Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
2.
J Leukoc Biol ; 108(6): 1727-1733, 2020 12.
Article in English | MEDLINE | ID: mdl-32640496

ABSTRACT

This study tested the hypothesis that heparanase (HPSE) is related to tumor metastasis and curcumin (CCM) inhibits tumor metastasis by down-regulating HPSE expression. MTT, Transwell assays, and RT-PCR were used to study the effects of CCM on the migration and invasion of CT26 cells and the expression of HPSE. CT26 cells were transfected with lentivirus to establish HPSE-overexpressing cells (OE) and corresponding negative control cells (NC). Signal pathways involved in down-regulating the expression of HPSE and inhibiting the migration and invasion of CT26 cells by CCM were screened by the liquid crystal chip. HPSE promoted CT26 cells migration and invasion, and CCM inhibited the proliferation and metastasis of CT26 cells. The results of RT-PCR indicated that CCM down-regulated HPSE expression. Liquid phase microarray showed that CCM inhibited the phosphorylation of P38 and STAT5 in CT26 cells and NC cells. In contrast, the inhibitory function of CCM was markedly enhanced when HPSE was overexpressed (P < 0.05). In short, HPSE is closely related to metastasis of colon cancer cells. CCM inhibits colon cancer cell migration and invasion by inhibiting HPSE expression, which may be related to P38 MAPK and JAK/STAT5 signal pathways.


Subject(s)
Colonic Neoplasms/immunology , Curcumin/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glucuronidase/immunology , Neoplasm Proteins/immunology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Gene Expression Regulation, Enzymologic/immunology , Gene Expression Regulation, Neoplastic/immunology , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Neoplasm Metastasis
3.
Int Immunopharmacol ; 85: 106582, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32438076

ABSTRACT

Increased oxidative stress and inflammation play an important role in the pathogenesis of diabetic cataract. Klotho, known as an anti-ageing protein, has antioxidative and anti-inflammatory properties. Klotho is expressed in limited tissues including the lens. Here we examined whether klotho expression is decreased in diabetic lens and, if so, whether klotho treatment can prevent diabetic cataract formation. Streptozotocin (STZ)-induced diabetic rats and age-matched control rats were treated with vehicle or klotho protein, starting at 1 week after STZ injection. Twelve weeks after treatment, cataract formation was observed in diabetic rats but not control rats. Cataract formation and scores were significantly less in klotho-treated diabetic rats than vehicle-treated diabetic rats. Levels of klotho in plasma, aqueous humor and lens were significantly decreased in vehicle-treated diabetic rats, compared with control rats, but were restored in klotho-treated diabetic rats. Additionally, vehicle-treated diabetic rats had increased oxidative stress and inflammation in the lens, which were associated with decreased antioxidant transcriptional master regulator Nrf2 activity and increased transcription factor NF-κB activity. All of these findings were ameliorated in klotho-treated diabetic rats. Notably, klotho treatment did not alter blood glucose in diabetic rats. These results indicate that klotho reduction may increase susceptibility of the lens to oxidative and inflammatory insults, promoting cataract formation under diabetic conditions. Klotho treatment can ameliorate the onset and progression of diabetic cataract via enhancing Nrf2-mediated antioxidant defense and suppressing NF-κB-mediated inflammatory responses. Klotho in the lens may be a novel therapeutic target for prevention of cataract formation in diabetes.


Subject(s)
Cataract/immunology , Diabetes Mellitus, Experimental/immunology , Glucuronidase/immunology , Lens, Crystalline/immunology , Animals , Blood Glucose , Cataract/blood , Diabetes Mellitus, Experimental/blood , Disease Progression , Glucuronidase/blood , Inflammation/immunology , Klotho Proteins , Male , NF-E2-Related Factor 2/immunology , NF-kappa B/immunology , Oxidative Stress , Rats, Wistar
4.
Mol Genet Metab ; 129(3): 219-227, 2020 03.
Article in English | MEDLINE | ID: mdl-32063397

