conditions. Approach and Results: To obtain mouse models with normal LDL levels, SR-BI-/- mice were bred to ApoBtg mice. Then, the F1 SR-BI±ApoBtg mice were backcrossed to SR-BI-/- to obtain SR-BI-/-ApoBtg, SR-BI-/-ApoBwt (apolipoprotein B wild type), and SR-BI+/+ApoBtg mice. We first examined the lipoprotein profile, which shows a 6.5-fold increase in LDL levels in SR-BI-/-ApoBtg mice compared with SR-BI-/-ApoBwt mice. Then, we induced stress with adrenocorticotropic hormone and cecal ligation and puncture. One hour after adrenocorticotropic hormone stimulation, SR-BI+/+ApoBtg control mice produced iGC (14.9-fold), but both SR-BI-/-ApoBwt and SR-BI-/-ApoBtg showed no iGC production (P<0.001). Three hours after cecal ligation and puncture treatment, SR-BI+/+ApoBtg control mice showed iGC production (6.4-fold), but both SR-BI-/-ApoBwt and SR-BI-/-ApoBtg mice showed no iGC production (P<0.001). CONCLUSIONS: SR-BI-/-ApoBtg mice fail to produce iGC in stress conditions even though with restored LDL levels in circulation. These findings clarify that the HDL/SR-BI, not LDL/LDLr, pathway is responsible for iGC production in stress conditions.


Subject(s)
Glucocorticoids/biosynthesis , Receptors, LDL/physiology , Scavenger Receptors, Class B/physiology , Stress, Psychological/metabolism , Adrenocorticotropic Hormone/pharmacology , Animals , Lipoproteins, LDL/blood , Mice , Mice, Inbred C57BL
9.
Immunity ; 50(6): 1401-1411.e4, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31076358

ABSTRACT

Inflammasome activation and subsequent pyroptosis are critical defense mechanisms against microbes. However, overactivation of inflammasome leads to death of the host. Although recent studies have uncovered the mechanism of pyroptosis following inflammasome activation, how pyroptotic cell death drives pathogenesis, eventually leading to death of the host, is unknown. Here, we identified inflammasome activation as a trigger for blood clotting through pyroptosis. We have shown that canonical inflammasome activation by the conserved type III secretion system (T3SS) rod proteins from Gram-negative bacteria or noncanonical inflammasome activation by lipopolysaccharide (LPS) induced systemic blood clotting and massive thrombosis in tissues. Following inflammasome activation, pyroptotic macrophages released tissue factor (TF), an essential initiator of coagulation cascades. Genetic or pharmacological inhibition of TF abolishes inflammasome-mediated blood clotting and protects against death. Our data reveal that blood clotting is the major cause of host death following inflammasome activation and demonstrate that inflammasome bridges inflammation with thrombosis.


Subject(s)
Blood Coagulation , Inflammasomes/metabolism , Pyroptosis , Thrombosis/etiology , Thrombosis/metabolism , Animals , Bacterial Infections/complications , Bacterial Infections/microbiology , Biomarkers , Caspases/metabolism , Cell-Derived Microparticles/immunology , Cell-Derived Microparticles/metabolism , Disease Models, Animal , Humans , Lipopolysaccharides/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Monocytes/immunology , Monocytes/metabolism , Signal Transduction , Thromboplastin/metabolism , Thrombosis/blood , Thrombosis/mortality
10.
Article in English | MEDLINE | ID: mdl-30555417

ABSTRACT

High-density lipoprotein (HDL) have long been studied for their protective role against cardiovascular diseases, however recently relationship between HDL and cancer came into focus. Several epidemiological studies have shown an inverse correlation between HDL-cholesterol (HDL-C) and cancer risk, and some have even implied that HDL-C can be used as a predictive measure for survival prognosis in for specific sub-population of certain types of cancer. HDL itself is an endogenous nanoparticle capable of removing excess cholesterol from the periphery and returning it to the liver for excretion. One of the main receptors for HDL, scavenger receptor type B-I (SR-BI), is highly upregulated in endocrine cancers, notably due to the high demand for cholesterol by cancer cells. Thus, the potential to exploit administration of cholesterol-free reconstituted or synthetic HDL (sHDL) to deplete cholesterol in endocrine cancer cell and stunt their growth of use chemotherapeutic drug loaded sHDL to target payload delivery to cancer cell has become increasingly attractive. This review focuses on the role of HDL and HDL-C in cancer and application of sHDLs as endocrine cancer therapeutics.

