Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters











Publication year range
1.
Cardiovasc Toxicol ; 21(10): 808-819, 2021 10.
Article in English | MEDLINE | ID: mdl-34173191

ABSTRACT

Pentraxin 3 (PTX3) is synthesized locally and released into the circulation, reflecting local inflammation in the cardiovascular system. Therefore, we conducted a study to explore the effect of PTX3 in spontaneously hypertensive heart failure (SHHF) rats. Sprague Dawley (SD) and SHHF rats were treated with recombinant PTX3 protein, and the blood pressure (BP) and echocardiographic parameters were collected. Radioimmunoassay, enzyme immunoassay and enzyme-linked immunosorbent assay (ELISA) were applied to detect plasma levels of atrial/B-type natriuretic peptide (ANP/BNP) and PTX3. The pathological changes in the myocardial tissues were observed by hematoxylin and eosin (HE) and Masson stainings. The mRNA and protein expressions were detected by quantitative real-time reverse-transcription polymerase chain reaction (qPCR) and western blotting. Cardiomyocyte apoptosis was evaluated by TUNEL staining and DNA fragmentation test. Increased plasma concentrations of PTX3 were found in SHHF rats compared with SD rats, which was further enhanced by recombinant PTX3 protein. After injection with recombinant PTX3 protein, the heart function was improved in SHHF rats with the decreased systolic and diastolic BP, and the reduced plasma levels of ANP and BNP. Moreover, PTX3 improved the myocardial damage and interstitial fibrosis in SHHF rats with reduced cardiomyocyte apoptosis and decreased mRNA expressions of pro-inflammatory factors in myocardial tissues. PTX3 could decrease the BP and plasma levels of ANP and BNP in SHHF rats, as well as improve the inflammation, cardiomyocyte apoptosis, and pathological changes of myocardial tissues, suggesting it may be a useful intervention in the treatment of SHHF.


Subject(s)
Apoptosis/drug effects , Blood Pressure/drug effects , C-Reactive Protein/pharmacology , Heart Failure/drug therapy , Hypertension/drug therapy , Myocytes, Cardiac/drug effects , Serum Amyloid P-Component/pharmacology , Animals , Atrial Natriuretic Factor/blood , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Heart Failure/metabolism , Heart Failure/physiopathology , Hypertension/metabolism , Hypertension/physiopathology , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/blood , Rats, Inbred SHR , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Ventricular Function, Left/drug effects
2.
Immunohorizons ; 4(6): 352-362, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32576593

ABSTRACT

Pulmonary fibrosis is a chronic and generally fatal disorder characterized by progressive formation of scar-like tissue in the lungs. Sialic acids are often found as the terminal sugar on extracellular glycoconjugates such as protein glycosylations. Sialidases, also known as neuraminidases, desialylate glycoconjugates. Serum amyloid P (SAP), a pentameric serum glycoprotein that has two sialic acids on each polypeptide, inhibits the differentiation of monocytes into fibrocytes and promotes human PBMCs to accumulate high extracellular levels of IL-10. When SAP is desialylated with sialidase, the effects of SAP on fibrocyte differentiation and IL-10 accumulation are strongly inhibited. Intriguingly, in patients with pulmonary fibrosis, there are increased levels of sialidase activity in the bronchoalveolar lavage fluid, increased levels of sialidases in the lungs, and decreased levels of SAP in the sera. To elucidate the role of SAP desialylation in idiopathic pulmonary fibrosis (IPF) pathogenesis, we purified SAP from the serum of IPF patients and healthy controls and measured the extent of sialylation and bioactivity of the purified SAP. We find that some IPF patients have abnormally high levels of the sialidase NEU3 in their sera and that the SAP in the sera of IPF patients has an abnormally high extent of desialylation and an abnormally low ability to inhibit fibrocyte differentiation and induce extracellular IL-10 accumulation by PBMC. These results suggest that SAP desialylation may play a role in IPF pathogenesis and that inhibiting NEU3 could be a potential therapeutic target for IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/prevention & control , Leukocytes, Mononuclear/metabolism , Neuraminidase/metabolism , Serum Amyloid P-Component/metabolism , Aged , Bronchoalveolar Lavage Fluid , Case-Control Studies , Cell Differentiation/drug effects , Cells, Cultured , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Lung/metabolism , Lung/physiopathology , Male , Mass Spectrometry , Middle Aged , Neuraminidase/antagonists & inhibitors , Serum Amyloid P-Component/pharmacology
3.
Front Immunol ; 9: 2328, 2018.
Article in English | MEDLINE | ID: mdl-30459752