ABSTRACT

Vestronidase alfa (recombinant human beta-glucuronidase) is an enzyme replacement therapy (ERT) for Mucopolysaccharidosis (MPS) VII, a highly heterogeneous, ultra-rare disease. Twelve subjects, ages 8-25 years, completed a Phase 3, randomized, placebo-controlled, blind-start, single crossover study (UX003-CL301; NCT02377921), receiving 24-48 weeks of vestronidase alfa 4 mg/kg IV. All 12 subjects completed the blind-start study, which showed significantly reduced urinary glycosaminoglycans (GAG) and clinical improvement in a multi-domain responder index, and enrolled in a long-term, open-label, extension study (UX003-CL202; NCT02432144). Here, we report the final results of the extension study, up to an additional 144 weeks after completion of the blind-start study. Three subjects (25%) completed all 144 weeks of study, eight subjects (67%) ended study participation before Week 144 to switch to commercially available vestronidase alfa, and one subject discontinued due to non-compliance after receiving one infusion of vestronidase alfa in the extension study. The safety profile of vestronidase alfa in the extension study was consistent with observations in the preceding blind-start study, with most adverse events mild to moderate in severity. There were no treatment or study discontinuations due to AEs and no noteworthy changes in a standard safety chemistry panel. Out of the eleven subjects who tested positive for anti-drug antibodies at any time during the blind-start or extension study, including the baseline assessment in the blind-start study, seven subjects tested positive for neutralizing antibodies and all seven continued to demonstrate a reduction in urinary GAG levels. There was no association between antibody formation and infusion associated reactions. Subjects receiving continuous vestronidase alfa treatment showed a sustained urinary GAG reduction and clinical response evaluated using a multi-domain responder index that includes assessments in pulmonary function, motor function, range of motion, mobility, and visual acuity. Reduction in fatigue was also maintained in the overall population. As ERT is not expected to cross the blood brain barrier, limiting the impact on neurological signs of disease, and not all subjects presented with neurological symptoms, outcomes related to central nervous system pathology are not focused on in this report. Results from this study show the long-term safety and durability of clinical efficacy in subjects with MPS VII with long-term vestronidase alfa treatment.


Subject(s)
Enzyme Replacement Therapy , Glucuronidase/therapeutic use , Glycosaminoglycans/urine , Mucopolysaccharidosis VII/drug therapy , Adolescent , Adult , Antibodies, Neutralizing , Blood-Brain Barrier/drug effects , Child , Cross-Over Studies , Female , Glucuronidase/administration & dosage , Glucuronidase/adverse effects , Glucuronidase/immunology , Humans , Male , Mucopolysaccharidosis VII/immunology , Mucopolysaccharidosis VII/physiopathology , Rare Diseases/therapy , Treatment Outcome
5.
Intern Emerg Med ; 15(3): 381-393, 2020 04.
Article in English | MEDLINE | ID: mdl-31919781

ABSTRACT

Experimental and clinical studies aimed at investigating the mechanism(s) underlying vascular complications of diabetes indicate that a great number of molecules are involved in the pathogenesis of these complications. Most of these molecules are inflammatory mediators or markers generated by immune or adipose tissue. Some of them, i.e. resistin and sortilin, have been shown to be involved in the cross talk between adipocytes and inflammatory cells. This interaction is an attractive area of research, particularly in type 2 diabetes and obesity. Other proteins, such as adiponectin and visfatin, appear to be more promising as possible vascular markers. In addition, some molecules involved in calcium/phosphorus metabolism, such as klotho and FGF23, have an involvement in the pathogenesis of diabetic vasculopathy, which appears to be dependent on the degree of vascular impairment. Inflammatory markers are a promising tool for treatment decisions while measuring plasma levels of adipokines, sortilin, Klotho and FGF23 in adequately sized longitudinal studies is expected to allow a more precise characterization of diabetic vascular disease and the optimal use of personalized treatment strategies.


Subject(s)
Adipose Tissue/immunology , Biomarkers/analysis , Cardiovascular Diseases/diagnosis , Immune System/immunology , Signal Transduction/immunology , Adaptor Proteins, Vesicular Transport/analysis , Adaptor Proteins, Vesicular Transport/blood , Adaptor Proteins, Vesicular Transport/immunology , Adipokines/analysis , Adipokines/blood , Adipokines/immunology , Adipose Tissue/physiopathology , Biomarkers/blood , C-Reactive Protein/analysis , C-Reactive Protein/immunology , Cardiovascular Diseases/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Exosomes/immunology , Fibroblast Growth Factor-23 , Glucuronidase/analysis , Glucuronidase/blood , Glucuronidase/immunology , HMGB Proteins/analysis , HMGB Proteins/blood , HMGB Proteins/immunology , Humans , Immune System/physiopathology , Interleukin-1/analysis , Interleukin-1/blood , Interleukin-1/immunology , Klotho Proteins , Osteoprotegerin/analysis , Osteoprotegerin/blood , Osteoprotegerin/immunology , Prevalence , Serum Amyloid P-Component/analysis , Serum Amyloid P-Component/immunology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
6.
Aging (Albany NY) ; 11(14): 5246-5257, 2019 07 28.
Article in English | MEDLINE | ID: mdl-31386628

ABSTRACT

Circulating Klotho peptide hormone has anti-aging activity and affects tissue maintenance. Hypomorphic mutant Klotho [kl/kl] mice on C57BL/6xC3H, BALB/c and 129 genetic backgrounds, show decreased Klotho expression that correlate with accelerated aging including pre-mature death due to abnormally high levels of serum vitamin D. These mice also show multiple impairments in the immune system. However, it remains unresolved if the defects in the immune system stem from decreased Klotho expression or high vitamin D levels in the serum. Transfer of the kl/kl allele to pure C57BL/6 genetic background [B6-kl/kl] significantly reduced expression of Klotho at all ages. Surprisingly, B6-kl/kl mice showed normalized serum vitamin D levels, amelioration of severe aging-related phenotypes and normal lifespan. This paper reports a detailed analysis of the immune system in B6-kl/kl mice in the absence of detrimental levels of serum vitamin D. Remarkably, the data reveal that in the absence of overt systemic stress, such as abnormally high vitamin D levels, reduced expression of Klotho does not have a major effect on the generation and maintenance of the immune system.