11.
Arterioscler Thromb Vasc Biol ; 38(11): 2706-2717, 2018 11.
Article in English | MEDLINE | ID: mdl-30354229

ABSTRACT

Objective- Continuous T-cell production from thymus is essential in replenishing naïve T-cell pool and maintaining optimal T-cell functions. However, the underlying mechanisms regulating the T-cell development in thymus remains largely unknown. Approach and Results- We identified SR-BI (scavenger receptor class B type 1), an HDL (high-density lipoprotein) receptor, as a novel modulator in T-cell development. We found that SR-BI deficiency in mice led to reduced thymus size and decreased T-cell production, which was accompanied by narrowed peripheral naïve T-cell pool. Further investigation revealed that SR-BI deficiency impaired progenitor thymic homing, causing a dramatic reduction in the percentage of earliest thymic progenitors, but did not affect other downstream T-cell developmental steps inside the thymus. As a result of the impaired progenitor thymic homing, SR-BI-deficient mice displayed delayed thymic regeneration postirradiation. Using a variety of experimental approaches, we revealed that the impaired T-cell development in SR-BI-deficient mice was not caused by hematopoietic SR-BI deficiency or SR-BI deficiency-induced hypercholesterolemia, but mainly attributed to the SR-BI deficiency in adrenal glands, as adrenal-specific SR-BI-deficient mice exhibited similar defects in T-cell development and thymic regeneration with SR-BI-deficient mice. Conclusions- This study demonstrates that SR-BI deficiency impaired T-cell development and delayed thymic regeneration by affecting progenitor thymic homing in mice, elucidating a previously unrecognized link between SR-BI and adaptive immunity.


Subject(s)
Adrenal Glands/metabolism , Cell Proliferation , Lymphocyte Activation , Regeneration , Scavenger Receptors, Class B/metabolism , T-Lymphocytes/metabolism , Thymocytes/metabolism , Thymus Gland/metabolism , Adaptive Immunity , Adrenal Glands/immunology , Animals , Cells, Cultured , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Leukocyte Common Antigens/deficiency , Leukocyte Common Antigens/genetics , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Receptors, LDL/deficiency , Receptors, LDL/genetics , Scavenger Receptors, Class B/deficiency , Scavenger Receptors, Class B/genetics , Signal Transduction , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/immunology , Thymocytes/immunology , Thymus Gland/immunology
12.
Pathol Res Pract ; 212(6): 555-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27067809

ABSTRACT

Scavenger receptor class B type 1 (SR-B1) is an integral membrane protein that is expressed in numerous cells and tissue types. The primary role of SR-B1 is to facilitate uptake of cholesteryl esters from high-density lipoproteins (HDL) in the liver. Altered SR-B1 expression contributes to human diseases. This study assessed association of SR-B1 expression in breast tissue specimens with breast cancer development and prognosis. Tissue specimens from 30 cases of adjacent normal breast tissues, ductal carcinoma in situ (DCIS) and invasive ductal breast cancer (IDCA) were subjected to Western blot analysis, and 135 cases of DCIS and IDCA were used for quantitative immunohistochemical analysis of SR-B1 expression. The data showed that SR-B1 was significantly overexpressed in IDCA tissues compared to normal breast and DCIS tissues. SR-B1 expression was associated with pre-menopausal status, tumor size, and worse overall survival of patients. The data from this ex vivo study suggests that up-regulated SR-B1 protein expression is associated with malignant behaviors of breast cancer and that SR-B1 is an independent predictor for poor survival in breast cancer patients.


