Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Antibiotics (Basel) ; 13(3)2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38534707

ABSTRACT

Over the past century, antibiotic usage has skyrocketed in the treatment of critically ill patients. There have been increasing calls to establish guidelines for appropriate treatment and durations of antibiosis. Antibiotic treatment, even when appropriately tailored to the patient and infection, is not without cost. Short term risks-hepatic/renal dysfunction, intermediate effects-concomitant superinfections, and long-term risks-potentiating antimicrobial resistance (AMR), are all possible consequences of antimicrobial administration. These risks are increased by longer periods of treatment and unnecessarily broad treatment courses. Recently, the literature has focused on multiple strategies to determine the appropriate duration of antimicrobial therapy. Further, there is a clinical shift to multi-modal approaches to determine the most suitable timepoint at which to end an antibiotic course. An approach utilising biomarker assays and an inter-disciplinary team of pharmacists, nurses, physicians, and microbiologists appears to be the way forward to develop sound clinical decision-making surrounding antibiotic treatment.

4.
FASEB J ; 34(3): 4041-4054, 2020 03.
Article in English | MEDLINE | ID: mdl-31930569

ABSTRACT

Recent evidence indicates that elevated placental adenosine signaling contributes to preeclampsia (PE). However, the molecular basis for the chronically enhanced placental adenosine signaling in PE remains unclear. Here, we report that hypoxia-inducible factor-1α (HIF-1α) is crucial for the enhancement of placental adenosine signaling. Utilizing a pharmacologic approach to reduce placental adenosine levels, we found that enhanced adenosine underlies increased placental HIF-1α in an angiotensin receptor type 1 receptor agonistic autoantibody (AT1 -AA)-induced mouse model of PE. Knockdown of placental HIF-1α in vivo suppressed the accumulation of adenosine and increased ecto-5'-nucleotidase (CD73) and adenosine A2B receptor (ADORA2B) in the placentas of PE mouse models induced by AT1 -AA or LIGHT, a TNF superfamily cytokine (TNFSF14). Human in vitro studies using placental villous explants demonstrated that increased HIF-1α resulting from ADORA2B activation facilitates the induction of CD73, ADORA2B, and FLT-1 expression. Overall, we demonstrated that (a) elevated placental HIF-1α by AT1 -AA or LIGHT upregulates CD73 and ADORA2B expression and (b) enhanced adenosine signaling through upregulated ADORA2B induces placental HIF-1α expression, which creates a positive feedback loop that promotes FLT-1 expression leading to disease development. Our results suggest that adenosine-based therapy targeting the malicious cycle of placental adenosine signaling may elicit therapeutic effects on PE.


Subject(s)
Adenosine/metabolism , Autoantibodies/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , RNA, Small Interfering/metabolism , Animals , Autoantibodies/genetics , Blood Pressure/genetics , Blood Pressure/physiology , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Pre-Eclampsia/genetics , Pregnancy , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
5.
Sci Rep ; 6: 29637, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27436223

ABSTRACT

Although Lands' cycle was discovered in 1958, its function and cellular regulation in membrane homeostasis under physiological and pathological conditions remain largely unknown. Nonbiased high throughput metabolomic profiling revealed that Lands' cycle was impaired leading to significantly elevated erythrocyte membrane lysophosphatidylcholine (LysoPC) content and circulating and erythrocyte arachidonic acid (AA) in mice with sickle cell disease (SCD), a prevalent hemolytic genetic disorder. Correcting imbalanced Lands' cycle by knockdown of phospholipase 2 (cPLA2) or overexpression of lysophosphatidycholine acyltransferase 1 (LPCAT1), two key enzymes of Lands' cycle in hematopoietic stem cells, reduced elevated erythrocyte membrane LysoPC content and circulating AA levels and attenuated sickling, inflammation and tissue damage in SCD chimeras. Human translational studies validated SCD mouse findings and further demonstrated that imbalanced Lands' cycle induced LysoPC production directly promotes sickling in cultured mouse and human SCD erythrocytes. Mechanistically, we revealed that hypoxia-mediated ERK activation underlies imbalanced Lands' cycle by preferentially inducing the activity of PLA2 but not LPCAT in human and mouse SCD erythrocytes. Overall, our studies have identified a pathological role of imbalanced Lands' cycle in SCD erythrocytes, novel molecular basis regulating Lands' cycle and therapeutic opportunities for the disease.


