Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 5552, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34548486

ABSTRACT

Sepsis is a life-threatening condition caused by the extreme release of inflammatory mediators into the blood in response to infection (e.g., bacterial infection, COVID-19), resulting in the dysfunction of multiple organs. Currently, there is no direct treatment for sepsis. Here we report an abiotic hydrogel nanoparticle (HNP) as a potential therapeutic agent for late-stage sepsis. The HNP captures and neutralizes all variants of histones, a major inflammatory mediator released during sepsis. The highly optimized HNP has high capacity and long-term circulation capability for the selective sequestration and neutralization of histones. Intravenous injection of the HNP protects mice against a lethal dose of histones through the inhibition of platelet aggregation and migration into the lungs. In vivo administration in murine sepsis model mice results in near complete survival. These results establish the potential for synthetic, nonbiological polymer hydrogel sequestrants as a new intervention strategy for sepsis therapy and adds to our understanding of the importance of histones to this condition.


Subject(s)
Hydrogels/therapeutic use , Nanoparticles/therapeutic use , Sepsis/drug therapy , Animals , Blood Platelets/drug effects , Cell Adhesion , Cell Survival/drug effects , Disease Models, Animal , Histones/antagonists & inhibitors , Histones/metabolism , Histones/toxicity , Hydrogels/chemistry , Hydrogels/metabolism , Hydrogels/pharmacology , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice , Nanoparticles/chemistry , Nanoparticles/metabolism , Platelet Aggregation/drug effects , Polyethylene Glycols/chemistry , Polyethylene Glycols/metabolism , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Protein Binding , Sepsis/mortality , Survival Rate
2.
Nat Commun ; 11(1): 6408, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33328478

ABSTRACT

Extracellular histones in neutrophil extracellular traps (NETs) or in chromatin from injured tissues are highly pathological, particularly when liberated by DNases. We report the development of small polyanions (SPAs) (~0.9-1.4 kDa) that interact electrostatically with histones, neutralizing their pathological effects. In vitro, SPAs inhibited the cytotoxic, platelet-activating and erythrocyte-damaging effects of histones, mechanistic studies revealing that SPAs block disruption of lipid-bilayers by histones. In vivo, SPAs significantly inhibited sepsis, deep-vein thrombosis, and cardiac and tissue-flap models of ischemia-reperfusion injury (IRI), but appeared to differ in their capacity to neutralize NET-bound versus free histones. Analysis of sera from sepsis and cardiac IRI patients supported these differential findings. Further investigations revealed this effect was likely due to the ability of certain SPAs to displace histones from NETs, thus destabilising the structure. Finally, based on our work, a non-toxic SPA that inhibits both NET-bound and free histone mediated pathologies was identified for clinical development.


Subject(s)
Extracellular Traps/drug effects , Histones/metabolism , Polymers/pharmacology , Sepsis/blood , Sepsis/drug therapy , Animals , Erythrocytes/drug effects , Erythrocytes/pathology , Female , Histones/toxicity , Humans , Lipid Bilayers , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocardial Infarction/blood , Platelet Activation/drug effects , Polyelectrolytes , Polymers/chemistry , Rats, Wistar , Reperfusion Injury/blood , Reperfusion Injury/pathology , Sepsis/pathology
3.
J Trauma Acute Care Surg ; 87(3): 614-622, 2019 09.
Article in English | MEDLINE | ID: mdl-31454337

ABSTRACT

BACKGROUND: Extracellular histones are major mediators of organ dysfunction and death in sepsis, and they may cause microcirculatory dysfunction. Heparins have beneficial effects in sepsis and have been reported to bind to histones and neutralize their cytotoxicity. The aim of this study was to investigate the impact of histones on intestinal microcirculation and the intestinal endothelium and to discuss the protective effect of unfractionated heparin (UFH) on the endothelial cytotoxicity and microcirculatory dysfunction induced by histones. METHODS: Anesthetized rats were infused with 30 mg/kg calf thymus histones, and UFH was administered intravenously at a concentration of 100 IU/kg per hour. The intestinal microcirculation was visualized and measured with incident dark field microscope. Plasma von Willebrand factor (vWF) and soluble thrombomodulin were detected, and structural changes in the rat intestinal microvascular endothelium were examined. The effects of histones and UFH on cell survival rates, vWF release and calcium influx were investigated in human intestinal microvascular endothelial cells (HIMECs). RESULTS: Histone infusion caused severe intestinal microcirculatory dysfunction in the absence of obvious hemodynamic changes, and UFH protected intestinal microcirculation in histone-infused rats. Concentrations of the plasma endothelial injury markers vWF and soluble thrombomodulin were elevated, and structural abnormalities were found in the intestinal microvascular endothelium in the histone-infused rats. These events were attenuated by UFH. In vitro, UFH significantly reduced the histone-induced cytotoxicity of HIMECs, reduced the release of vWF from the cytoplasm into the culture medium, and inhibited calcium influx into HIMECs. CONCLUSION: Histones induce intestinal microcirculatory dysfunction followed by direct injury to the endothelial cells; UFH protects the intestinal microcirculation partly by antagonizing the endothelial toxicity of histones.