ABSTRACT

Pentraxins such as serum amyloid P (SAP; also known as PTX2) regulate several aspects of the innate immune system. SAP inhibits the differentiation of monocyte-derived fibroblast-like cells called fibrocytes, promotes the formation of immuno-regulatory macrophages, and inhibits neutrophil adhesion to extracellular matrix proteins. In this minireview, we describe how these effects of SAP have led to its possible use as a therapeutic, and how modulating SAP effects might be used for other therapeutics. Fibrosing diseases such as pulmonary fibrosis, cardiac fibrosis, liver fibrosis, and renal fibrosis are associated with 30-45% of deaths in the US. Fibrosis involves both fibrocyte differentiation and profibrotic macrophage differentiation, and possibly because SAP inhibits both of these processes, in 9 different animal models, SAP inhibited fibrosis. In Phase 1B and Phase 2 clinical trials, SAP injections reduced the decline in lung function in pulmonary fibrosis patients, and in a small Phase 2 trial SAP injections reduced fibrosis in myelofibrosis patients. Acute respiratory distress syndrome/ acute lung injury (ARDS/ALI) involves the accumulation of neutrophils in the lungs, and possibly because SAP inhibits neutrophil adhesion, SAP injections reduced the severity of ARDS in an animal model. Conversely, depleting SAP is a potential therapeutic for amyloidosis, topically removing SAP from wound fluid speeds wound healing in animal models, and blocking SAP binding to one of its receptors makes cultured macrophages more aggressive toward tuberculosis bacteria. These results suggest that modulating pentraxin signaling might be useful for a variety of diseases.


Subject(s)
Serum Amyloid P-Component/pharmacology , Amyloidosis/drug therapy , Amyloidosis/etiology , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Clinical Trials as Topic , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/metabolism , Fibrosis/drug therapy , Fibrosis/etiology , Fibrosis/metabolism , Humans , Immunomodulation/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Multigene Family , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Serum Amyloid P-Component/genetics , Serum Amyloid P-Component/metabolism , Serum Amyloid P-Component/therapeutic use , Signal Transduction/drug effects , Tuberculosis/drug therapy , Tuberculosis/microbiology , Wound Healing/drug effects
4.
Cell Biol Int ; 42(11): 1556-1563, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30095216

ABSTRACT

Cardiovascular disease is a leading cause of death worldwide, requiring the development of new therapeutic strategies including stem cell therapy. Pentraxins (PTXs) are a superfamily of proteins highly involved in different myocardial disorders, and thus this study aimed to identify the modulation of long pentraxin 3 (PTX3) in the differentiation of mouse embryonic stem cells (mESCs) toward cardiomyocytes. Cell toxicity of PTX3 was detected by MTT and LDH assays in mESCs. Embryoid bodies (EBs) were differentiated using hanging drop method, and the beating was observed under microscope. Expressional levels of early cardiac progenitor marker genes were assessed by qRT-PCR. Expression of marker proteins in early myocardial development and the activation of JNK signaling pathway was evaluated by Western blot. PTX3 treatment at 50 ng/mL significantly promoted the expression of cardiac-specific marker genes including Nkx2.5, Mef2c, Tbx5, dHand, and αMHC, and increased the expression of cardiac maturity indicative markers including connexin 43 and troponin C1. PTX3 enhanced the phosphorylation of JNK across the incubation duration, whereas the activation of p38 remained the same as control group. Co-treatment of JNK signaling pathway inhibitor SP600125 impaired the PTX3-promoted transcription of Nkx2.5, Mef2c, Tbx5, dHand, and αMHC. This study revealed the promotion of PTX3 in the differentiation of mESCs into cardiomyocytes and the underlying mechanism.


Subject(s)
C-Reactive Protein/pharmacology , Cell Differentiation/drug effects , MAP Kinase Signaling System/drug effects , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Myocardium/cytology , Serum Amyloid P-Component/pharmacology , Animals , Biomarkers/metabolism , Cell Survival/drug effects , Connexin 43/metabolism , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Embryoid Bodies/metabolism , Gene Expression Regulation/drug effects , Mice , Mouse Embryonic Stem Cells/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Organ Specificity , Troponin C/metabolism
5.
JAMA ; 319(22): 2299-2307, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29800034

ABSTRACT

Importance: Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease with poor prognosis. Approved therapies do not halt disease progression. Objective: To determine the effect of recombinant human pentraxin 2 vs placebo on change from baseline to week 28 in mean forced vital capacity (FVC) percentage of predicted value. Design, Setting, and Participants: Phase 2, randomized, double-blind, placebo-controlled trial conducted at 18 sites in 7 countries of eligible patients with IPF (N = 117; aged 40-80 years; FVC ≥50% and ≤90% predicted; ratio of forced expiratory volume in the first second/FVC >0.70; diffusing capacity for carbon monoxide [Dlco] ≥25% and ≤90% predicted; and distance of ≥150 m on the 6-minute walk test). Study period was August 2015-May 2017. Interventions: Patients were randomized to receive either recombinant human pentraxin 2 (10 mg/kg intravenous every 4 weeks, n = 77) or placebo (n = 39) for 24 weeks, and stratified by concurrent IPF treatment status. Main Outcomes and Measures: The primary end point was the least-squares mean change in FVC percentage of predicted value from baseline to week 28 (minimal clinically important difference, decline of 2%-6%). Secondary end points included mean change in lung volumes (total, normal, and interstitial lung abnormalities) on high-resolution computed tomography (HRCT) and 6-minute walk distance (minimal clinically important difference, 24-45 m). Results: Of 117 randomized patients, 116 received at least 1 dose of study drug (mean age, 68.6 years; 81.0% men; mean time since IPF diagnosis, 3.8 years), and 111 (95.7%) completed the study. The least-squares mean change in FVC percentage of predicted value from baseline to week 28 in patients treated with recombinant human pentraxin 2 was -2.5 vs -4.8 for those in the placebo group (difference, +2.3 [90% CI, 1.1 to 3.5]; P = .001). No significant treatment differences were observed in total lung volume (difference, 93.5 mL [90% CI, -27.7 to 214.7]), quantitative parenchymal features on HRCT (normal lung volume difference, -1.2% [90% CI, -4.4 to 1.9]; interstitial lung abnormalities difference, 1.1% [90% CI, -2.2 to 4.3]), or measurement of Dlco (difference, -0.4 [90% CI, -2.6 to 1.7]). The change in 6-minute walk distance was -0.5 m for patients treated with recombinant human pentraxin 2 vs -31.8 m for those in the placebo group (difference, +31.3 m [90% CI, 17.4 to 45.1]; P < .001). The most common adverse events in the recombinant human pentraxin 2 vs placebo group were cough (18% vs 5%), fatigue (17% vs 10%), and nasopharyngitis (16% vs 23%). Conclusions and Relevance: In this preliminary study, recombinant human pentraxin 2 vs placebo resulted in a slower decline in lung function over 28 weeks for patients with idiopathic pulmonary fibrosis. Further research should more fully assess efficacy and safety. Trial Registration: clinicaltrials.gov Identifier: NCT02550873.