Subject(s)
Anemia, Sickle Cell/metabolism , Arachidonic Acid/blood , Erythrocytes/metabolism , Lysophosphatidylcholines/metabolism , Metabolomics/methods , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Animals , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Female , Gene Knockdown Techniques , Group IV Phospholipases A2/genetics , Humans , Male , Mice
6.
J Am Heart Assoc ; 5(7)2016 06 30.
Article in English | MEDLINE | ID: mdl-27364991

ABSTRACT

BACKGROUND: The pathogenesis of essential hypertension is multifactorial with different underlying mechanisms contributing to disease. We have recently shown that TNF superfamily member 14 LIGHT (an acronym for homologous to lymphotoxins, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes, also known as TNFSF14) induces hypertension when injected into mice. Research reported here was undertaken to examine the role of transglutaminase (TGase) in LIGHT-induced hypertension. METHODS AND RESULTS: Initial experiments showed that plasma and kidney TGase activity was induced by LIGHT infusion (13.91±2.92 versus 6.75±1.92 mU/mL and 19.86±3.55 versus 12.00±0.97 mU/10 µg) and was accompanied with hypertension (169±7.16 versus 117.17±11.57 mm Hg at day 14) and renal impairment (proteinuria, 61.33±23.21 versus 20.38±9.01 µg/mg; osmolality, 879.57±93.02 versus 1407.2±308.04 mmol/kg). The increase in renal TGase activity corresponded to an increase in RNA for the tissue TGase isoform, termed TG2. Pharmacologically, we showed that LIGHT-induced hypertension and renal impairment did not occur in the presence of cystamine, a well-known competitive inhibitor of TGase activity. Genetically, we showed that LIGHT-mediated induction of TGase, along with hypertension and renal impairment, was dependent on interleukin-6 and endothelial hypoxia inducible factor-1α. We also demonstrated that interleukin-6, endothelial hypoxia inducible factor-1α, and TGase are required for LIGHT-induced production of angiotensin receptor agonistic autoantibodies. CONCLUSIONS: Thus, LIGHT-induced hypertension, renal impairment, and production of angiotensin receptor agonistic autoantibodies require TGase, most likely the TG2 isoform. Our findings establish TGase as a critical link between inflammation, hypertension, and autoimmunity.


Subject(s)
Blood Pressure/drug effects , Hypertension/immunology , Inflammation/immunology , Transglutaminases/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Animals , Autoantibodies/immunology , Blood Pressure/immunology , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Hypertension/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/immunology , Kidney/immunology , Kidney/metabolism , Mice , Mice, Knockout , Protein Glutamine gamma Glutamyltransferase 2 , Proteinuria/immunology , Proteinuria/metabolism , Receptors, Angiotensin/immunology , Renal Insufficiency/immunology , Renal Insufficiency/metabolism , Transglutaminases/genetics , Transglutaminases/metabolism
7.
Clin Immunol ; 168: 64-71, 2016 07.
Article in English | MEDLINE | ID: mdl-27181993

ABSTRACT

To characterize antibody specificities associated with pre-eclampsia (PE), bacterial displayed peptide library screening and evolution was applied to identify peptide epitopes recognized by plasma antibodies present in women with PE near the time of delivery. Pre-eclamptic women exhibited elevated IgG1 titers towards a peptide epitope KRPSCIGCK within the Epstein-Barr virus nuclear antigen 1 (EBNA-1). EBNA-1 epitope antibodies cross-reacted with a similar epitope within the extracellular N-terminus of the human G protein-coupled receptor, GPR50, expressed in human placental tissue and immortalized placental trophoblast cells. We observed increased antibody binding activity to epitopes from EBNA-1 and GPR50 among women with PE (n=42) compared to healthy-outcome pregnancies (n=43) and nulligravid samples (n=21). The EBNA-1 peptide potently blocked binding of the PE-associated antibody to the GPR50 epitope (IC50=58-81pM). These results reveal the existence of molecular mimicry between EBNA-1 and placental GPR50, supporting a mechanism for IgG1 deposition in the pre-eclamptic placenta.