Subject(s)
Heparin/pharmacology , Histones/toxicity , Intestines/blood supply , Microcirculation/drug effects , Animals , Cell Survival/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/ultrastructure , Humans , In Vitro Techniques , Intestines/drug effects , Male , Microscopy, Electron , Rats , Rats, Wistar
4.
Am J Physiol Heart Circ Physiol ; 316(6): H1309-H1322, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30848676

ABSTRACT

Histone proteins are elevated in the circulation after traumatic injury owing to cellular lysis and release from neutrophils. Elevated circulating histones in trauma contribute to coagulopathy and mortality through a mechanism suspected to involve endothelial cell (EC) dysfunction. However, the functional consequences of histone exposure on intact blood vessels are unknown. Here, we sought to understand the effects of clinically relevant concentrations of histones on the endothelium in intact, resistance-sized, mesenteric arteries (MAs). EC Ca2+ was measured with high spatial and temporal resolution in MAs from mice selectively expressing the EC-specific, genetically encoded ratiometric Ca2+ indicator, Cx40-GCaMP-GR, and vessel diameter was measured by edge detection. Application of purified histone protein directly to the endothelium of en face mouse and human MA preparations produced large Ca2+ signals that spread within and between ECs. Surprisingly, luminal application of histones had no effect on the diameter of pressurized arteries. Instead, after prolonged exposure (30 min), it reduced dilations to endothelium-dependent vasodilators and ultimately caused death of ~25% of ECs, as evidenced by markedly elevated cytosolic Ca2+ levels (793 ± 75 nM) and uptake of propidium iodide. Removal of extracellular Ca2+ but not depletion of intracellular Ca2+ stores prevented histone-induced Ca2+ signals. Histone-induced signals were not suppressed by transient receptor potential vanilloid 4 (TRPV4) channel inhibition (100 nM GSK2193874) or genetic ablation of TRPV4 channels or Toll-like receptor receptors. These data demonstrate that histones are robust activators of noncanonical EC Ca2+ signaling, which cause vascular dysfunction through loss of endothelium-dependent dilation in resistance-sized MAs. NEW & NOTEWORTHY We describe the first use of the endothelial cell (EC)-specific, ratiometric, genetically encoded Ca2+ indicator, Cx40-GCaMP-GR, to study the effect of histone proteins on EC Ca2+ signaling. We found that histones induce an influx of Ca2+ in ECs that does not cause vasodilation but instead causes Ca2+ overload, EC death, and vascular dysfunction in the form of lost endothelium-dependent dilation.


Subject(s)
Calcium Signaling/drug effects , Endothelium, Vascular/drug effects , Histones/toxicity , Mesenteric Arteries/drug effects , Vasodilation/drug effects , Animals , Arterial Pressure , Cell Death , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Mesenteric Arteries/metabolism , Mesenteric Arteries/pathology , Mice, Inbred C57BL , Mice, Knockout , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Toll-Like Receptor 4/metabolism , Vascular Resistance
5.
Cell Death Dis ; 10(3): 167, 2019 02 18.
Article in English | MEDLINE | ID: mdl-30778043