Subject(s)
Homeodomain Proteins/therapeutic use , Idiopathic Pulmonary Fibrosis/drug therapy , Serum Amyloid P-Component/therapeutic use , Vital Capacity/drug effects , Aged , Double-Blind Method , Female , Homeodomain Proteins/adverse effects , Homeodomain Proteins/pharmacology , Humans , Idiopathic Pulmonary Fibrosis/physiopathology , Least-Squares Analysis , Male , Middle Aged , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Serum Amyloid P-Component/adverse effects , Serum Amyloid P-Component/pharmacology , Walk Test
6.
PLoS One ; 13(4): e0195758, 2018.
Article in English | MEDLINE | ID: mdl-29672566

ABSTRACT

Pentraxin-3 (PTX3) is a long-form member of the pentraxin family of proteins that has been studied in inflammatory diseases and in various organs. We found that PTX3 protects kidney cells during ischemia and proinflammatory acute kidney injury. The aim of this study was to develop an in vitro experimental model of acute kidney injury and to analyze the protective mechanism of exogenous recombinant PTX3. In this study, cells of the HK-2 renal tubular cell line were treated with a calcium ionophore (A23187), which induced injury by increasing intracellular calcium concentrations and inducing calpain activity and the generation of reactive oxygen species. Exposure of cells to PTX3 significantly attenuated these effects. In addition, the activity of caspase-3 and PARP-1 were decreased in ischemic cells exposed to exogenous recombinant PTX3. PTX3 stabilized the mitochondrial membrane potential and suppressed apoptosis, resulting in the protection of renal tubular cells from ischemic injury.


Subject(s)
Acute Kidney Injury/metabolism , C-Reactive Protein/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Serum Amyloid P-Component/pharmacology , Acute Kidney Injury/pathology , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Humans , Hypoxia/drug therapy , Hypoxia/metabolism , Ischemia/drug therapy , Ischemia/metabolism , Matrix Metalloproteinases , Protective Agents/pharmacology
7.
Pharmacol Rep ; 70(1): 22-28, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29306759

ABSTRACT

BACKGROUND: Circulating fibrocytes (CFs) have been shown to participate in subepithelial fibrosis of asthma with chronic airflow limitation by acting as an important source of fibroblasts deposited beneath airway epithelia. Serum amyloid P (SAP) is an innate inhibitor of fibrocytes differentiation. Store-operated Ca2+ entry (SOCE) is the major Ca2+ influx of non-excitable cells. In this study, the role of SOCE in the regulation of fibrocytes differentiation and the effects of Th2 cytokine IL-4 and SAP on SOCE of fibrocytes were investigated. METHODS: Peripheral blood mononuclear cells or monocytes were cultured in serum-free medium for 7days to differentiate into fibrocytes; the expression of SOC channels was determined with PCR, SOCE was measured with Ca2+ fluorescence imaging. RESULTS: IL-4 significantly promoted monocyte derived fibrocytes differentiation in vitro. It also increased both SOCE which was induced by thapsigargin or UTP and molecules STIM1 and Orai1 which were related to expression of SOC channels in fibrocytes. Fibrocytes differentiation induced by IL-4 and SOC channels activity could be inhibited by SOC channel blocker SKF-96365. As expected, SAP significantly inhibited IL-4-induced differentiation of fibrocytes, the activity of SOCE and the expression of STIM1 and Orai1 in IL-4-treated fibrocytes. CONCLUSION: IL-4 and SAP reversely regulates cultured fibrocytes differentiation in vitro by respectively promoting or inhibiting the expression and activity of SOC channels in fibrocytes.