Subject(s)
Bone Marrow/immunology , Glucuronidase/immunology , Glucuronidase/metabolism , Thymus Gland/immunology , Aging , Animals , Klotho Proteins , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Vitamin D
7.
Life Sci ; 234: 116773, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31422095

ABSTRACT

AIMS: NLRP3 inflammasome activation is essential for the development and prognosis of diabetic cardiomyopathy (DCM). The anti-aging protein Klotho is suggested to modulate tissue inflammatory responses. The aim of the present study was to examine the protective effects of Klotho on DCM. MAIN METHODS: A streptozotocin-induced diabetes mouse model was established to assess the effects of Klotho in vivo, which was administered for 12 weeks. The characteristics of type 1 DCM were evaluated by general status, echocardiography, and histopathology. The expression of associated factors was determined by RT-qPCR and western blotting. Parallel experiments to determine the molecular mechanism through which Klotho prevents DCM were performed using H9C2 cells exposed to high glucose (35 mM). KEY FINDINGS: Diabetes-induced increases in serum creatine kinase-muscle/brain and lactate dehydrogenase levels, cardiac fibrosis, cardiomyocyte apoptosis, and cardiac dysfunction were ameliorated by Klotho. Additionally, Klotho suppressed TXNIP expression, NLRP3 inflammasome activation, and expression of the inflammatory cytokines tumor necrosis factor ɑ, interleukin-1ß, and interleukin-18 in vivo. In high glucose-cultured cardiomyocytes, Klotho and N-acetylcysteine significantly downregulated intracellular reactive oxygen species generation and TXNIP/NLRP3 inflammasome activation. Pretreatment of H9C2 cells with NLRP3 siRNA or Klotho prevented high glucose-induced inflammation and apoptosis in H9C2 cells. SIGNIFICANCE: Our results demonstrate that the protective effect of Klotho on diabetes-induced cardiac injury is associated with inhibition of the NLRP3 inflammasome pathway, suggesting its therapeutic potential for DCM.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Diabetic Cardiomyopathies/immunology , Glucuronidase/immunology , Inflammasomes/immunology , Inflammation/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Animals , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/therapeutic use , Cardiotonic Agents/immunology , Cardiotonic Agents/therapeutic use , Cell Line , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/pathology , Glucuronidase/therapeutic use , Inflammation/drug therapy , Inflammation/etiology , Inflammation/pathology , Klotho Proteins , Male , Mice, Inbred C57BL , Rats , Reactive Oxygen Species/immunology
8.
Proc Natl Acad Sci U S A ; 115(48): E11388-E11396, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30413620

ABSTRACT

Located within the brain's ventricles, the choroid plexus produces cerebrospinal fluid and forms an important barrier between the central nervous system and the blood. For unknown reasons, the choroid plexus produces high levels of the protein klotho. Here, we show that these levels naturally decline with aging. Depleting klotho selectively from the choroid plexus via targeted viral vector-induced knockout in Klothoflox/flox mice increased the expression of multiple proinflammatory factors and triggered macrophage infiltration of this structure in young mice, simulating changes in unmanipulated old mice. Wild-type mice infected with the same Cre recombinase-expressing virus did not show such alterations. Experimental depletion of klotho from the choroid plexus enhanced microglial activation in the hippocampus after peripheral injection of mice with lipopolysaccharide. In primary cultures, klotho suppressed thioredoxin-interacting protein-dependent activation of the NLRP3 inflammasome in macrophages by enhancing fibroblast growth factor 23 signaling. We conclude that klotho functions as a gatekeeper at the interface between the brain and immune system in the choroid plexus. Klotho depletion in aging or disease may weaken this barrier and promote immune-mediated neuropathogenesis.