Subject(s)
Breast Neoplasms/genetics , CD36 Antigens/genetics , Carcinoma, Ductal, Breast/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Gene Expression Regulation, Neoplastic , Up-Regulation , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/mortality , Carcinoma, Intraductal, Noninfiltrating/pathology , Female , Humans , Middle Aged , Prognosis , Survival Rate
13.
PLoS Biol ; 14(2): e1002375, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26863604

ABSTRACT

In Hedgehog (Hh) signaling, binding of Hh to the Patched-Interference Hh (Ptc-Ihog) receptor complex relieves Ptc inhibition on Smoothened (Smo). A longstanding question is how Ptc inhibits Smo and how such inhibition is relieved by Hh stimulation. In this study, we found that Hh elevates production of phosphatidylinositol 4-phosphate (PI(4)P). Increased levels of PI(4)P promote, whereas decreased levels of PI(4)P inhibit, Hh signaling activity. We further found that PI(4)P directly binds Smo through an arginine motif, which then triggers Smo phosphorylation and activation. Moreover, we identified the pleckstrin homology (PH) domain of G protein-coupled receptor kinase 2 (Gprk2) as an essential component for enriching PI(4)P and facilitating Smo activation. PI(4)P also binds mouse Smo (mSmo) and promotes its phosphorylation and ciliary accumulation. Finally, Hh treatment increases the interaction between Smo and PI(4)P but decreases the interaction between Ptc and PI(4)P, indicating that, in addition to promoting PI(4)P production, Hh regulates the pool of PI(4)P associated with Ptc and Smo.


Subject(s)
Drosophila Proteins/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , Hedgehog Proteins/metabolism , Phosphatidylinositol Phosphates/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cilia/metabolism , Drosophila , Mice , NIH 3T3 Cells , Patched Receptors , Patched-1 Receptor , Phosphorylation , Receptors, Cell Surface/metabolism , Smoothened Receptor
14.
Tumour Biol ; 37(3): 3581-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26456958

ABSTRACT

Scavenger receptor class B type I (SR-BI) has been linked to the development and progression of breast cancer. However, its clinical significance in breast cancer remains unclear. Here, we evaluated SR-BI expression in a well-characterized breast cancer tissue microarray by immunohistochemistry. High SR-BI expression was observed in 54 % of all breast cancer cases and was significantly associated with advanced pTNM stage (P = 0.002), larger tumor size (P = 0.023), lymph node metastasis (P = 0.012), and the absence of ER (P = 0.014). The Kaplan-Meier survival analysis revealed that patients with high SR-BI expression had significantly shorter overall survival (OS) (P = 0.004). Moreover, multivariate analysis with adjustment for other prognostic factors confirmed that SR-BI was an independent prognostic factor for patient outcome (P = 0.017). Overall, our study demonstrated that high SR-BI expression was related to conventional parameters indicative of more aggressive tumor type and may serve as a new prognostic marker for poor clinical outcome in human breast cancer.


Subject(s)
Biomarkers, Tumor/biosynthesis , Breast Neoplasms/metabolism , Scavenger Receptors, Class B/biosynthesis , Adult , Aged , Breast Neoplasms/pathology , Female , Follow-Up Studies , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymphatic Metastasis , Middle Aged , Multivariate Analysis , Neoplasm Staging , Prognosis , Proportional Hazards Models , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Tissue Array Analysis/methods , Tissue Array Analysis/statistics & numerical data , Tumor Burden
15.
Front Pharmacol ; 6: 244, 2015.
Article in English | MEDLINE | ID: mdl-26557091

ABSTRACT

High-density lipoprotein (HDL) is a key component of circulating blood and plays essential roles in regulation of vascular endothelial function and immunity. Clinical data demonstrate that HDL levels drop by 40-70% in septic patients, which is associated with a poor prognosis. Experimental studies using Apolipoprotein A-I (ApoAI) null mice showed that HDL deficient mice are susceptible to septic death, and overexpressing ApoAI in mice to increase HDL levels protects against septic death. These clinical and animal studies support our hypothesis that a decrease in HDL level is a risk factor for sepsis, and raising circulating HDL levels may provide an efficient therapy for sepsis. In this review, we discuss the roles of HDL in sepsis and summarize the efforts of using synthetic HDL as a potential therapy for sepsis.