ABSTRACT

Sepsis is life-threatening and often leads to acute brain damage. Dexmedetomidine, an α2-adrenoceptor agonist, has been reported to possess neuroprotective effects against various brain injury but underlying mechanisms remain elusive. In this study, in vitro and in vivo models of sepsis were used to explore the effects of dexmedetomidine on the inflammasome activity and its associated glia pyroptosis and neuronal death. In vitro, inflammasome activation and pyroptosis were found in astrocytes following lipopolysaccharide (LPS) exposure. Dexmedetomidine significantly alleviated astrocyte pyroptosis and inhibited histone release induced by LPS. In vivo, LPS treatment in rats promoted caspase-1 immunoreactivity in astrocytes and caused an increase in the release of pro-inflammatory cytokines of IL-1ß and IL-18, resulting in neuronal injury, which was attenuated by dexmedetomidine; this neuroprotective effect was abolished by α2-adrenoceptor antagonist atipamezole. Dexmedetomidine significantly reduced the high mortality rate caused by LPS challenge. Our data demonstrated that dexmedetomidine may protect glia cells via reducing pyroptosis and subsequently protect neurons, all of which may preserve brain function and ultimately improve the outcome in sepsis.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/therapeutic use , Astrocytes/drug effects , Brain/drug effects , Dexmedetomidine/therapeutic use , Pyroptosis/drug effects , Sepsis/drug therapy , Adrenergic alpha-2 Receptor Agonists/pharmacology , Animals , Astrocytes/metabolism , Brain/immunology , Brain/metabolism , Cytokines/pharmacology , Dexmedetomidine/agonists , Dexmedetomidine/pharmacology , Disease Models, Animal , Histones/metabolism , Histones/toxicity , Humans , Inflammasomes/metabolism , Lipopolysaccharides/pharmacology , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroglia/metabolism , Neurons/cytology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , PC12 Cells , Rats , Rats, Sprague-Dawley , Sepsis/immunology , Sepsis/mortality , Tumor Necrosis Factor-alpha/metabolism
6.
Mucosal Immunol ; 12(1): 39-50, 2019 01.
Article in English | MEDLINE | ID: mdl-30115999

ABSTRACT

Extracellular histones are present in the airways because of cell death occurring during inflammation. They promote inflammation and cause tissue damage due to their cationic nature. The anionic phosphoglycoprotein osteopontin (OPN) is expressed at high levels during airway inflammation and has been ascribed both pro- and anti-inflammatory roles. In this study, it was hypothesized that OPN may neutralize the harmful activities of extracellular histones at the airway mucosal surface. In a model of histone-induced acute lung injury, OPN-/- mice showed increased inflammation and tissue injury, and succumbed within 24 h, whereas wild-type mice showed lower degrees of inflammation and no mortality. In lipopolysaccharide-induced acute lung injury, wild-type mice showed less inflammation and tissue injury than OPN-/- mice. In bronchoalveolar lavage fluid from ARDS patients, high levels of OPN and also histone-OPN complexes were detected. In addition, OPN bound to histones with high affinity in vitro, resulting in less cytotoxicity and reduced formation of tissue-damaging neutrophil extracellular traps (NETs). The interaction between OPN and histones was dependent on posttranslational modification of OPN, i.e., phosphorylation. The findings demonstrate a novel role for OPN, modulating the pro-inflammatory and cytotoxic properties of free histones.


Subject(s)
Acute Lung Injury/immunology , Extracellular Traps/immunology , Neutrophils/immunology , Osteopontin/metabolism , Respiratory Distress Syndrome/immunology , Acute Lung Injury/chemically induced , Animals , Cells, Cultured , Cytotoxicity, Immunologic , Disease Models, Animal , Extracellular Space , Histones/toxicity , Humans , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteopontin/genetics , Phosphorylation
7.
Am J Transplant ; 18(8): 1890-1903, 2018 08.
Article in English | MEDLINE | ID: mdl-29446207

ABSTRACT

Clinical evidence has indicated a possible link between renal injury and remote liver injury. We investigated whether extracellular histone mediates remote hepatic damage after renal graft ischemia-reperfusion injury, while vascular endothelial growth factor (VEGF) is protective against remote hepatic injury. In vitro, hepatocyte HepG2 cultures were treated with histone. In vivo, the Brown-Norway renal graft was stored in 4°C preservation solution for 24 hours and then transplanted into a Lewis rat recipient; blood samples and livers from recipients were harvested 24 hours after surgery. Prolonged cold ischemia in renal grafts enhanced liver injury 24 hours after engraftment. Caspase-1, ASC, NLRP3, and AIM2 expressions in hepatocyte, CD68+ -infiltrating macrophages, tissue, and serum interleukin-1ß and -18 were greatly elevated, indicating that pyroptosis occurred in the liver and resulted in acute liver functional impairment. Blocking the caspase-1 pathway decreased the number of necrotic hepatocytes. VEGF treatment suppressed the hepatocyte pyroptosis and liver function was partially restored. Our data suggested that renal allograft ischemia-reperfusion injury is likely associated with acute liver damage due to hepatocyte pyroptosis induced by histone and such injury may be protected by VEGF administration. VEGF, therefore, may serve as a new strategy against other remote organ injuries related to renal transplantation.