Subject(s)
Antibodies, Viral/immunology , Herpesvirus 4, Human/immunology , Nerve Tissue Proteins/immunology , Placenta/immunology , Pre-Eclampsia/immunology , Receptors, G-Protein-Coupled/immunology , Adult , Amino Acid Sequence , Antibodies, Viral/metabolism , Cross Reactions/immunology , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Epitopes/metabolism , Epstein-Barr Virus Nuclear Antigens/immunology , Epstein-Barr Virus Nuclear Antigens/metabolism , Female , HEK293 Cells , Herpesvirus 4, Human/metabolism , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunohistochemistry , Nerve Tissue Proteins/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Pregnancy , Protein Binding/immunology , Receptors, G-Protein-Coupled/metabolism
8.
J Am Heart Assoc ; 4(12)2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26675250

ABSTRACT

BACKGROUND: Preeclampsia (PE) is a life-threatening hypertensive disorder of pregnancy associated with autoantibodies, termed AT1-AA, that activate the AT1 angiotensin receptor. Although the pathogenic nature of these autoantibodies has been extensively studied, little is known about the molecular cause of their generation. METHODS AND RESULTS: Here we show that tissue transglutaminase (TG2), an enzyme that conducts posttranslational modification of target proteins, directly modified the 7-amino acid (7-aa) epitope peptide that localizes to the second extracellular loop of the AT1 receptor. These findings led us to further discover that plasma transglutaminase activity was induced and contributed to the production of AT1-AA and disease development in an experimental model of PE induced by injection of LIGHT, a tumor necrosis factor superfamily member. Key features of PE were regenerated by adoptive transfer of purified IgG from LIGHT-injected pregnant mice and blocked by the 7-amino acid epitope peptide. Translating our mouse research to humans, we found that plasma transglutaminase activity was significantly elevated in PE patients and was positively correlated with AT1-AA levels and PE features. CONCLUSIONS: Overall, we provide compelling mouse and human evidence that elevated transglutaminase underlies AT1-AA production in PE and highlight novel pathogenic biomarkers and innovative therapeutic possibilities for the disease.


Subject(s)
Autoantibodies/immunology , GTP-Binding Proteins/physiology , Pre-Eclampsia/etiology , Receptors, Angiotensin/physiology , Transglutaminases/physiology , Adult , Animals , Biomarkers/blood , Blotting, Western , Epitopes/immunology , Female , GTP-Binding Proteins/blood , Humans , Mice , Mice, Inbred C57BL , Pre-Eclampsia/physiopathology , Pregnancy , Protein Glutamine gamma Glutamyltransferase 2 , Receptor, Angiotensin, Type 1/physiology , Transglutaminases/blood
10.
Hypertension ; 65(6): 1307-15, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25847948