Subject(s)
Aging/immunology , Brain/immunology , Choroid Plexus/immunology , Glucuronidase/immunology , Aging/genetics , Animals , Female , Glucuronidase/genetics , Hippocampus/immunology , Humans , Klotho Proteins , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology
9.
Autoimmunity ; 51(6): 297-303, 2018 09.
Article in English | MEDLINE | ID: mdl-30369266

ABSTRACT

During NET formation, the content of neutrophils granules is released into the intercellular milieu. Consisting of many proteases and ROS species, formed NETs were shown to degrade cytokines (Schauer, Nat Med, 2014); while the content of neutrophil's azurophilic granules proved to contain glycosidases, secreted upon activation (Thaysen-Andersen, JBC, 2015), and formation of autoantibodies to neutrophil beta-glucoronidase was connected with the level of anti-MPO antibodies (Ab) (Martensson, Autoimmunity, 1992). Taking into account these facts, we aimed to investigate the possibility of NET-related changes in glycan composition on circulating IgG molecules and IgG-IgM immune complexes in multiple sclerosis (MS). This autoimmune disorder still has no reliable detection markers or established ways of treatment, besides widely accepted interferon therapy, making it a particularly interesting clinical condition. By applying capture lectin-ELISA, we analysed binding of α2,6 sialyl-specific lectins SNA, PSqL, and core α1,6-fucose specific lectin AAL to circulating IgG and related complexes in five groups of MS patients: untreated (17 persons); undergoing therapy with interferon (IFN) ß-1 b (15 persons), corticosteroids (methylprednisolone) (12 persons) and anti-B-cell monoclonal Ab (12 persons: Ocrelizumab, 6 persons and alemtuzumab, 6 persons). A group of 23 healthy donors served as control. Significant increase in neutrophil elastase activity, observed in the group of patients under corticosteroid treatment was also accompanied by sialyl-specific PSqL and SNA lectin binding to captured IgG molecules. Subsequent analysis demonstrated that sialic acid residues were exposed on free IgG and on circulating IgG-IgM immune complexes. Increased lectin binding was not observed for anti-myelin basic protein (one of the major autoAb in MS) Ab compared to total serum Ab. IFN therapy was accompanied by low neutrophil elastase activity and low amount of circulating immune complexes. Incubation of in vitro generated NETs with human serum revealed the digestion of high-molecular weight immune complexes with subsequent exposure of hidden glycoepitops. Obtained data indicate the potential of neutrophil-derived proteases to modify (partially degrade) circulating immune complexes leading to exposure of internal glycoepitops.


Subject(s)
Autoantibodies/blood , Extracellular Traps/enzymology , Glucuronidase/metabolism , Leukocyte Elastase/metabolism , Multiple Sclerosis/immunology , Adolescent , Adult , Aged , Autoantibodies/immunology , Epitopes/immunology , Epitopes/metabolism , Extracellular Traps/immunology , Female , Glucuronidase/immunology , Glycosylation , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Immunosuppressive Agents/therapeutic use , Leukocyte Elastase/immunology , Male , Middle Aged , Multiple Sclerosis/blood , Multiple Sclerosis/drug therapy , Neutrophils/enzymology , Neutrophils/immunology , Young Adult
10.
Cytokine ; 111: 106-111, 2018 11.
Article in English | MEDLINE | ID: mdl-30138898

ABSTRACT

AIMS: The proarrhythmic effect of fibroblast growth factor 23 (FGF23) was observed in patients with end stage kidney disease (ESKD). However, there is no data on the role of FGF23 and soluble Klotho (sKlotho) in the pathogenesis of atrial fibrillation (AF) beyond ESKD. The aim of the study was to assess the peripheral vein and left atrial (LA) serum levels of FGF23 and sKlotho along with calcium-phosphates parameters in patients with AF undergoing percutaneous radiofrequency pulmonary vein isolation (PVI). METHODS AND RESULTS: Sixty-nine consecutive patients (mean age: 55.8 ±â€¯9.7 years, F/M: 26/43, CHA2DS2-Vasc: 1.7 ±â€¯1.1) with paroxysmal/persistent AF undergoing PVI were included into the study. Blood samples were taken during PVI - baseline from the peripheral vein, then from the LA immediately after a septal puncture. RESULTS: There were significant differences in the concentrations of peripheral and LA serum sKlotho, intact FGF23 (iFGF23), calcium and phosphates; peripheral FGF23, calcium and phosphates levels were significantly higher, and sKlotho levels were significantly lower than the LA concentrations. Serum sKlotho levels correlated with the CHADS2-VASc score (r = 0.254, p = 0.034) and glucose level (r = 0.300, p = 0.005). Serum sKlotho gradient (LA - peripheral vein) correlated with the baseline AF burden in the Holter monitoring (r = -0.389, p = 0.003). PVI efficacy was confirmed in 52 (75%) patients. There was a significant difference in the iFGF23 gradient between patients with AF and without AF (80.3 vs. -47.6 pg/ml, p = 0.009) in the six-month follow-up. A receiver operating characteristic (ROC) analysis revealed that an iFGF23 gradient >28.7 pg/ml (AUC = 0.742, p = 0.002) was a predictor for AF recurrence. CONCLUSIONS: There is a gradient between the LA and peripheral vein in the markers of calcium-phosphate metabolism in patients undergoing PVI. Lower sKlotho and higher iFGF23 serum levels are associated with episodes of AF. Serum iFGF23 gradient is a potent predictor for the recurrence of AF.