Subject(s)
Histones/toxicity , Inflammation/prevention & control , Kidney Transplantation/adverse effects , Liver Diseases/prevention & control , Pyroptosis , Reperfusion Injury/surgery , Vascular Endothelial Growth Factor A/metabolism , Allografts , Animals , Cytoprotection , Inflammation/etiology , Inflammation/metabolism , Liver Diseases/etiology , Liver Diseases/metabolism , Male , Rats , Rats, Inbred BN , Rats, Inbred Lew , Vascular Endothelial Growth Factor A/genetics
8.
J Control Release ; 268: 335-342, 2017 Dec 28.
Article in English | MEDLINE | ID: mdl-29061513

ABSTRACT

Many of macromolecular toxins induce cell death by directly interacting with cells or induction of inflammatory cytokines. Abiotic polymer ligands (PLs) composed of functional monomers are able to bind and neutralize toxins in vivo and are of great interest for efficient antidotes. However, little has been reported about recognition and neutralization of target molecules in the bloodstream because of readily elimination from the bloodstream. Here, we report a rational design of PLs-decorated lipid nanoparticles (PL-NPs) for neutralizing a target toxin in vivo. PL that decorated on the NPs would cooperatively interacts with target biomacromolecules since the lipid molecules in NPs have a high degree of freedom. In the present study, N-isopropylacrylamide based PLs interacting with histones, major mediators of sepsis, were synthesized. Affinity between PL-NPs and histones depends on monomer composition and polymer length. The optimized PL-NP showed little affinity for plasma proteins. The PL-NPs inhibited the toxicity of histones both in vitro and in vivo, suggesting that PLs on the NPs cooperatively bound to histones and neutralized their toxicity. In addition, circulation time of optimized PL was significantly prolonged by the modification onto NPs. These results provide a platform for designing antidote nanoparticles neutralizing toxic biomacromolecules.


Subject(s)
Antidotes/administration & dosage , Histones/toxicity , Nanoparticles/administration & dosage , Animals , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Design , Ligands , Lipids/administration & dosage , Male , Mice, Inbred BALB C , Polymers/administration & dosage
9.
Shock ; 48(6): 666-673, 2017 12.
Article in English | MEDLINE | ID: mdl-28538085

ABSTRACT

Extracellular histones are cationic damage-associated molecular pattern molecules capable of directly inducing cellular injury via charge-mediated interactions with plasma membranes. Accordingly, histones released into the plasma during critical illness are known to contribute to the onset and propagation of lung injury. Vascular injury (with consequent degradation of the endothelial glycocalyx) simultaneously releases anionic heparan sulfate fragments (hexa- to octasaccharides in size) into the plasma. It is unknown whether this endogenous release of heparan sulfate fragments modulates charge-dependent histone cytotoxicity, or if exogenous heparan sulfate fragments could therapeutically attenuate histone-induced lung injury. Using isothermic calorimetry, we found that extracellular histones only bind to heparan sulfate fragments ≥ 10 saccharides in size, suggesting that glycocalyx-derived heparan sulfate hexa/octasaccharides are incapable of intercepting/neutralizing circulating histones. However, we found that even heparan sulfate fragments incapable of histone binding (e.g., tetrasaccharides) attenuated histone-induced lung injury in vivo, suggesting a direct, size-independent protective effect of heparan sulfate. We found that histones had no effect on human neutrophils ex vivo but exerted toll-like receptor-independent cytotoxicity on human pulmonary microvascular endothelial cells in vitro. This cytotoxicity could be prevented by either the addition of negatively charged (i.e., highly sulfated) heparan sulfate tetrasaccharides (incapable of binding histones) or decasaccharides (capable of binding histones). Taken together, our findings suggest that heparan sulfate oligosaccharides may directly exert pulmonary endothelial-protective effects that attenuate histone-mediated lung injury.