16.
Crit Care Med ; 43(11): e490-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26308430

ABSTRACT

OBJECTIVES: Corticosteroid therapy is frequently used in septic patients given the rationale that there is an increased demand for corticosteroid in sepsis, and up to 60% of severe septic patients experience adrenal insufficiency. However, the efficacy of corticosteroid therapy and whether the therapy should be based on the results of adrenal function testing are highly controversial. The lack of an adrenal insufficiency animal model and our poor understanding of the pathogenesis caused by adrenal insufficiency present significant barriers to address this long-standing clinical issue. DESIGN: Prospective experimental study. SETTING: University laboratory. SUBJECTS: Scavenger receptor BI null and adrenal-specific scavenger receptor BI null mice. INTERVENTIONS: Sepsis was induced by cecal ligation and puncture. MEASUREMENTS AND MAIN RESULTS: Using scavenger receptor BI mice as the first relative adrenal insufficiency animal model, we found that corticosteroid therapy significantly improved the survival in cecal ligation and puncture-treated scavenger receptor BI mice but causes more septic death in wild-type mice. We identified a corticosteroid cocktail that provides effective protection 18 hours post cecal ligation and puncture; using adrenal-specific scavenger receptor BI mice as an inducible corticosteroid-deficient animal model, we found that inducible corticosteroid specifically suppresses interleukin-6 production without affecting tumor necrosis factor-α, nitric oxide, and interleukin-10 production. We further found that inducible corticosteroid does not induce peripheral lymphocyte apoptosis but promotes phagocytic activity of macrophages and neutrophils. CONCLUSIONS: This study demonstrates that corticosteroid treatment benefits mice with adrenal insufficiency but harms mice without adrenal insufficiency. This study also reveals that inducible corticosteroid has both immunosuppressive and immunopermissive properties, suppressing interleukin-6 production, promoting phagocytosis of immune effector cells, but not inducing peripheral lymphocyte apoptosis. These findings support our hypothesis that corticosteroid is an effective therapy for a subgroup of septic patients with adrenal insufficiency but harms septic patients without adrenal insufficiency and encourage further efforts to test this hypothesis in clinic.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Adrenal Insufficiency/drug therapy , Sepsis/drug therapy , Adrenal Insufficiency/physiopathology , Animals , Disease Models, Animal , Male , Mice , Mice, Knockout , Random Allocation , Risk Assessment , Sepsis/mortality , Sepsis/physiopathology , Survival Rate , Treatment Outcome
17.
J Biol Chem ; 290(25): 15496-15511, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25947382