Subject(s)
Atrial Fibrillation/blood , Fibroblast Growth Factors/blood , Glucuronidase/blood , Atrial Fibrillation/immunology , Atrial Fibrillation/pathology , Atrial Fibrillation/therapy , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/immunology , Follow-Up Studies , Glucuronidase/immunology , Humans , Klotho Proteins , Male , Middle Aged
11.
Int J Mol Sci ; 19(9)2018 Aug 21.
Article in English | MEDLINE | ID: mdl-30134544

ABSTRACT

Immunosuppression may occur for a number of reasons related to an individual's frailty, debility, disease or from therapeutic iatrogenic intervention or misadventure. A large percentage of morbidity and mortality in immunodeficient populations is related to an inadequate response to infectious agents with slow response to antibiotics, enhancements of antibiotic resistance in populations, and markedly increased prevalence of acute inflammatory response, septic and infection related death. Given known relationships between intracellular calcium ion concentrations and cytotoxicity and cellular death, we looked at currently available data linking blockade of calcium ion channels and potential decrease in expression of sepsis among immunosuppressed patients. Notable are relationships between calcium, calcium channel, vitamin D mechanisms associated with sepsis and demonstration of antibiotic-resistant pathogens that may utilize channels sensitive to calcium channel blocker. We note that sepsis shock syndrome represents loss of regulation of inflammatory response to infection and that vitamin D, parathyroid hormone, fibroblast growth factor, and klotho interact with sepsis defense mechanisms in which movement of calcium and phosphorus are part of the process. Given these observations we consider that further investigation of the effect of relatively inexpensive calcium channel blockade agents of infections in immunosuppressed populations might be worthwhile.


Subject(s)
Calcium Channel Blockers/therapeutic use , Calcium Channels/immunology , Communicable Diseases/drug therapy , Immunocompromised Host , Sepsis/drug therapy , Calcium/immunology , Calcium/metabolism , Calcium Channels/genetics , Communicable Diseases/genetics , Communicable Diseases/immunology , Communicable Diseases/mortality , Drug Resistance, Microbial/genetics , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/immunology , Gene Expression Regulation , Glucuronidase/genetics , Glucuronidase/immunology , Humans , Klotho Proteins , Parathyroid Hormone/genetics , Parathyroid Hormone/immunology , Phosphorus/immunology , Phosphorus/metabolism , Risk , Sepsis/genetics , Sepsis/immunology , Sepsis/mortality , Survival Analysis , Vitamin D/immunology , Vitamin D/metabolism
12.
J Histochem Cytochem ; 66(4): 305-319, 2018 04.
Article in English | MEDLINE | ID: mdl-29290138

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) are implicated as inflammatory mediators in a variety of settings, including chemokine activation, which is required to recruit circulating leukocytes to infection sites. Heparan sulfate (HS) polysaccharide chains are highly interactive and serve co-receptor roles in multiple ligand:receptor interactions. HS may also serve as a storage depot, sequestering ligands such as cytokines and restricting their access to binding partners. Heparanase, through its ability to fragment HS chains, is a key regulator of HS function and has featured prominently in studies of HS's involvement in inflammatory processes. This review focuses on recent discoveries regarding the role of HSPGs, HS, and heparanase during inflammation, with particular focus on the brain. HS chains emerge as critical go-betweens in multiple aspects of the inflammatory response-relaying signals between receptors and cells. The molecular interactions proposed to occur between HSPGs and the pathogen receptor toll-like receptor 4 (TLR4) are discussed, and we summarize some of the contrasting roles that HS and heparanase have been assigned in diseases associated with chronic inflammatory states, including Alzheimer's disease (AD). We conclude by briefly discussing how current knowledge could potentially be applied to augment HS-mediated events during sustained neuroinflammation, which contributes to neurodegeneration in AD.


Subject(s)
Alzheimer Disease/immunology , Brain/immunology , Heparan Sulfate Proteoglycans/immunology , Immunity, Innate , Inflammation/immunology , Neuroglia/immunology , Alzheimer Disease/pathology , Animals , Brain/pathology , Cytokines/immunology , Glucuronidase/immunology , Heparitin Sulfate/immunology , Humans , Inflammation/pathology , Neuroglia/pathology , Neuroimmunomodulation , Toll-Like Receptor 4/immunology
13.
J Histochem Cytochem ; 66(4): 321-336, 2018 04.
Article in English | MEDLINE | ID: mdl-29290153

ABSTRACT

Key events that occur during inflammation include the recruitment, adhesion, and transmigration of leukocytes from the circulation to the site of inflammation. These events are modulated by chemokines, integrins, and selectins and the interaction of these molecules with glycosaminoglycans, predominantly heparan sulfate (HS). The development of HS/heparin mimetics that interfere or inhibit the interactions that occur between glycosaminoglycans and modulators of inflammation holds great potential for use as anti-inflammatory therapeutics. This review will detail the role of HS in the events that occur during inflammation, their interaction and modulation of inflammatory mediators, and the current advances in the development of HS/heparin mimetics as anti-inflammatory biotherapeutics.