Subject(s)
Heparitin Sulfate/pharmacology , Histones/toxicity , Lung Injury , Oligosaccharides/pharmacology , Animals , Heparitin Sulfate/chemistry , Lung Injury/chemically induced , Lung Injury/drug therapy , Lung Injury/metabolism , Lung Injury/pathology , Male , Mice , Oligosaccharides/chemistry
10.
FEBS J ; 284(11): 1688-1699, 2017 06.
Article in English | MEDLINE | ID: mdl-28371406

ABSTRACT

Neutrophil extracellular traps (NET) are formed against pathogens. However, various diseases are directly linked to this meshwork of DNA. The cytotoxic properties of extracellular histones especially seem to be an important trigger during these diseases. Furthermore, NET accumulation on implants is discussed to result in an impaired efficiency or failure, depending on the category of implant. Interestingly, mucins have been investigated as surface coatings potentially capable of reducing neutrophil adhesion. Similarly, polysialic acid was shown to inactivate the cytotoxic properties of extracellular histones. We wanted to combine the probability to decrease the adhesion of neutrophils using mucins with the capability of sialic acid polymers to counteract histone-mediated cytotoxicity. To this end, we elongate cervical mucins using bacterial polysialyltransferases. Subsequent cell-based experiments demonstrated the activity of elongated mucins against histone-mediated cytotoxicity. Thus, polysialylated mucins may represent a novel component to coat implants or to combat diseases with exaggerated NET formation.


Subject(s)
Bacterial Proteins/metabolism , Cervix Mucus/chemistry , Extracellular Traps/physiology , Histones/antagonists & inhibitors , Mucins/metabolism , Neisseria meningitidis/enzymology , Sialic Acids/metabolism , Sialyltransferases/metabolism , Animals , Cattle , Cell Adhesion , Cell Line , Chickens , Estrus , Female , Histones/physiology , Histones/toxicity , In Vitro Techniques , Neutrophils/cytology , Swine
11.
Rheumatology (Oxford) ; 56(6): 1013-1018, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28073955

ABSTRACT

Objective: Recent studies have suggested that aPS-PT antibody is one of the most relevant autoantibodies to APS. This study aimed to demonstrate the pathogenicity of aPS-PT antibody in vivo . Methods: At first, cultured rat vascular endothelial cells (RECs) were exposed to calf thymus-derived histones. Two hours later, lactate dehydrogenase release from the RECs and expression of PS on the cell surface were assessed. Next, we administered an i.v. injection of calf thymus-derived histones into Wistar rats (12.5 µg/g weight of 8-week-old female rats), and 2 h later they were given an i.v. injection of aPS-PT mAb (1.25 mg/g weight, n = 6) or an equal dose of rat IgM as controls (n = 5). Three days later, histological examination was conducted. Results: Calf thymus-derived histones (>12.5 µg/ml) could injure RECs in vitro . Simultaneously, annexin V could bind to the RECs; thereby, this result indicated that cell-free histone exposure of vascular endothelial cells induced cell surface expression of PS, which is naturally present inside the plasma membrane. Thrombosis developed with higher frequency in the rats given an i.v. injection of aPS-PT mAb than in controls. Conclusion: We established a rat model of thrombosis induced by i.v. injection of aPS-PT mAb.


Subject(s)
Antibodies, Monoclonal/pharmacology , Phosphatidylserines/immunology , Prothrombin/immunology , Thrombosis/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Autoantibodies/administration & dosage , Autoantibodies/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Female , Histones/toxicity , Injections, Intravenous , Phosphatidylserines/metabolism , Rats, Wistar
12.
Toxicol Lett ; 264: 106-113, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27825936

ABSTRACT

Furan is a rodent hepatocarcinogen ubiquitously found in the environment and heat-processed foods. Furan undergoes cytochrome P450 2E1-catalyzed bioactivation to cis-2-butene-1,4-dial (BDA), which has been shown to form an electrophilic conjugate (GSH-BDA) with glutathione. Both BDA and GSH-BDA yield covalent adducts with lysine residues in proteins. Dose- and time-dependent epigenetic histone alterations have been observed in furan-treated rats. While the covalent modification of histones by chemical carcinogens has long been proposed, histone-carcinogen adducts have eluded detection in vivo. In this study, we investigated if the covalent modification of histones by furan may occur in vivo prior to epigenetic histone alterations. Using a "bottom-up" methodology, involving the analysis of tryptic peptides by liquid chromatography - high resolution mass spectrometry, we obtained evidence for a cross-link between GSH-BDA and lysine 107 of histone H2B isolated from the livers of male F344 rats treated with tumorigenic doses of furan. This cross-link was detected at the shortest treatment period (90 days) in the lowest dose group (0.92mg/kg body weight/day), prior to the identification of epigenetic changes, and occurred at a lysine residue that is a target for epigenetic modifications and crucial for nucleosome stability. Our results represent the first unequivocal proof of the occurrence of carcinogen-modified histones in vivo and suggest that such modification happens at the initial stages of furan-induced carcinogenesis. This type of alteration may be general in scope, opening new insights into the mechanisms of chemical carcinogenesis/toxicity and new opportunities for the development of early compound-specific biomarkers of exposure.