ABSTRACT

Accumulation of hypoxia inducible factor-1α (HIF-1α) is commonly an acute and beneficial response to hypoxia, whereas chronically elevated HIF-1α is associated with multiple disease conditions, including preeclampsia, a serious hypertensive disease of pregnancy. However, the molecular basis underlying the persistent elevation of placental HIF-1α in preeclampsia and its role in the pathogenesis of preeclampsia are poorly understood. Here we report that Hif-1α mRNA and HIF-1α protein were elevated in the placentas of pregnant mice infused with angiotensin II type I receptor agonistic autoantibody, a pathogenic factor in preeclampsia. Knockdown of placental Hif-1α mRNA by specific siRNA significantly attenuated hallmark features of preeclampsia induced by angiotensin II type I receptor agonistic autoantibody in pregnant mice, including hypertension, proteinuria, kidney damage, impaired placental vasculature, and elevated maternal circulating soluble fms-like tyrosine kinase-1 levels. Next, we discovered that Hif-1α mRNA levels and HIF-1α protein levels were induced in an independent preeclampsia model with infusion of the inflammatory cytokine tumor necrosis factor superfamily member 14 (LIGHT). SiRNA knockdown experiments also demonstrated that elevated HIF-1α contributed to LIGHT-induced preeclampsia features. Translational studies with human placentas showed that angiotensin II type I receptor agonistic autoantibody or LIGHT is capable of inducing HIF-1α in a hypoxia-independent manner. Moreover, increased HIF-1α was found to be responsible for angiotensin II type I receptor agonistic autoantibody or LIGHT-induced elevation of Flt-1 gene expression and production of soluble fms-like tyrosine kinase-1 in human villous explants. Overall, we demonstrated that hypoxia-independent stimulation of HIF-1α gene expression in the placenta is a common pathogenic mechanism promoting disease progression. Our findings reveal new insight to preeclampsia and highlight novel therapeutic possibilities for the disease.


Subject(s)
Fetal Hypoxia/physiopathology , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Placenta/metabolism , Pre-Eclampsia/therapy , Pregnancy, Animal , Animals , Autoantibodies/administration & dosage , Disease Models, Animal , Female , Humans , Mice , Placenta/physiopathology , Pre-Eclampsia/physiopathology , Pregnancy , RNA, Messenger/analysis , RNA, Small Interfering/analysis , Random Allocation , Receptor, Angiotensin, Type 2/administration & dosage , Sensitivity and Specificity , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Up-Regulation
11.
Hypertension ; 65(2): 430-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25452470

ABSTRACT

C-reactive protein (CRP), an innate immune mediator, is elevated in the circulation before symptoms in patients with preeclampsia, a severe hypertensive pregnancy disorder with high mortality and morbidity. However, the specific sources underlying increased CRP and the role of elevated CRP in preeclampsia are undefined. Here, we report that circulating CRP levels are significantly increased in a large cohort of normotensive pregnant individuals when compared with nulligravid women and is further increased in patients with preeclampsia. These findings led us to discover further that placental syncytiotrophoblasts are previously unrecognized cellular sources of CRP and underlie elevated CRP in normotensive pregnant women and the additional increase in patients with preeclampsia. Next, we demonstrated that injection of CRP induces preeclampsia features, including hypertension (157 mm Hg CRP treated versus 119 mm Hg control), proteinuria (35.0 mg/µg CRP treated versus 14.1 mg/µg control), kidney, and placental damage and increased levels of sFlt-1 in pregnant mice but not in nonpregnant mice. Our study implicates that phosphocholine transferase, a placental-specific enzyme post-translationally modifying neurokinin B, is essential for the pathogenic role of CRP in preeclampsia through activation of the neurokinin 3 receptor. Overall, our studies have provided significant new insight on the pathogenic role of CRP in preeclampsia and highlighted innovative therapeutic strategies.


Subject(s)
C-Reactive Protein/physiology , Choline-Phosphate Cytidylyltransferase/physiology , Neurokinin B/metabolism , Pre-Eclampsia/etiology , Receptors, Neurokinin-3/physiology , Animals , Biomarkers , C-Reactive Protein/analysis , C-Reactive Protein/toxicity , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Disease Models, Animal , Double-Blind Method , Female , Humans , Kidney/pathology , Mice , Mice, Inbred C57BL , Phosphorylation , Phosphorylcholine/metabolism , Placenta/pathology , Pre-Eclampsia/chemically induced , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , Protein Binding , Protein Processing, Post-Translational , Quinolines/pharmacology , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Receptors, Neurokinin-3/antagonists & inhibitors , Receptors, Neurokinin-3/metabolism , Recombinant Proteins/toxicity , Single-Blind Method , Vascular Endothelial Growth Factor Receptor-1/blood
12.
Circulation ; 131(8): 730-41, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25538227