ABSTRACT

Studies in human populations have shown a significant correlation between procollagen C-endopeptidase enhancer protein 2 (PCPE2) single nucleotide polymorphisms and plasma HDL cholesterol concentrations. PCPE2, a 52-kDa glycoprotein located in the extracellular matrix, enhances the cleavage of C-terminal procollagen by bone morphogenetic protein 1 (BMP1). Our studies here focused on investigating the basis for the elevated concentration of enlarged plasma HDL in PCPE2-deficient mice to determine whether they protected against diet-induced atherosclerosis. PCPE2-deficient mice were crossed with LDL receptor-deficient mice to obtain LDLr(-/-), PCPE2(-/-) mice, which had elevated HDL levels compared with LDLr(-/-) mice with similar LDL concentrations. We found that LDLr(-/-), PCPE2(-/-) mice had significantly more neutral lipid and CD68+ infiltration in the aortic root than LDLr(-/-) mice. Surprisingly, in light of their elevated HDL levels, the extent of aortic lipid deposition in LDLr(-/-), PCPE2(-/-) mice was similar to that reported for LDLr(-/-), apoA-I(-/-) mice, which lack any apoA-I/HDL. Furthermore, LDLr(-/-), PCPE2(-/-) mice had reduced HDL apoA-I fractional clearance and macrophage to fecal reverse cholesterol transport rates compared with LDLr(-/-) mice, despite a 2-fold increase in liver SR-BI expression. PCPE2 was shown to enhance SR-BI function by increasing the rate of HDL-associated cholesteryl ester uptake, possibly by optimizing SR-BI localization and/or conformation. We conclude that PCPE2 is atheroprotective and an important component of the reverse cholesterol transport HDL system.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Cholesterol Esters/metabolism , Glycoproteins/metabolism , Lipoproteins, HDL/metabolism , Scavenger Receptors, Class B/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Aorta/metabolism , Aorta/pathology , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Atherosclerosis/chemically induced , Atherosclerosis/genetics , Atherosclerosis/pathology , Biological Transport, Active/genetics , CHO Cells , Cholesterol Esters/genetics , Cricetulus , Glycoproteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Lipoproteins, HDL/genetics , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Receptors, LDL/genetics , Receptors, LDL/metabolism , Scavenger Receptors, Class B/genetics
18.
J Biol Chem ; 289(21): 14666-73, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24719333

ABSTRACT

Recent studies revealed that scavenger receptor BI (SR-BI or Scarb1) plays a critical protective role in sepsis. However, the mechanisms underlying this protection remain largely unknown. In this study, using Scarb1(I179N) mice, a mouse model specifically deficient in hepatic SR-BI, we report that hepatic SR-BI protects against cecal ligation and puncture (CLP)-induced sepsis as shown by 75% fatality in Scarb1(I179N) mice, but only 21% fatality in C57BL/6J control mice. The increase in fatality in Scarb1(I179N) mice was associated with an exacerbated inflammatory cytokine production. Further study demonstrated that hepatic SR-BI exerts its protection against sepsis through its role in promoting LPS clearance without affecting the inflammatory response in macrophages, the glucocorticoid production in adrenal glands, the leukocyte recruitment to peritoneum or the bacterial clearance in liver. Our findings reveal hepatic SR-BI as a critical protective factor in sepsis and point out that promoting hepatic SR-BI-mediated LPS clearance may provide a therapeutic approach for sepsis.


Subject(s)
Lipopolysaccharides/immunology , Liver/immunology , Scavenger Receptors, Class B/immunology , Sepsis/immunology , Adrenal Glands/immunology , Adrenal Glands/metabolism , Animals , Blotting, Western , Cecum/surgery , Cell Line , Cytokines/immunology , Cytokines/metabolism , Glucocorticoids/biosynthesis , Glucocorticoids/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Leukocytes/immunology , Leukocytes/metabolism , Ligation/adverse effects , Liver/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Peritoneum/immunology , Peritoneum/metabolism , Point Mutation , Punctures/adverse effects , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Sepsis/etiology , Sepsis/microbiology
19.
Arterioscler Thromb Vasc Biol ; 34(5): 966-75, 2014 May.
Article in English | MEDLINE | ID: mdl-24603680