Subject(s)
Carcinogenesis/drug effects , Carcinogens/toxicity , Furans/toxicity , Histones/toxicity , Animals , Carcinogenicity Tests , Furans/metabolism , Glutathione/chemistry , Liver/chemistry , Liver/metabolism , Male , Peptides/chemistry , Rats , Rats, Inbred F344 , Trypsin/chemistry
13.
Microcirculation ; 23(3): 240-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26806786

ABSTRACT

OBJECTIVE: Extracellular histones have strong toxicity against the vascular ECs, however, the damage is significantly attenuated in the serum. Although several plasma proteins such as albumin, APC, and PTX3 are known to inhibit the actions of histones, it is still unclear as to which plasma proteins play predominant role. The purpose of this study was to search for the major inhibitors in the serum. METHODS: ECs were cultured in serum-free medium and histone H3 was added. The effects of albumin, low-, medium-, and high-concentration of APC and PTX3 were examined by time-lapse morphological observation and by immunofluorescent staining. The treatment effects were also assessed by the cell viability assay. RESULTS: Both 5% and 2.5% albumin, medium- and high-concentration APC, and medium- and high-concentration PTX3 exerted significant protective effect. In case of damage induced by high-concentration histone H3, all of albumin, APC and PTX3 exerted effects in a concentration-dependent manner. Above results were also confirmed by the cell viability assay. CONCLUSION: Because albumin and PTX3 inhibited histone-induced damage at physiological levels found in serum, these proteins are expected to be major histone inhibitors in vivo.


Subject(s)
Albumins/metabolism , C-Reactive Protein/metabolism , Endothelial Cells/metabolism , Histones/toxicity , Serum Amyloid P-Component/metabolism , Animals , Cell Survival/drug effects , Endothelial Cells/pathology , Histones/pharmacology , Rats
14.
Front Biosci (Landmark Ed) ; 20(8): 1259-70, 2015 06 01.
Article in English | MEDLINE | ID: mdl-25961556

ABSTRACT

Although circulating histones were demonstrated as major mediators of death in septic mice models, their roles in septic patients are not clarified. The present study sought to evaluate the clinical relevance of the circulating histone levels in septic children, and the antagonizing effects of heparin on circulating histones. Histone levels in the plasma of septic children were significantly higher than healthy controls, and positively correlated with disease severity. Histone treatment could activate NF-κB pathway of the endothelial cells and induce the secretion of large amount of cytokines that further amplify inflammation, subsequently leading to organ damage. Co-injection of low dose heparin with lethal dose histones could protect mouse from organ damage and death by antagonizing circulating histones, and similar effects were also observed in other septic models. Collectively, these findings indicated that circulating histones might serve as key factors in the pathogenesis of sepsis and their levels in plasma might be a marker for disease progression and prognosis. Furthermore, low dose heparin might be an effective therapy to hamper sepsis progression and reduce the mortality.


Subject(s)
Heparin/therapeutic use , Histones/blood , Protective Agents/therapeutic use , Sepsis/blood , Animals , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Histones/toxicity , Human Umbilical Vein Endothelial Cells , Humans , Infant , Male , Mice , Mice, Inbred C57BL
15.
Blood ; 125(14): 2286-96, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25631771

ABSTRACT

Extracellular histones are mediators of tissue injury and organ dysfunction; therefore they constitute potential therapeutic targets in sepsis, inflammation, and thrombosis. Histone cytotoxicity in vitro decreases in the presence of plasma. Here, we demonstrate that plasma inter-α inhibitor protein (IAIP) neutralizes the cytotoxic effects of histones and decreases histone-induced platelet aggregation. These effects are mediated through the negatively charged glycosaminoglycans (GAGs) chondroitin sulfate and high-molecular-weight hyaluronan (HMW-HA) associated with IAIP. Cell surface anionic glycosaminoglycans heparan sulfate and HA protect the cells against histone-mediated damage in vitro. Surface plasmon resonance showed that both IAIP and HMW-HA directly bind to recombinant histone H4. In vivo neutralization of histones with IAIP and HMW-HA prevented histone-induced thrombocytopenia, bleeding, and lung microvascular thrombosis, decreased neutrophil activation, and averted histone-induced production of inflammatory cytokines and chemokines. IAIP and HMW-HA colocalized with histones in necrotic tissues and areas that displayed neutrophil extracellular traps. Increasing amounts of IAIP-histone complexes detected in the plasma of septic baboons correlated with increase in histones and/or nucleosomes and consumption of plasma IAIP. Our data suggest that IAIP, chondroitin sulfate, and HMW-HA are potential therapeutic agents to protect against histone-induced cytotoxicity, coagulopathy, systemic inflammation, and organ damage during inflammatory conditions such as sepsis and trauma.