Subject(s)
Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/pharmacology , Drug Discovery , Heparitin Sulfate/immunology , Heparitin Sulfate/pharmacology , Immunity, Innate , Inflammation/immunology , Animals , Anti-Inflammatory Agents/chemistry , Biomimetics/methods , Chemokines/immunology , Drug Discovery/methods , Glucuronidase/antagonists & inhibitors , Glucuronidase/immunology , Heparitin Sulfate/chemistry , Humans , Immunity, Innate/drug effects , Inflammation/drug therapy
14.
Sci Rep ; 7(1): 14956, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29097791

ABSTRACT

The heparan sulfate endoglycosidase heparanase (HPSE) is involved in tumor growth, chronic inflammation and fibrosis. Since a role for HPSE in chronic liver disease has not been demonstrated to date, the current study was aimed at investigating the involvement of HPSE in the pathogenesis of chronic liver injury. Herein, we revealed that HPSE expression increased in mouse livers after carbon tetrachloride (CCl4)-mediated chronic induction of fibrosis, but with a trend to decline during progression of the disease. In mouse fibrotic liver tissues HPSE immunostaining was restricted in necro-inflammatory areas, co-localizing with F4/80 macrophage marker and TNF-α. TNF-α treatment induced HPSE expression as well as HPSE secretion in U937 macrophages. Moreover, macrophage-secreted HPSE regulated the expression of α-SMA and fibronectin in hepatic stellate LX-2 cells. Finally, HPSE activity increased in the plasma of patients with liver fibrosis but it inversely correlated with liver stiffness. Our results suggest the involvement of HPSE in early phases of reaction to liver damage and inflammatory macrophages as an important source of HPSE. HPSE seems to play a key role in the macrophage-mediated activation of hepatic stellate cells (HSCs), thus suggesting that HPSE targeting could be a new therapeutic option in the treatment of liver fibrosis.


Subject(s)
Glucuronidase/analysis , Liver Cirrhosis/pathology , Liver/pathology , Macrophages/pathology , Animals , Carbon Tetrachloride , Cell Line , Glucuronidase/immunology , Hepatic Stellate Cells/immunology , Hepatic Stellate Cells/pathology , Humans , Liver/immunology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/immunology , Macrophages/immunology , Male , Mice, Inbred BALB C
15.
Br J Haematol ; 176(1): 111-117, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27748520

ABSTRACT

Lack of reliable laboratory parameters is the main challenge in the management of fetal and neonatal alloimmune thrombocytopenia (FNAIT). Despite the long-known association between the HLA-DRB3*01:01 allele and human platelet antigen 1a (HPA-1a) alloimmunisation, maternal human leucocyte antigen (HLA) typing has been of little clinical value. Recently, other DRB3 allele variants have been suggested to predict the severity of FNAIT. In this nationwide population-based retrospective cohort study, we performed extensive HLA typing of 96 women, accounting for 87% of our cohort of 110 families with confirmed or possible HPA-1a-immunisation. The HLA type was compared with anti-HPA-1a levels, severity of neonatal disease and responsiveness to maternally administrated intravenous gammaglobulin (IVIG). HLA haplotypes were constructed to investigate further HLA associations. Despite significantly lower anti-HPA-1a levels in DRB3*01:01-negative women, the carrier status of this particular allele could not be used to confirm or rule out FNAIT in the absence of detectable antibodies. In the haplotype analysis, the DRB3*01:01 allele was the actual factor associated with FNAIT. No other HLA allele was shown to be of additional value as a predictor of severe FNAIT or non-responsiveness to IVIG treatment. Thus, HLA genotyping was not found useful in differentiating high- and low-risk pregnancies or in guiding antenatal treatment in affected families.