ABSTRACT

BACKGROUND: Preeclampsia is a prevalent hypertensive disorder of pregnancy and a leading cause of maternal and neonatal morbidity and mortality worldwide. This pathogenic condition is speculated to be caused by placental abnormalities that contribute to the maternal syndrome. However, the specific factors and signaling pathways that lead to impaired placentas and maternal disease development remain elusive. METHODS AND RESULTS: Using 2 independent animal models of preeclampsia (genetically engineered pregnant mice with elevated adenosine exclusively in placentas and a pathogenic autoantibody-induced preeclampsia mouse model), we demonstrated that chronically elevated placental adenosine was sufficient to induce hallmark features of preeclampsia, including hypertension, proteinuria, small fetuses, and impaired placental vasculature. Genetic and pharmacological approaches revealed that elevated placental adenosine coupled with excessive A2B adenosine receptor (ADORA2B) signaling contributed to the development of these features of preeclampsia. Mechanistically, we provided both human and mouse evidence that elevated placental CD73 is a key enzyme causing increased placental adenosine, thereby contributing to preeclampsia. CONCLUSIONS: We determined that elevated placental adenosine signaling is a previously unrecognized pathogenic factor for preeclampsia. Moreover, our findings revealed the molecular basis underlying the elevation of placental adenosine and the detrimental role of excess placental adenosine in the pathophysiology of preeclampsia, and thereby, we highlight novel therapeutic targets.


Subject(s)
Adenosine/metabolism , Placenta/metabolism , Pre-Eclampsia/etiology , Pre-Eclampsia/physiopathology , Signal Transduction/physiology , Up-Regulation/physiology , 5'-Nucleotidase/metabolism , Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Adult , Animals , Autoantibodies/adverse effects , Disease Models, Animal , Female , Fetal Growth Retardation/etiology , Fetal Growth Retardation/physiopathology , Gene Deletion , Humans , Mice, Knockout , Pre-Eclampsia/chemically induced , Pregnancy , Receptor, Adenosine A2B/genetics , Receptor, Adenosine A2B/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
13.
Hypertension ; 63(5): 1086-93, 2014 May.
Article in English | MEDLINE | ID: mdl-24446060

ABSTRACT

The presence of maternal autoantibodies has been previously associated with preeclampsia, although the composition of the antibody repertoire in preeclampsia has not been well characterized. Given this, we applied a bacterial display peptide library to identify peptides that preferentially react with plasma antibodies from patients with preeclampsia (n=15) versus healthy-outcome pregnancies (n=18). Screening using fluorescence-activated cell sorting identified 38 peptides that preferentially bind to antibodies from individuals with preeclampsia. These preeclampsia-specific peptides possessed similar motifs of R(G)/S(G)/-WW(G)/S, RWW(G)/S, or WGWGXX(R)/K distinct from the angiotensin II type 1 receptor epitope AFHYESQ. Seven library-isolated peptides and a cell surface-displayed angiotensin II type 1 receptor epitope were used to construct a diagnostic algorithm with a training set of 18 new preeclamptic and 22 healthy-outcome samples from geographically distinct cohorts. Cross-validation within the training group resulted in averaged areas underneath a receiver operating characteristic curve of 0.78 and 0.72 with and without the known receptor epitope, respectively. In a small validation set (12 preeclamptic; 8 healthy), the algorithm consisting only of library-isolated peptides correctly classified 10 preeclamptic and 6 healthy samples using a predefined cutoff that achieved 61% sensitivity (95% confidence interval, 36%-83%) at 95% specificity (95% confidence interval, 77%-100%) in training set (n=40) cross-validation. Our results indicate that antibodies with specificities other than anti-angiotensin II type 1 receptor are prevalent in preeclampsia patients and may be useful as diagnostic biomarkers.