Subject(s)
Alpha-Globulins/metabolism , Glycosaminoglycans/metabolism , Hemorrhage/prevention & control , Histones/toxicity , Inflammation/prevention & control , Sepsis/prevention & control , Thrombocytopenia/prevention & control , Thrombosis/prevention & control , Animals , Apoptosis , Blood Coagulation , Blotting, Western , Cells, Cultured , Cytokines/metabolism , Flow Cytometry , Glycocalyx/metabolism , HL-60 Cells , Hemorrhage/etiology , Hemorrhage/metabolism , Humans , Inflammation/etiology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Nucleosomes/metabolism , Papio , Platelet Aggregation , Sepsis/etiology , Sepsis/metabolism , Thrombocytopenia/etiology , Thrombocytopenia/metabolism , Thrombosis/etiology , Thrombosis/metabolism
16.
J Cataract Refract Surg ; 40(11): 1885-93, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25442884

ABSTRACT

PURPOSE: To determine the effect of histones on corneal endothelial cells generated during cataract surgery. SETTING: Kagoshima University Hospital, Kagoshima, Japan. DESIGN: Experimental study. METHODS: Standard phacoemulsification was performed on enucleated pig eyes. Histones in the anterior segment of the eye were determined by immunohistochemistry. Cultured human corneal endothelial cells were exposed to histones for 18 hours, and cell viability was determined by 2-(2-methoxy-4-nitrophenyl)-3-(4-nitro-phenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium, monosodium salt assay. The concentration of interleukin-6 (IL-6) in the culture medium of human corneal endothelial cells was measured using enzyme-linked immunosorbent assay. The effects of signal inhibitors U0126, SB203580, and SP600125 were evaluated. The protective effect of hyaluronan against histones was evaluated in human corneal endothelial cells with and without hyaluronan. RESULTS: Cellular debris containing histones was observed in the anterior chamber of pig eyes after phacoemulsification. Exposure of human corneal endothelial cells to 50 µg/mL of histones or more led to cytotoxic effects. The IL-6 concentration was significantly increased dose dependently after exposure of human corneal endothelial cells to histones (P<.01). The histone-induced IL-6 production was significantly decreased by extracellular signal-regulated kinases 1/2 and p-38 mitogen-activated protein kinase inhibitors (P<.01). Co-incubation of hyaluronan and histones caused formation of histone aggregates, decreased the cytotoxic effects of the histones, and blocked the increase in IL-6 (P<.01). CONCLUSIONS: Histones were released extracellularly during phacoemulsification and exposure of human corneal endothelial cells to histones increased the IL-6 secretion. The intraoperative use of hyaluronan may decrease the cytotoxic effects of histones released during cataract surgery. FINANCIAL DISCLOSURE: No author has a financial or proprietary interest in any material or method mentioned.


Subject(s)
Endothelium, Corneal/drug effects , Histones/toxicity , Hyaluronic Acid/pharmacology , Phacoemulsification , Viscosupplements/pharmacology , Animals , Anterior Chamber/metabolism , Cell Survival , Cells, Cultured , Cytoprotection/drug effects , Endothelium, Corneal/metabolism , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique, Indirect , Histones/metabolism , Humans , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism , Swine , Zonula Occludens-1 Protein/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
17.
J Innate Immun ; 6(6): 819-30, 2014.
Article in English | MEDLINE | ID: mdl-24942226

ABSTRACT

Collateral damage caused by extracellular histones has an immediate impact on morbidity and mortality in many disease models. A significant increase in the levels of extracellular histones is seen in critically ill patients with trauma and sepsis. We showed that histones are released from necrotic cells in patients with invasive skin infections. Under in vitro conditions, endogenous p33, an endothelial surface protein also known as the gC1q receptor, interacts with histones released from damaged endothelial cells. Functional analyses have revealed that recombinantly expressed p33 completely neutralizes the harmful features of histones, i.e. hemolysis of erythrocytes, lysis of endothelial cells and platelet aggregation. We also noted that mice treated with a sublethal dose of histones developed severe signs of hemolysis, thrombocytopenia and lung tissue damage already 10 min after inoculation. These complications were fully counteracted when p33 was administered together with the histones. Moreover, application of p33 significantly improved survival in mice receiving an otherwise lethal dose of histones. Together, our data suggest that treatment with p33 is a promising therapeutic approach in severe infectious diseases.