Subject(s)
Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Calcium/metabolism , Cell Differentiation/drug effects , Interleukin-4/pharmacology , Mesenchymal Stem Cells/drug effects , Neoplasm Proteins/drug effects , ORAI1 Protein/drug effects , Serum Amyloid P-Component/pharmacology , Stromal Interaction Molecule 1/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Imidazoles/pharmacology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Neoplasm Proteins/metabolism , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism , Time Factors
8.
BMC Immunol ; 18(1): 47, 2017 Dec 04.
Article in English | MEDLINE | ID: mdl-29202702

ABSTRACT

BACKGROUND: Pentraxins are a family of highly conserved secreted proteins that regulate the innate immune system, including monocytes and macrophages. C-reactive protein (CRP) is a plasma protein whose levels can rise to 1000 µg/ml from the normal <3 µg/ ml during inflammation. RESULTS: We find that CRP inhibits proliferation of the human myeloid leukemia cell line Mono Mac 6 with an IC50 of 75 µg/ ml by inducing apoptosis of these cells. The related proteins serum amyloid P (SAP) and pentraxin 3 (PTX3) do not inhibit Mono Mac 6 proliferation. CRP has no significant effect on the proliferation of other leukemia cell lines such as HL-60, Mono Mac 1, K562, U937, or THP-1, or the survival of normal peripheral blood cells. The effect of CRP appears to be dependent on the CRP receptor FcγRI, and is negatively regulated by a phosphatidylinositol -3-kinase pathway. CONCLUSION: These data reveal differential signaling by pentraxins on immune cells, and suggest that CRP can regulate the proliferation of some myeloid leukemia cells.


Subject(s)
Apoptosis/drug effects , C-Reactive Protein/pharmacology , Leukemia, Myeloid, Acute/physiopathology , Serum Amyloid P-Component/pharmacology , Antigens, CD/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Inhibitory Concentration 50 , Leukocytes, Mononuclear/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Receptors, Fc/genetics , Receptors, Fc/metabolism , Receptors, IgG/metabolism , Receptors, Immunologic/genetics , Signal Transduction/drug effects
9.
Hepatology ; 66(3): 953-968, 2017 09.
Article in English | MEDLINE | ID: mdl-28422322

ABSTRACT

Acute-on-chronic liver injury is characterized by an important inflammatory response frequently associated with endotoxemia. In this context, acute-phase proteins such as Pentraxin-3 (PTX3) are released; however, little is known about their role in chronic liver disease. The aim of this study was to elucidate the role of PTX3 in liver injury. The role of PTX3 was evaluated in cultured human cells, liver tissue slices, and mice with acute-on-chronic liver injury. PTX3 expression was assessed in tissue and serum samples from 54 patients with alcoholic hepatitis. PTX3 expression was up-regulated in animal models of liver injury and strongly induced by lipopolysaccharide (LPS). Liver cell fractionation showed that macrophages and activated hepatic stellate cells were the main cell types expressing PTX3 in liver injury. Ex vivo and in vivo studies showed that PTX3 treatment attenuated LPS-induced liver injury, inflammation, and cell recruitment. Mechanistically, PTX3 mediated the hepatic stellate cell wound-healing response. Moreover, PTX3 modulated LPS-induced inflammation in human primary liver macrophages and peripheral monocytes by enhancing a TIR domain-containing adapter-inducing interferon-dependent response and favoring a macrophage interleukin-10-like phenotype. Additionally, hepatic and plasma PTX3 levels were increased in patients with alcoholic hepatitis, a prototypic acute-on-chronic condition; and its expression correlated with disease severity scores, endotoxemia, infections, and short-term mortality, thus suggesting that expression of PTX3 found in patients could be a counterregulatory response to injury. CONCLUSION: Experimental and human evidence suggests that, in addition to being a potential biomarker for alcoholic hepatitis, PTX3 participates in the wound-healing response and attenuates LPS-induced liver injury and inflammation; therefore, administration of PTX3 could be a promising therapeutic strategy in acute-on-chronic conditions, particularly those associated with endotoxemia. (Hepatology 2017;66:953-968).


Subject(s)
Acute-On-Chronic Liver Failure/pathology , C-Reactive Protein/genetics , Cytokines/metabolism , Gene Expression Regulation , Serum Amyloid P-Component/genetics , Acute-On-Chronic Liver Failure/genetics , Animals , Biopsy, Needle , C-Reactive Protein/pharmacology , Disease Models, Animal , Disease Progression , Female , Hepatic Stellate Cells/metabolism , Humans , Immunohistochemistry , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Random Allocation , Retrospective Studies , Serum Amyloid P-Component/pharmacology , Up-Regulation
10.
Cereb Cortex ; 27(6): 3437-3448, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28334103

ABSTRACT

Neuronal pentraxin 1 (NPTX1) has been implicated in Alzheimer's disease, being present in and around dystrophic neurons in plaques, affecting glutamatergic transmission postsynaptically and mediating effects of amyloidß. Here, we confirm the presence of NPTX1 around plaques in postmortem Alzheimer's disease brain and report that acutely applied human NPTX1 increases paired-pulse ratio at mouse CA3-CA1 hippocampal synapses, indicating a decrease in glutamate release. In contrast, chronic exposure to NPTX1, NPTX2, or NPTX receptor decreases paired-pulse ratio, mimicking some of the earliest changes in mice expressing familial Alzheimer's disease genes. The peripheral pentraxin, serum amyloid P component (SAP), causes similar synaptic effects to NPTX1. The presence of SAP on amyloid plaques in Alzheimer's disease confirms that it can enter the brain. We show that SAP and neuronal pentraxins can interact and that SAP can enter the brain if the blood-brain barrier is compromised, suggesting that peripheral pentraxins could affect central synaptic transmission via this interaction, especially in the event of blood-brain barrier breakdown.