Subject(s)
Histocompatibility Testing , Predictive Value of Tests , Thrombocytopenia, Neonatal Alloimmune/immunology , Adult , Alleles , Cohort Studies , Female , Genotype , Glucuronidase/immunology , HLA-DRB3 Chains/immunology , Humans , Infant, Newborn , Mothers , Pregnancy , Retrospective Studies , Severity of Illness Index , Thrombocytopenia, Neonatal Alloimmune/diagnosis , Thrombocytopenia, Neonatal Alloimmune/pathology , Young Adult
16.
Drug Resist Updat ; 29: 54-75, 2016 11.
Article in English | MEDLINE | ID: mdl-27912844

ABSTRACT

Heparanase, the sole heparan sulfate degrading endoglycosidase, regulates multiple biological activities that enhance tumor growth, angiogenesis and metastasis. Heparanase expression is enhanced in almost all cancers examined including various carcinomas, sarcomas and hematological malignancies. Numerous clinical association studies have consistently demonstrated that upregulation of heparanase expression correlates with increased tumor size, tumor angiogenesis, enhanced metastasis and poor prognosis. In contrast, knockdown of heparanase or treatments of tumor-bearing mice with heparanase-inhibiting compounds, markedly attenuate tumor progression further underscoring the potential of anti-heparanase therapy for multiple types of cancer. Heparanase neutralizing monoclonal antibodies block myeloma and lymphoma tumor growth and dissemination; this is attributable to a combined effect on the tumor cells and/or cells of the tumor microenvironment. In fact, much of the impact of heparanase on tumor progression is related to its function in mediating tumor-host crosstalk, priming the tumor microenvironment to better support tumor growth, metastasis and chemoresistance. The repertoire of the physio-pathological activities of heparanase is expanding. Specifically, heparanase regulates gene expression, activates cells of the innate immune system, promotes the formation of exosomes and autophagosomes, and stimulates signal transduction pathways via enzymatic and non-enzymatic activities. These effects dynamically impact multiple regulatory pathways that together drive inflammatory responses, tumor survival, growth, dissemination and drug resistance; but in the same time, may fulfill some normal functions associated, for example, with vesicular traffic, lysosomal-based secretion, stress response, and heparan sulfate turnover. Heparanase is upregulated in response to chemotherapy in cancer patients and the surviving cells acquire chemoresistance, attributed, at least in part, to autophagy. Consequently, heparanase inhibitors used in tandem with chemotherapeutic drugs overcome initial chemoresistance, providing a strong rationale for applying anti-heparanase therapy in combination with conventional anti-cancer drugs. Heparin-like compounds that inhibit heparanase activity are being evaluated in clinical trials for various types of cancer. Heparanase neutralizing monoclonal antibodies are being evaluated in pre-clinical studies, and heparanase-inhibiting small molecules are being developed based on the recently resolved crystal structure of the heparanase protein. Collectively, the emerging premise is that heparanase expressed by tumor cells, innate immune cells, activated endothelial cells as well as other cells of the tumor microenvironment is a master regulator of the aggressive phenotype of cancer, an important contributor to the poor outcome of cancer patients and a prime target for therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Gene Expression Regulation, Neoplastic , Glucuronidase/antagonists & inhibitors , Glycoside Hydrolase Inhibitors/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Autophagy/drug effects , Autophagy/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Glucuronidase/genetics , Glucuronidase/immunology , Humans , Immunity, Innate/drug effects , Inflammation , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Signal Transduction , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
17.
Infez Med ; 24(3): 237-40, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27668906

ABSTRACT

A male newborn developed a post-natal cytomegalovirus (CMV) infection, arising in the clinical setting of congenital thrombocytopenia, which was diagnosed as being alloimmune. The evidence of active CMV infection in an infant showing slow-resolution lower airways infection, persistent neonatal and low platelet volume thrombocytopenia, and diffuse eczema (associated to very high levels of serum immunoglobulin E) led to the diagnosis of Wiskott-Aldrich syndrome (WAS) before the third month of life, despite the presence of several confounding clinical factors. The correct interpretation of all clinical features supported the precocious diagnosis of WAS.


Subject(s)
Cytomegalovirus Infections/etiology , Hypergammaglobulinemia/etiology , Immunoglobulin E/blood , Thrombocytopenia, Neonatal Alloimmune/etiology , Wiskott-Aldrich Syndrome/diagnosis , Adult , Cytomegalovirus Infections/immunology , Early Diagnosis , Eczema/etiology , Female , Glucuronidase/immunology , Humans , Immunity, Maternally-Acquired , Immunocompromised Host , Infant , Male , Milk Hypersensitivity/complications , Thrombocytopenia, Neonatal Alloimmune/immunology , Wiskott-Aldrich Syndrome/complications , Wiskott-Aldrich Syndrome/immunology
18.
Proc Natl Acad Sci U S A ; 113(3): 704-9, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26729870