ABSTRACT

OBJECTIVE: Thymocyte apoptosis is a major event in sepsis; however, how this process is regulated remains poorly understood. APPROACH AND RESULTS: Septic stress induces glucocorticoids production which triggers thymocyte apoptosis. Here, we used scavenger receptor BI (SR-BI)-null mice, which are completely deficient in inducible glucocorticoids in sepsis, to investigate the regulation of thymocyte apoptosis in sepsis. Cecal ligation and puncture induced profound thymocyte apoptosis in SR-BI(+/+) mice, but no thymocyte apoptosis in SR-BI(-/-) mice because of lack of inducible glucocorticoids. Unexpectedly, supplementation of glucocorticoids only partly restored thymocyte apoptosis in SR-BI(-/-) mice. We demonstrated that high-density lipoprotein (HDL) is a critical modulator for thymocyte apoptosis. SR-BI(+/+) HDL significantly enhanced glucocorticoid-induced thymocyte apoptosis, but SR-BI(-/-) HDL had no such activity. Further study revealed that SR-BI(+/+) HDL modulates glucocorticoid-induced thymocyte apoptosis via promoting glucocorticoid receptor translocation, but SR-BI(-/-) HDL loses such regulatory activity. To understand why SR-BI(-/-) HDL loses its regulatory activity, we analyzed HDL cholesterol contents. There was 3-fold enrichment of unesterified cholesterol in SR-BI(-/-) HDL compared with SR-BI(+/+) HDL. Normalization of unesterified cholesterol in SR-BI(-/-) HDL by probucol administration or lecithin cholesteryl acyltransferase expression restored glucocorticoid-induced thymocyte apoptosis, and incorporating unesterified cholesterol into SR-BI(+/+) HDL rendered SR-BI(+/+) HDL dysfunctional. Using lckCre-GR(fl/fl) mice in which thymocytes lack cecal ligation and puncture-induced thymocyte apoptosis, we showed that lckCre-GR(fl/fl) mice were significantly more susceptible to cecal ligation and puncture-induced septic death than GR(fl/fl) control mice, suggesting that glucocorticoid-induced thymocyte apoptosis is required for protection against sepsis. CONCLUSIONS: The findings in this study reveal a novel regulatory mechanism of thymocyte apoptosis in sepsis by SR-BI and HDL.


Subject(s)
Apoptosis , Cholesterol, HDL/blood , Scavenger Receptors, Class B/metabolism , Sepsis/metabolism , Thymocytes/metabolism , Animals , Apoptosis/drug effects , Cecum/microbiology , Cecum/surgery , Cells, Cultured , Corticosterone/metabolism , Disease Models, Animal , Female , Humans , Ligation , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Probucol/pharmacology , Protein Transport , Punctures , Receptors, Glucocorticoid/metabolism , Scavenger Receptors, Class B/deficiency , Scavenger Receptors, Class B/genetics , Sepsis/blood , Sepsis/microbiology , Sepsis/pathology , Signal Transduction , Thymocytes/drug effects , Thymocytes/pathology
20.
Shock ; 41(6): 463-75, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24569509

ABSTRACT

Many preclinical studies in critical care medicine and related disciplines rely on hypothesis-driven research in mice. The underlying premise posits that mice sufficiently emulate numerous pathophysiologic alterations produced by trauma/sepsis and can serve as an experimental platform for answering clinically relevant questions. Recently, the lay press severely criticized the translational relevance of mouse models in critical care medicine. A series of provocative editorials were elicited by a highly publicized research report in the Proceedings of the National Academy of Sciences (PNAS; February 2013), which identified an unrecognized gene expression profile mismatch between human and murine leukocytes following burn/trauma/endotoxemia. Based on their data, the authors concluded that mouse models of trauma/inflammation are unsuitable for studying corresponding human conditions. We believe this conclusion was not justified. In conjunction with resulting negative commentary in the popular press, it can seriously jeopardize future basic research in critical care medicine. We will address some limitations of that PNAS report to provide a framework for discussing its conclusions and attempt to present a balanced summary of strengths/weaknesses of use of mouse models. While many investigators agree that animal research is a central component for improved patient outcomes, it is important to acknowledge known limitations in clinical translation from mouse to man. The scientific community is responsible to discuss valid limitations without overinterpretation. Hopefully, a balanced view of the strengths/weaknesses of using animals for trauma/endotoxemia/critical care research will not result in hasty discount of the clear need for using animals to advance treatment of critically ill patients.


Subject(s)
Genomics , Inflammation/genetics , Animals , Female , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...