Subject(s)
Blood-Brain Barrier/physiopathology , C-Reactive Protein/metabolism , Glutamic Acid/metabolism , Hippocampus/physiology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Animals, Newborn , Blood-Brain Barrier/pathology , C-Reactive Protein/genetics , C-Reactive Protein/pharmacology , Evoked Potentials/drug effects , Evoked Potentials/physiology , Female , GABA Antagonists/pharmacology , HEK293 Cells , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Neurons/drug effects , Pyridazines/pharmacology , Serum Amyloid P-Component/pharmacology , Synapses/drug effects , Synapses/genetics , Synapses/metabolism
11.
Sci Rep ; 7: 43736, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28252047

ABSTRACT

Proliferative vitreoretinopathy (PVR) is mediated by proliferation and epithelial mesenchymal transition (EMT) of retinal pigment epithelium (RPE). Because heavy chain-hyaluronic acid/pentraxin 3 (HC-HA/PTX3) purified from human amniotic membrane exerts anti-inflammatory and anti-scarring actions, we hypothesized that HC-HA/PTX3 could inhibit these PVR-related processes in vitro. In this study, we first optimized an ARPE-19 cell culture model to mimic PVR by defining cell density, growth factors, and cultivation time. Using this low cell density culture model and HA as a control, we tested effects of HC-HA/PTX3 on the cell viability (cytotoxicity), proliferation (EGF + FGF-2) and EMT (TGF-ß1). Furthermore, we determined effects of HC-HA/PTX3 on cell migration (EGF + FGF-2 + TGF-ß1) and collagen gel contraction (TGF-ß1). We found both HA and HC-HA/PTX3 were not toxic to unstimulated RPE cells. Only HC-HA/PTX3 dose-dependently inhibited proliferation and EMT of stimulated RPE cells by down-regulating Wnt (ß-catenin, LEF1) and TGF-ß (Smad2/3, collagen type I, α-SMA) signaling, respectively. Additionally, HA and HC-HA/PTX3 inhibited migration but only HC-HA/PTX3 inhibited collagen gel contraction. These results suggest HC-HA/PTX3 is a non-toxic, potent inhibitor of proliferation and EMT of RPE in vitro, and HC-HA/PTX3's ability to inhibit PVR formation warrants evaluation in an animal model.


Subject(s)
C-Reactive Protein/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , Hyaluronic Acid/pharmacology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Serum Amyloid P-Component/pharmacology , Biomarkers , Cell Movement , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , Epithelial Cells/pathology , Humans , Protein Transport , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Vitreoretinopathy, Proliferative/etiology , Vitreoretinopathy, Proliferative/metabolism , Vitreoretinopathy, Proliferative/pathology
12.
Sci Rep ; 7: 42195, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28165063

ABSTRACT

Chronic graft-versus-host disease (cGVHD) is a major complication of hematopoietic stem cell transplantation. Dry eye disease is the prominent ocular sequel of cGVHD and is caused by excessive inflammation and fibrosis in the lacrimal glands. Heavy chain-Hyaluronan/Pentraxin 3 (HC-HA/PTX3) is a complex purified from human amniotic membrane (AM) and known to exert anti-inflammatory and anti-scarring actions. In this study, we utilized a mouse model of cGVHD to examine whether HC-HA/PTX3 could attenuate dry eye disease elicited by cGVHD. Our results indicated that subconjunctival and subcutaneous injection of HC-HA/PTX3 preserved tear secretion and conjunctival goblet cell density and mitigated inflammation and scarring of the conjunctiva. Such therapeutic benefits were associated with suppression of scarring and infiltration of inflammatory/immune cells in the lacrimal glands. Furthermore, HC-HA/PTX3 significantly reduced the extent of infiltration of CD45+ CD4+ IL-17+ cells, CD45+ CD34+ collagen I+ CXCR4+ fibrocytes, and HSP47+ activated fibroblasts that were accompanied by upregulation of collagen type Iα1, collagen type IIIα1 and NF-kB in lacrimal glands. Collectively, these pre-clinical data help prove the concept that subcutaneous and subconjunctival injection of HC-HA/PTX3 is a novel approach to prevent dry eye disease caused by cGVHD and allow us to test its safety and efficacy in future human clinical trials.


Subject(s)
C-Reactive Protein/pharmacology , Cicatrix/prevention & control , Conjunctiva/drug effects , Dry Eye Syndromes/drug therapy , Graft vs Host Disease/pathology , Hyaluronic Acid/pharmacology , Lacrimal Apparatus/drug effects , Serum Amyloid P-Component/pharmacology , Amnion/chemistry , Animals , Bone Marrow Transplantation/adverse effects , C-Reactive Protein/isolation & purification , Chronic Disease , Cicatrix/etiology , Cicatrix/immunology , Cicatrix/pathology , Conjunctiva/immunology , Conjunctiva/pathology , Disease Models, Animal , Dry Eye Syndromes/etiology , Dry Eye Syndromes/immunology , Dry Eye Syndromes/pathology , Female , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/pathology , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Humans , Hyaluronic Acid/isolation & purification , Injections, Intraocular , Injections, Subcutaneous , Lacrimal Apparatus/immunology , Lacrimal Apparatus/pathology , Male , Mice , Serum Amyloid P-Component/isolation & purification , Transplantation, Homologous , Whole-Body Irradiation
13.
Cell Physiol Biochem ; 40(5): 1029-1038, 2016.
Article in English | MEDLINE | ID: mdl-27941350