Subject(s)
Histones/toxicity , Mitochondrial Proteins/pharmacology , Shock , Animals , Blood Platelets/immunology , Blood Platelets/pathology , Endothelial Cells/immunology , Endothelial Cells/pathology , Female , Hemolysis/drug effects , Hemolysis/immunology , Humans , Mice , Mice, Inbred BALB C , Platelet Aggregation/drug effects , Platelet Aggregation/immunology , Recombinant Proteins/pharmacology , Shock/chemically induced , Shock/immunology , Shock/pathology , Shock/prevention & control
19.
J Control Release ; 113(3): 245-54, 2006 Jul 20.
Article in English | MEDLINE | ID: mdl-16806557

ABSTRACT

Protein/peptide-mediated gene delivery has recently emerged as a powerful approach in non-viral gene transfer. In previous studies, we and other groups found that histones efficiently mediate gene transfer (histonefection). Histonefection has been demonstrated to be effective with various members of the histone family. The DNA binding domains and natural nuclear localisation signal sequences make histones excellent candidates for effective gene transfer. In addition, their positive charge promotes binding to anionic molecules and helps them to overcome the negative charge of cells that is an important barrier to cellular penetration. Histonefection appears to have particular promise in cancer gene transfer and therapy.


Subject(s)
Cell Nucleus/metabolism , DNA/metabolism , Histones/metabolism , RNA/metabolism , Transfection/methods , Active Transport, Cell Nucleus , Animals , Cell Membrane/metabolism , Cell Membrane/physiology , DNA/genetics , Endocytosis , Histones/genetics , Histones/toxicity , Humans , RNA/genetics
20.
J Antimicrob Chemother ; 55(5): 735-41, 2005 May.
Article in English | MEDLINE | ID: mdl-15772144

ABSTRACT

OBJECTIVES: Infections with multidrug-resistant microorganisms (e.g. Pseudomonas aeruginosa and Staphylococcus aureus) cause immense complications in wound care and in the treatment of immunosuppressed patients. Like most antimicrobial peptides, histones are relatively small polycationic proteins located in each eukaryotic nucleus, which naturally supercoil DNA. The aim of this study was to investigate the in vitro and in vivo activity of histone H1.2 in infected burn wounds and its potential toxicity. METHODS: To characterize the antimicrobial properties of histone H1.2 against potential causative organisms of burn wound infections, the in vitro radial diffusion assay and modified NCCLS microbroth dilution MIC assay were carried out. Haemolytic and cytotoxic properties were determined in human red blood cells and primary human keratinocytes. In vivo antimicrobial activity was tested in an infected rat burn model with P. aeruginosa (ATCC 27853). All results were compared with the naturally occurring broad-spectrum antimicrobial peptide protegrin-1 and with antibiotics clinically used against the corresponding bacteria. RESULTS: Human histone H1.2 exerted good antimicrobial activity against all tested microorganisms without significant haemolytic activity. Surprisingly, histone H1.2 showed cytotoxicity with an LD50 of 7.91 mg/L in primary human keratinocytes. The in vivo burn model data revealed a significant three-fold higher reduction in bacterial counts within 4 h compared with carrier control. CONCLUSIONS: These findings indicate that histone H1.2 is a potential candidate for use as a local and, because of its low haemolytic activity, systemic antimicrobial agent. However, further investigations are needed to specify the cytotoxicity and the dose-response relationship for histone H1.2.


Subject(s)
Burns/complications , Histones/toxicity , Histones/therapeutic use , Pseudomonas Infections/drug therapy , Wound Infection/drug therapy , Animals , Antimicrobial Cationic Peptides , Bacteria/drug effects , Cells, Cultured , Disease Models, Animal , Erythrocytes/drug effects , Hemolysis , Histones/administration & dosage , Histones/pharmacology , Humans , Keratinocytes/drug effects , Microbial Sensitivity Tests , Proteins/pharmacology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Rats , Skin/cytology , Toxicity Tests , Wound Infection/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...