ABSTRACT

Heparanase is an endoglycosidase that cleaves heparan sulfate side chains of proteoglycans, resulting in disassembly of the extracellular matrix underlying endothelial and epithelial cells and associating with enhanced cell invasion and metastasis. Heparanase expression is induced in carcinomas and sarcomas, often associating with enhanced tumor metastasis and poor prognosis. In contrast, the function of heparanase in hematological malignancies (except myeloma) was not investigated in depth. Here, we provide evidence that heparanase is expressed by human follicular and diffused non-Hodgkin's B-lymphomas, and that heparanase inhibitors restrain the growth of tumor xenografts produced by lymphoma cell lines. Furthermore, we describe, for the first time to our knowledge, the development and characterization of heparanase-neutralizing monoclonal antibodies that inhibit cell invasion and tumor metastasis, the hallmark of heparanase activity. Using luciferase-labeled Raji lymphoma cells, we show that the heparanase-neutralizing monoclonal antibodies profoundly inhibit tumor load in the mouse bones, associating with reduced cell proliferation and angiogenesis. Notably, we found that Raji cells lack intrinsic heparanase activity, but tumor xenografts produced by this cell line exhibit typical heparanase activity, likely contributed by host cells composing the tumor microenvironment. Thus, the neutralizing monoclonal antibodies attenuate lymphoma growth by targeting heparanase in the tumor microenvironment.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Glucuronidase/immunology , Lymphoma/pathology , Animals , Antibodies, Monoclonal/pharmacology , Cell Proliferation/drug effects , Glucuronidase/isolation & purification , HEK293 Cells , Humans , Luciferases/metabolism , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Molecular Weight , Neoplasm Metastasis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Saponins/pharmacology , Tumor Burden/drug effects , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
19.
Infect Immun ; 84(1): 241-53, 2016 01.
Article in English | MEDLINE | ID: mdl-26502911

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) causes invasive, drug-resistant skin and soft tissue infections. Reports that S. aureus bacteria survive inside macrophages suggest that the intramacrophage environment may be a niche for persistent infection; however, mechanisms by which the bacteria might evade macrophage phagosomal defenses are unclear. We examined the fate of the S. aureus-containing phagosome in THP-1 macrophages by evaluating bacterial intracellular survival and phagosomal acidification and maturation and by testing the impact of phagosomal conditions on bacterial viability. Multiple strains of S. aureus survived inside macrophages, and in studies using the MRSA USA300 clone, the USA300-containing phagosome acidified rapidly and acquired the late endosome and lysosome protein LAMP1. However, fewer phagosomes containing live USA300 bacteria than those containing dead bacteria associated with the lysosomal hydrolases cathepsin D and ß-glucuronidase. Inhibiting lysosomal hydrolase activity had no impact on intracellular survival of USA300 or other S. aureus strains, suggesting that S. aureus perturbs acquisition of lysosomal enzymes. We examined the impact of acidification on S. aureus intramacrophage viability and found that inhibitors of phagosomal acidification significantly impaired USA300 intracellular survival. Inhibition of macrophage phagosomal acidification resulted in a 30-fold reduction in USA300 expression of the staphylococcal virulence regulator agr but had little effect on expression of sarA, saeR, or sigB. Bacterial exposure to acidic pH in vitro increased agr expression. Together, these results suggest that S. aureus survives inside macrophages by perturbing normal phagolysosome formation and that USA300 may sense phagosomal conditions and upregulate expression of a key virulence regulator that enables its intracellular survival.


Subject(s)
Cathepsin D/immunology , Glucuronidase/immunology , Lysosomal Membrane Proteins/immunology , Macrophages/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Bacterial Proteins/biosynthesis , Cell Line , Humans , Macrophages/enzymology , Macrophages/microbiology , Microbial Viability/immunology , Phagocytosis/immunology , Phagosomes/microbiology , Sigma Factor/biosynthesis , Staphylococcal Infections/microbiology , Trans-Activators/biosynthesis , Transcription Factors , Virulence Factors
20.
Biochem Biophys Res Commun ; 469(4): 878-83, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26713365

ABSTRACT

To explore possible roles of heparanase in cancer-host crosstalk, we examined whether heparanase influences expression of inflammatory chemokines in colorectal cancer cells. Murine colorectal carcinoma cells incubated with heparanase upregulated MCP-1, KC, and RANTES genes and released MCP-1 and KC proteins. Heparanase-dependent production of IL-8 was detected in two human colorectal carcinoma cell lines. Addition of a heparanase inhibitor Heparastatin (SF4) did not influence MCP-1 production, while both latent and mature forms of heparanase augmented MCP-1 release, suggesting that heparanase catalytic activity was dispensable for MCP-1 production. In contrast, addition of heparin to the medium suppressed MCP-1 release in a dose-dependent manner. Similarly, targeted suppression of Ext1 by RNAi significantly suppressed cell surface expression of heparan sulfate and MCP-1 production in colon 26 cells. Taken together, it is concluded that colon 26 cells transduce the heparanase-mediated signal through heparan sulfate binding. We propose a novel function for heparanase independent of its endoglycosidase activity, namely as a stimulant for chemokine production.


Subject(s)
Chemokines/immunology , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/immunology , Glucuronidase/immunology , Heparitin Sulfate/immunology , Inflammasomes/immunology , Catalysis , Cell Line, Tumor , Enzyme Activation , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...