ABSTRACT

BACKGROUND/AIMS: Tubulointerstitial fibrosis can lead to end-stage renal disease. Pentraxin 3 (PTX3) is an acute phase protein produced by resident and innate immunity cells. We investigated the effect of PTX3 on cultured human proximal tubular epithelial (HK-2) cells and a rat unilateral ureteral obstruction (UUO) model of renal fibrosis. METHODS: Gain-of-function experiments were used to examine the effect of recombinant human PTX3 (Rh-PTX3) on HK-2 cells. Cell proliferation (MTT assay) and in vitro cell migration were measured. The levels of PTX3, p-JNK, and EMT markers were measured using immunohistochemistry, RT-PCR, and western blotting in UUO rats and HK-2 cells. RESULTS: HK-2 cells treated with Rh PTX3 did not affect cell viability, but significantly increased cell migration. Moreover, Rh-PTX3 increased the expression of snail, slug, N-cadherin, and vimentin, decreased the expression of E-cadherin, and increased the phosphorylation of JNK. SP600126 (a specific JNK inhibitor) enhanced the effects of Rh-PTX3. Rats with UUO exhibited time-dependent increased levels of PTX3, p-JNK, and vimentin, and decreased expression of E-cadherin. CONCLUSIONS: Our results suggest that PTX3 induces cell migration via upregulation of EMT in a JNK-dependent mechanism, and highlight the role of PTX3 in the pathogenesis renal fibrosis.


Subject(s)
C-Reactive Protein/pharmacology , Epithelial-Mesenchymal Transition/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney/enzymology , Kidney/pathology , MAP Kinase Signaling System/drug effects , Serum Amyloid P-Component/pharmacology , Animals , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Disease Models, Animal , Disease Progression , Enzyme Activation/drug effects , Fibrosis , Humans , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/pathology , Male , Phosphorylation/drug effects , Rats, Sprague-Dawley , Ureteral Obstruction/pathology
14.
J Exp Med ; 213(9): 1723-40, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27481130

ABSTRACT

Primary myelofibrosis (PMF) is a fatal neoplastic disease characterized by clonal myeloproliferation and progressive bone marrow (BM) fibrosis thought to be induced by mesenchymal stromal cells stimulated by overproduced growth factors. However, tissue fibrosis in other diseases is associated with monocyte-derived fibrocytes. Therefore, we sought to determine whether fibrocytes play a role in the induction of BM fibrosis in PMF. In this study, we show that BM from patients with PMF harbors an abundance of clonal, neoplastic collagen- and fibronectin-producing fibrocytes. Immunodeficient mice transplanted with myelofibrosis patients' BM cells developed a lethal myelofibrosis-like phenotype. Treatment of the xenograft mice with the fibrocyte inhibitor serum amyloid P (SAP; pentraxin-2) significantly prolonged survival and slowed the development of BM fibrosis. Collectively, our data suggest that neoplastic fibrocytes contribute to the induction of BM fibrosis in PMF, and inhibiting fibrocyte differentiation with SAP may interfere with this process.


Subject(s)
Fibroblasts/physiology , Monocytes/cytology , Primary Myelofibrosis/etiology , Animals , Bone Marrow/pathology , Bone Marrow Transplantation , Cells, Cultured , Fibroblasts/drug effects , Fibrosis , Homeodomain Proteins/pharmacology , Humans , Mice , Mice, SCID , Nitriles , Primary Myelofibrosis/pathology , Pyrazoles/pharmacology , Pyrimidines , Recombinant Proteins/pharmacology , Serum Amyloid P-Component/pharmacology
15.
Sci Rep ; 6: 22695, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26936002

ABSTRACT

Amyloidosis is a protein-misfolding disorder characterized by the extracellular deposition of amyloid, a complex matrix composed of protein fibrils, hyper-sulphated glycosaminoglycans and serum amyloid P component (SAP). Accumulation of amyloid in visceral organs results in the destruction of tissue architecture leading to organ dysfunction and failure. Early differential diagnosis and disease monitoring are critical for improving patient outcomes; thus, whole body amyloid imaging would be beneficial in this regard. Non-invasive molecular imaging of systemic amyloid is performed in Europe by using iodine-123-labelled SAP; however, this tracer is not available in the US. Therefore, we evaluated synthetic, poly-basic peptides, designated p5 and p5+14, as alternative radiotracers for detecting systemic amyloidosis. Herein, we perform a comparative effectiveness evaluation of radiolabelled peptide p5+14 with p5 and SAP, in amyloid-laden mice, using dual-energy SPECT imaging and tissue biodistribution measurements. All three radiotracers selectively bound amyloid in vivo; however, p5+14 was significantly more effective as compared to p5 in certain organs. Moreover, SAP bound principally to hepatosplenic amyloid, whereas p5+14 was broadly distributed in numerous amyloid-laden anatomic sites, including the spleen, liver, pancreas, intestines and heart. These data support clinical validation of p5+14 as an amyloid radiotracer for patients in the US.