Subject(s)
Antibodies/immunology , Antibody Specificity/immunology , Pre-Eclampsia/immunology , Adult , Algorithms , Antibodies/blood , Biomarkers , Case-Control Studies , Epitopes/immunology , Female , Humans , Pregnancy , Receptor, Angiotensin, Type 1/immunology
14.
Hypertension ; 63(3): 595-606, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24324043

ABSTRACT

Preeclampsia, a prevalent hypertensive disorder of pregnancy, is believed to be secondary to uteroplacental ischemia. Accumulating evidence indicates that hypoxia-independent mediators, including inflammatory cytokines and growth factors, are associated with preeclampsia, but it is unclear whether these signals directly contribute to placental damage and disease development in vivo. We report that LIGHT, a novel tumor necrosis factor superfamily member, is significantly elevated in the circulation and placentas of preeclamptic women compared with normotensive pregnant women. Injection of LIGHT into pregnant mice induced placental apoptosis, small fetuses, and key features of preeclampsia, hypertension and proteinuria. Mechanistically, using neutralizing antibodies specific for LIGHT receptors, we found that LIGHT receptors herpes virus entry mediator and lymphotoxin ß receptor are required for LIGHT-induced placental impairment, small fetuses, and preeclampsia features in pregnant mice. Accordingly, we further revealed that LIGHT functions through these 2 receptors to induce secretion of soluble fms-like tyrosine kinase-1 and endothelin-1, 2 well-accepted pathogenic factors in preeclampsia, and thereby plays an important role in hypertension and proteinuria in pregnant mice. Lastly, we extended our animal findings to human studies and demonstrated that activation of LIGHT receptors resulted in increased apoptosis and elevation of soluble fms-like tyrosine kinase-1 secretion in human placental villous explants. Overall, our human and mouse studies show that LIGHT signaling is a previously unrecognized pathway responsible for placental apoptosis, elevated secretion of vasoactive factors, and subsequent maternal features of preeclampsia, and reveal new therapeutic opportunities for the management of the disease.


Subject(s)
Endothelins/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Pregnancy, Animal , Proteinuria/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Adult , Animals , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Pre-Eclampsia/physiopathology , Pregnancy , Proteinuria/etiology , Signal Transduction
15.
Hypertension ; 63(2): 353-61, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24191290

ABSTRACT

Preeclampsia is a life-threatening pregnancy disorder that is widely thought to be triggered by impaired placental development. However, the placenta-related pathogenic factors are not fully identified, and their underlying mechanisms in disease development remain unclear. Here, we report that the protein level and enzyme activity of tissue transglutaminase (TG2 or tTG), the most ubiquitous member of a family of enzymes that conducts post-translational modification of proteins by forming ε-(γ-glutamyl)-lysine isopeptide bonds, are significantly elevated in placentas of preeclamptic women. TG2 is localized in the placental syncytiotrophoblasts of patients with preeclampsia where it catalyzes the isopeptide modification of the angiotensin receptor type 1 (AT1). To determine the role of elevated TG2 in preeclampsia, we used a mouse model of preeclampsia based on injection of AT1-agonistic autoantibody. A pathogenic role for TG2 in preeclampsia is suggested by in vivo experiments in which cystamine, a potent transglutaminase inhibitor, or small interfering RNA-mediated TG2 knockdown significantly attenuated autoantibody-induced hypertension and proteinuria in pregnant mice. Cystamine treatment also prevented isopeptide modification of placental AT1 receptors in preeclamptic mice. Mechanistically, we revealed that AT1-agonistic autoantibody stimulation enhances the interaction between AT1 receptor and TG2 and results in increased AT1 receptor stabilization via transglutaminase-mediated isopeptide modification in trophoblasts. Mutagenesis studies further demonstrated that TG2-mediated isopeptide modification of AT1 receptors prevents ubiquitination-dependent receptor degradation. Taken together, our studies not only identify a novel pathogenic involvement of TG2 in preeclampsia but also suggest a previously unrecognized role of TG2 in the regulation of G protein-coupled receptor stabilization by inhibiting ubiquitination-dependent degradation.


Subject(s)
Dipeptides/metabolism , GTP-Binding Proteins/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Receptor, Angiotensin, Type 1/metabolism , Transglutaminases/metabolism , Animals , Cell Line , Disease Models, Animal , Female , GTP-Binding Proteins/genetics , Humans , Mice , Mutagenesis/physiology , Placenta/pathology , Pre-Eclampsia/pathology , Pregnancy , Protein Glutamine gamma Glutamyltransferase 2 , Protein Processing, Post-Translational/physiology , RNA, Small Interfering/genetics , Transglutaminases/genetics , Trophoblasts/cytology , Trophoblasts/metabolism , Ubiquitination/physiology
16.
BMC Cancer ; 13: 314, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23806209

ABSTRACT

BACKGROUND: Circulating tumor cells (CTCs) detection has previously been used for diagnosing gastric cancer. However, the previous studies failed to make an agreement whether the detection of CTCs contributes to the diagnosis of gastric cancer. METHODS: A systematic review and meta-analysis was performed to evaluate the overall accuracy of CTCs detection for diagnosing gastric cancer. PubMed, Embase and the Wanfang database were searched in all languages published up to Oct 2012. The pooled sensitivity (SEN), specificity (SPE), positive and negative likelihood ratios (PLR and NLR, respectively), diagnostic odds ratio (DOR) and summary receiver operating characteristic (sROC) curve were calculated to evaluate the overall test performance. RESULTS: Twenty studies were included in this systematic review and meta-analysis. The diagnostic value of CTCs detection for the gastric cancer was calculated to evaluate the overall test performance. The summary estimates of The pooled sensitivity, specificity, positive and negative likelihood ratios, diagnostic odds ratio were 0.42 (95% confidence interval (CI), 0.21-0.67), 0.99 (95% CI, 0.96-1.00), 58.2 (95% CI, 9.8-345.9), 0.58 (95% CI, 0.38-0.89), and 100 (95% CI, 15-663), respectively. The summary receiver operating characteristic curve was 0.97 (95% CI, 0.95-0.98). Deek's funnel plot asymmetry test found no evidence of study publication bias in the current study (P = 0.49). CONCLUSION: This systematic review suggests that CTCs detection alone cannot be recommended as a screening test for gastric cancer. However, it might be used as a noninvasive method for the confirmation of the gastric cancer diagnosis.


Subject(s)
Neoplastic Cells, Circulating , Stomach Neoplasms/diagnosis , Area Under Curve , Confidence Intervals , Humans , Likelihood Functions , Odds Ratio , ROC Curve , Stomach Neoplasms/blood
17.
Dev Growth Differ ; 51(2): 123-33, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19207183

ABSTRACT

Patterning of the developing vertebrate limb along the anterior-posterior axis is controlled by the zone of polarizing activity (ZPA) via the expression of Sonic hedgehog (Shh) and along the proximal-distal axis by the apical ectodermal ridge (AER) through the production of fibroblast growth factors (FGFs). ZPA grafting, as well as ectopic application of SHH to the anterior chick limb bud, demonstrate that digit patterning is largely influenced by these secreted factors. Although signal transduction pathways have been well characterized for SHH and for FGFs, little is known of how these signals are regulated extracellularly in the limb. The present study shows that alteration of the extracellular environment through trypsin treatment can have profound effects on digit patterning. These effects appear to be mediated by the induction of Shh in host tissues and by ectopic AER formation, implicating the extracellular matrix in regulating the signaling activities of key patterning genes in the limb.


Subject(s)
Body Patterning/drug effects , Chick Embryo/embryology , Gene Expression Regulation, Developmental , Hedgehog Proteins/physiology , Limb Buds/embryology , Trypsin/pharmacology , Animals , Body Patterning/physiology , Chick Embryo/abnormalities , Collagenases/pharmacology , Ectoderm/physiology , Ectoderm/transplantation , Embryonic Induction , Extracellular Matrix/physiology , Female , Fibrinolysin/pharmacology , Fibroblast Growth Factors/physiology , Limb Buds/abnormalities , Limb Buds/drug effects , Limb Buds/metabolism , Limb Buds/transplantation , Male , Mice , Morphogenesis/drug effects , Pancreas/embryology , Signal Transduction , Transplantation, Heterologous , Transplantation, Heterotopic , Wings, Animal/abnormalities , Wings, Animal/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...