Subject(s)
Amyloidosis/diagnostic imaging , Amyloidosis/metabolism , Peptides , Serum Amyloid P-Component/metabolism , Single Photon Emission Computed Tomography Computed Tomography/methods , Animals , Disease Models, Animal , Iodine Radioisotopes/pharmacology , Isotope Labeling/methods , Mice , Peptides/pharmacokinetics , Peptides/pharmacology , Radioactive Tracers , Serum Amyloid P-Component/pharmacology
16.
Microb Pathog ; 89: 128-39, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26427882

ABSTRACT

Pentraxin 3 (PTX3), a soluble pattern recognition receptor, plays an important role in innate immunity and has been implicated to be a candidate resistance gene against Streptococcus suis 2 infection. To discover the antibacterial effect of porcine PTX3 against S. suis 2, the 42-kDa PTX3 protein was expressed by Chinese hamster ovary cells (CHO), and an additional eukaryotic expression vector pVAX-ptx3 was constructed. The expressed porcine PTX3 mediated a range of antibacterial activities including increasing phagocytic capacity of primary porcine alveolar macrophages (PAM) against S. suis 2 and inhibiting adhesion of S. suis 2 to human epidermoid cancer cells (Hep-2). In mouse model, pre-intramuscular injecting with pVAX-ptx3 reduced mortality and reduced bacteria loads in blood, spleen, lung and brain compared with that of control group during 2-12 h following intraperitoneal injection (i.p.) with S. suis 2. Meanwhile, the expressions of IL-6 and IL-8 in blood were increased in pVAX-ptx3 group, whereas no obvious changes about IL-10. In piglet model, bacteria load in blood of pVAX-ptx3 group was significantly lower than that of control group after i.p. with S. suis 2, correspondingly, expression of IL-6 and IL-8 were significantly increased in pVAX-ptx3 group. In contrast, white blood cell (WBC) and neutrophil cell (NEU) count of peripheral blood in pVAX-ptx3 group were lower than that of control group. These studies described a novel antibacterial role for porcine PTX3 against S. suis 2 both in vitro and in vivo and suggested that porcine PTX3 may be a potential biological agent against S. suis 2 in pig and be used for the clinical prevention and treatment of streptococcosis caused by S. suis 2.


Subject(s)
C-Reactive Protein/pharmacology , Immunologic Factors/pharmacology , Serum Amyloid P-Component/pharmacology , Streptococcus suis/drug effects , Animal Structures/microbiology , Animals , Bacterial Adhesion/drug effects , Bacterial Load , C-Reactive Protein/administration & dosage , C-Reactive Protein/genetics , C-Reactive Protein/isolation & purification , Epithelial Cells/microbiology , Immunologic Factors/administration & dosage , Injections, Intramuscular , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Mice , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Serum Amyloid P-Component/administration & dosage , Serum Amyloid P-Component/genetics , Serum Amyloid P-Component/isolation & purification , Streptococcal Infections/microbiology , Streptococcal Infections/prevention & control , Streptococcus suis/immunology , Streptococcus suis/physiology , Survival Analysis , Swine
17.
Oncotarget ; 6(15): 13790-802, 2015 May 30.
Article in English | MEDLINE | ID: mdl-25912421

ABSTRACT

Fibroblast growth factor-8b (FGF8b) affects the epithelial/stromal compartments of steroid hormone-regulated tumors by exerting an autocrine activity on cancer cells and a paracrine pro-angiogenic function, thus contributing to tumor progression. The FGF8b/FGF receptor (FGFR) system may therefore represent a target for the treatment of steroid hormone-regulated tumors. The soluble pattern recognition receptor long pentraxin-3 (PTX3) binds various FGFs, including FGF2 and FGF8b, thus inhibiting the angiogenic and tumorigenic activity of androgen-regulated tumor cells. Nevertheless, the complex/proteinaceous structure of PTX3 hampers its pharmacological exploitation. In this context, the acetylated pentapeptide Ac-ARPCA-NH2 (ARPCA), corresponding to the N-terminal amino acid sequence PTX3(100-104), was identified as a minimal FGF2-binding peptide able to antagonize the biological activity of FGF2. Here, we demonstrate that ARPCA binds FGF8b and inhibits its capacity to form FGFR1-mediated ternary complexes with heparan sulphate proteoglycans. As a FGF8b antagonist, ARPCA inhibits FGFR1 activation and signalling in endothelial cells, hampering the angiogenic activity exerted in vitro and in vivo by FGF8b. Also, ARPCA suppresses the angiogenic and tumorigenic potential of prototypic androgen/FGF8b-dependent Shionogi 115 mammary carcinoma cells and of androgen/FGF8b/FGF2-dependent TRAMP-C2 prostate cancer cells. In conclusion, ARPCA represents a novel FGF8b antagonist with translational implications for the therapy of steroid hormone-regulated tumors.


Subject(s)
C-Reactive Protein/pharmacology , Fibroblast Growth Factor 8/metabolism , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Peptide Fragments/pharmacology , Serum Amyloid P-Component/pharmacology , Animals , Cell Proliferation/drug effects , Chick Embryo , Fibroblast Growth Factor 8/genetics , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Models, Molecular , Neoplasms, Hormone-Dependent/blood supply , Neovascularization, Physiologic/drug effects
18.
Antimicrob Agents Chemother ; 58(10): 6284-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25070103

ABSTRACT

Posaconazole is currently used for the prophylaxis of invasive pulmonary aspergillosis (IPA). Limitations to posaconazole usage are drug-drug interactions and side effects. PTX3 is an innate immunity glycoprotein with opsonic activity, proven to be protective in IPA animal models. This study investigated the combination of posaconazole with PTX3. The results indicate synergy between PTX3 and posaconazole against aspergillosis, suggesting that a combination of reduced doses of posaconazole with the immune response enhancer PTX3 might represent a treatment option with a higher therapeutic index than posaconazole.


Subject(s)
Antifungal Agents/pharmacology , C-Reactive Protein/pharmacology , Invasive Pulmonary Aspergillosis/drug therapy , Serum Amyloid P-Component/pharmacology , Triazoles/pharmacology , Animals , Drug Synergism , Male , Rats
19.
Inflammation ; 37(5): 1895-901, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24854162

ABSTRACT

Acute kidney injury-induced organ fibrosis is recognized as a major risk factor for the development of chronic kidney disease, which remains one of the leading causes of death in the developed world. However, knowledge on molecules that may suppress the fibrogenic response after injury is lacking. The long pentraxin 3 (PTX3), a novel acute renal injury marker, has been reported to be involved in chronic renal injury, but the mechanism is still unknown. In this experiment, the mice subjected to acute kidney injury showed a slow recovery of kidney function compared with PTX3-treated animals. Collagen expression was absent in sham-operated kidneys; however, their expression was significantly increased after reperfusion. And, these changes were reduced in PTX3-treated mouse kidney. Fibrosis was associated with increased expression of IL-6 and extensive activation of Stat3. Administration of IL-6 increased collagen I expression and Stat3 activation in vitro in renal epithelial cells subjected to hypoxia-reoxygenation, which was suppressed by PTX3. Furthermore, we found that the decreased serum creatinine level and the reduced expression of collagen and smooth muscle actin induced by PTX3 were abolished by additional administration of IL-6. The associated p-Stat3 expression which was reduced by PTX3 administration was also inverted by additional IL-6 treatment. Our data suggest that PTX3 inhibits acute renal injury-induced interstitial fibrosis through suppression of IL-6/Stat3 pathway.


Subject(s)
Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , C-Reactive Protein/therapeutic use , Interleukin-6/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Serum Amyloid P-Component/therapeutic use , Signal Transduction/drug effects , Animals , C-Reactive Protein/pharmacology , Cells, Cultured , Fibrosis/drug therapy , Fibrosis/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/metabolism , Serum Amyloid P-Component/pharmacology , Signal Transduction/physiology
20.
Invest Ophthalmol Vis Sci ; 55(3): 1647-56, 2014 Mar 19.
Article in English | MEDLINE | ID: mdl-24519420

ABSTRACT

PURPOSE: Heavy chain-hyaluronic acid (HC-HA)/PTX3 purified from human amniotic membrane (AM) was previously observed to suppress inflammatory responses in vitro. We now examine whether HC-HA/PTX3 is able to exert a similar effect in vivo, using murine models for keratitis and corneal allograft rejection. METHODS: The in vitro effect of HC-HA/PTX3 was tested using OTII ovalbumin (OVA) transgenic, purified CD4(+) T cells, or IFN-γ/lipopolysaccharide (LPS)-stimulated RAW264.7 cells. Cytokine production was measured by ELISA, while cell surface markers and cell proliferation were determined by flow cytometry. In vivo effects of HC-HA/PTX3 were analyzed by quantifying the recruitment of enhanced green fluorescence-labeled macrophages and by measuring the expression of arginase 1 (Arg-1), IL-10, and IL-12 in LPS-induced keratitis in the macrophage Fas-induced apoptosis (Mafia) mouse. The effect of corneal allograft survival in a complete major histocompatibility complex (MHC) mismatched mouse model was assessed by grading corneal opacification. RESULTS: In vitro studies demonstrated that HC-HA/PTX3 significantly enhanced the expansion of FOXP3 T cells and suppressed cell proliferation and protein expression of IFN-γ, IL-2, CD25, and CD69 in activated CD4(+) T cells. Furthermore, immobilized HC-HA/PTX3 significantly upregulated IL-10 gene expression but downregulated that of IL-12 and IL-23 in activated RAW264.7 cells. Finally, in vivo subconjunctival injection of HC-HA/PTX3 significantly prolonged corneal allograft survival, suppressed macrophage infiltration, and promoted M2 polarization by upregulating Arg-1 and IL-10 but downregulating IL-12. CONCLUSIONS: HC-HA/PTX3 can suppress inflammatory responses in vivo by modulating both innate and adaptive immunity of macrophages and CD4(+) T cells.


Subject(s)
Adaptive Immunity/physiology , Amnion/immunology , C-Reactive Protein/pharmacology , Corneal Transplantation , Graft Rejection/immunology , Hyaluronic Acid/pharmacology , Immunity, Innate/physiology , Serum Amyloid P-Component/pharmacology , Acute-Phase Proteins , Allografts , Amnion/cytology , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Cytokines/biosynthesis , Cytokines/drug effects , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Graft Rejection/metabolism , Graft Rejection/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL