Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 40
1.
Front Immunol ; 13: 752315, 2022.
Article En | MEDLINE | ID: mdl-35222367

The EPICC peptides are a family of peptides that have been developed from the sequence of the capsid protein of human astrovirus type 1 and previously shown to inhibit the classical and lectin pathways of complement. The EPICC peptides have been further optimized to increase aqueous solubility and identify additional mechanisms of action. Our laboratory has developed the lead EPICC molecule, PA-dPEG24 (also known as RLS-0071), which is composed of a 15 amino acid peptide with a C-terminal monodisperse 24-mer PEGylated moiety. RLS-0071 has been demonstrated to possess other mechanisms of action in addition to complement blockade that include the inhibition of neutrophil-driven myeloperoxidase (MPO) activity, inhibition of neutrophil extracellular trap (NET) formation as well as intrinsic antioxidant activity mediated by vicinal cysteine residues contained within the peptide sequence. RLS-0071 has been tested in various ex vivo and in vivo systems and has shown promise for the treatment of both immune-mediated hematological diseases where alterations in the classical complement pathway plays an important pathogenic role as well as in models of tissue-based diseases such as acute lung injury and hypoxic ischemic encephalopathy driven by both complement and neutrophil-mediated pathways (i.e., MPO activity and NET formation). Next generation EPICC peptides containing a sarcosine residue substitution in various positions within the peptide sequence possess aqueous solubility in the absence of PEGylation and demonstrate enhanced complement and neutrophil inhibitory activity compared to RLS-0071. This review details the development of the EPICC peptides, elucidation of their dual-acting complement and neutrophil inhibitory activities and efficacy in ex vivo systems using human clinical specimens and in vivo efficacy in animal disease models.


Extracellular Traps , Peptides , Amino Acid Sequence , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Complement Pathway, Classical , Extracellular Traps/metabolism , Peptides/metabolism , Water
2.
PLoS One ; 16(10): e0259133, 2021.
Article En | MEDLINE | ID: mdl-34710157

Acute lung injury (ALI) often causes severe trauma that may progress to significant morbidity and mortality. ALI results from a combination of the underlying clinical condition of the patient (e.g., inflammation) with a secondary insult such as viral pneumonia or a blood transfusion. While the secondary insult may be variable, the rapidly progressive disease process leading to pulmonary failure is typically mediated by an overwhelming innate immunological or inflammatory reaction driven by excessive complement and neutrophil-mediated inflammatory responses. We recently developed a 'two-hit' ALI rat model mediated by lipopolysaccharide followed by transfusion of incompatible human erythrocytes resulting in complement activation, neutrophil-mediated ALI and free DNA in the blood indicative of neutrophil extracellular trap formation. The objective of this study was to evaluate the role of peptide inhibitor of complement C1 (RLS-0071), a classical complement pathway inhibitor and neutrophil modulator in this animal model. Adolescent male Wistar rats were infused with lipopolysaccharide followed by transfusion of incompatible erythrocytes in the presence or absence of RLS-0071. Blood was collected at various time points to assess complement C5a levels, free DNA and cytokines in isolated plasma. Four hours following erythrocyte transfusion, lung tissue was recovered and assayed for ALI by histology. Compared to animals not receiving RLS-0071, lungs of animals treated with a single dose of RLS-0071 showed significant reduction in ALI as well as reduced levels of C5a, free DNA and inflammatory cytokines in the blood. These results demonstrate that RLS-0071 can modulate neutrophil-mediated ALI in this novel rat model.


Acute Lung Injury/drug therapy , Anti-Inflammatory Agents/therapeutic use , Complement Activation/drug effects , Lung/drug effects , Neutrophil Infiltration/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Anti-Inflammatory Agents/administration & dosage , Cytokines/metabolism , Disease Models, Animal , Erythrocyte Transfusion , Humans , Lipopolysaccharides , Lung/pathology , Male , Rats , Rats, Wistar
3.
Viruses ; 13(8)2021 07 22.
Article En | MEDLINE | ID: mdl-34452288

Herpes simplex virus type 1 (HSV-1) is a prevalent human pathogen primarily transmitted through skin-to-skin contact, especially on and around mucosal surfaces where there is contact with contaminated saliva during periods of viral shedding. It is estimated that 90% of adults worldwide have HSV-1 antibodies. Cutaneous HSV-1 infections are characterized by a sensation of tingling or numbness at the initial infection site followed by an eruption of vesicles and then painful ulcers with crusting. These symptoms can take ten days to several weeks to heal, leading to significant morbidity. Histologically, infections cause ballooning degeneration of keratinocytes and formation of multinucleated giant cells, ultimately resulting in a localized immune response. Commonly prescribed treatments against HSV-1 infections are nucleoside analogs, such as acyclovir (ACV). However, the emergence of ACV-resistant HSV (ACVR-HSV) clinical isolates has created an urgent need for the development of compounds to control symptoms of cutaneous infections. RLS-0071, also known as peptide inhibitor of complement C1 (PIC1), is a 15-amino-acid anti-inflammatory peptide that inhibits classical complement pathway activation and modulates neutrophil activation. It has been previously shown to aid in the healing of chronic diabetic wounds by inhibiting the excessive activation of complement component C1 and infiltration of leukocytes. Here, we report that treatment of cutaneous infections of HSV-1 and ACVR-HSV-1 in BALB/cJ mice with RLS-0071 significantly reduced the rate of mortality, decreased zosteriform spread, and enhanced the healing of the infection-associated lesions compared to control-treated animals. Therefore, RLS-0071 may work synergistically with other antiviral drugs to aid in wound healing of HSV-1 cutaneous infection and may potentially aid in rapid wound healing of other pathology not limited to HSV-1.


Complement Inactivating Agents/therapeutic use , Herpes Simplex/drug therapy , Herpesvirus 1, Human/drug effects , Peptides/therapeutic use , Acyclovir/pharmacology , Animals , Antiviral Agents/pharmacology , Complement Inactivating Agents/pharmacology , Drug Resistance, Viral , Herpes Simplex/pathology , Herpes Simplex/virology , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/genetics , Mice , Mice, Inbred BALB C , Peptides/pharmacology , Thymidine Kinase/genetics
4.
J Hematol ; 10(1): 18-21, 2021 Feb.
Article En | MEDLINE | ID: mdl-33643505

A 14-year-old female patient with sickle cell disease developed a severe delayed hemolytic transfusion reaction (DHTR) leading to multiple transfusions and intensive care management. To better understand the extent to which the classical complement pathway was contributing to her DHTR, we utilized the complement hemolysis using human erythrocytes (CHUHE) assay and the classical complement pathway inhibitor, PIC1. Residual discarded de-identified plasma and erythrocytes from the patient obtained from routine phlebotomy was acquired. These reagents were used in the CHUHE assay in the presence of increasing concentrations of PIC1. Complement-mediated hemolysis of the patient's erythrocytes occurred in her plasma and complement permissive buffer. Increasing concentrations of PIC1 dose-dependently inhibited hemolysis to levels found for the negative control - complement inhibitor buffer. Complement-mediated hemolysis was demonstrated by the CHUHE assay for this patient with sickle cell disease and severe DHTR. PIC1 inhibition of hemolysis suggested that the classical complement pathway was contributing to her DHTR.

5.
Mol Immunol ; 124: 9-17, 2020 08.
Article En | MEDLINE | ID: mdl-32485436

BACKGROUND: Platelet refractoriness remains a challenging clinical dilemma although significant advancements have been made in identifying human leukocyte antigen (HLA) matched or HLA compatible units. Antiplatelet antibodies are the major risk factor for immune-mediated platelet refractoriness, yet the role of antibody-initiated complement-mediated platelet destruction remains poorly understood. STUDY DESIGN AND METHODS: Human complement-mediated opsonization and killing of platelets was assayed ex vivo using antibody-sensitized human platelets incubated with complement-sufficient human sera. A new animal model of platelet refractoriness utilizing Wistar rats transfused with human platelets is described. RESULTS: Human platelets sensitized with anti-platelet antibodies were rapidly opsonized with iC3b upon incubation in human sera. This opsonization could be completely blocked with a classical pathway complement inhibitor, PA-dPEG24. Complement activation decreased platelet viability, which was also reversible with complement inhibitor PA-dPEG24. A new rat model of platelet refractoriness was developed that demonstrated some platelet removal from the blood stream was complement mediated. CONCLUSIONS: Complement activation initiated by anti-platelet antibodies leads to complement opsonization and decreased platelet viability. A new rat model of platelet refractoriness was developed that adds a new tool for elucidating the mechanisms of platelet refractoriness.


Blood Platelets/immunology , Complement Activation/immunology , Disease Models, Animal , Isoantibodies/immunology , Animals , Complement Pathway, Classical , Humans , Male , Rats , Rats, Wistar , Transplantation, Heterologous
6.
PLoS One ; 15(4): e0230482, 2020.
Article En | MEDLINE | ID: mdl-32310973

Acute transfusion reactions can manifest in many forms including acute hemolytic transfusion reaction, allergic reaction and transfusion-related acute lung injury. We previously developed an acute hemolytic transfusion reaction rat model mediated by transfusion of incompatible human erythrocytes against which rats have preexisting antibodies resulting in classical complement pathway mediated intravascular hemolysis. In this study, the acute hemolytic transfusion reaction model was adapted to yield an acute lung injury phenotype. Adolescent male Wistar rats were primed in the presence or absence of lipopolysaccharide followed by transfusion of incompatible erythrocytes. Blood was collected at various time points during the course of the experiment to determine complement C5a levels and free DNA in isolated plasma. At 4 hours, blood and lung tissue were recovered and assayed for complete blood count and histological acute lung injury, respectively. Compared to sham animals or animals receiving increasing amounts of incompatible erythrocytes (equivalent to a 15-45% transfusion) in the absence of lipopolysaccharide, lungs of animals receiving lipopolysaccharide and a 30% erythrocyte transfusion showed dramatic alveolar wall thickening due to neutrophil infiltration. C5a levels were significantly elevated in these animals indicating that complement activation contributes to lung damage. Additionally, these animals demonstrated a significant increase of free DNA in the blood over time suggestive of neutrophil extracellular trap formation previously associated with transfusion-related acute lung injury in humans and mice. This novel 'two-hit' model utilizing incompatible erythrocyte transfusion in the presence of lipopolysaccharide yields a robust acute lung injury phenotype.


Acute Lung Injury , Disease Models, Animal , Erythrocyte Transfusion , Lipopolysaccharides/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Animals , Blood Group Incompatibility/metabolism , Complement C5a/metabolism , DNA/blood , Erythrocytes/metabolism , Extracellular Traps/metabolism , Humans , Male , Neutrophil Infiltration , Rats , Rats, Wistar , Transfusion Reaction/pathology
7.
PLoS One ; 14(12): e0226875, 2019.
Article En | MEDLINE | ID: mdl-31891617

BACKGROUND: A product of rational molecular design, PA-dPEG24 is the lead derivative of the PIC1 family of peptides with multiple functional abilities including classical complement pathway inhibition, myeloperoxidase inhibition, NET inhibition and antioxidant activity. PA-dPEG24 is composed of a sequence of 15 amino acid, IALILEPICCQERAA, and contains a monodisperse 24-mer PEGylated moiety at its C terminus to increase aqueous solubility. Here we explore a sarcosine substitution scan of the PA peptide to evaluate impacts on solubility in the absence of PEGylation and functional characteristics. METHODS: Sixteen sarcosine substitution variants were synthesized and evaluated for solubility in water. Aqueous soluble variants were then tested in standard complement, myeloperoxidase, NET formation and antioxidant capacity assays. RESULTS: Six sarcosine substitution variants were aqueous soluble without requiring PEGylation. Substitution with sarcosine of the isoleucine at position eight yielded a soluble peptide that surpassed the parent molecule for complement inhibition and myeloperoxidase inhibition. Substitution with sarcosine of the cysteine at position nine improved solubility, but did not otherwise change the functional characteristics compared with the parent compound. However, replacement of both vicinal cysteine residues at positions 9 and 10 with a single sarcosine residue reduced functional activity in most of the assays tested. CONCLUSIONS: Several of the sarcosine PIC1 variant substitutions synthesized yielded improved solubility as well as a number of unanticipated structure-function findings that provide new insights. Several sarcosine substitution variants demonstrate increased potency over the parent peptide suggesting enhanced therapeutic potential for inflammatory disease processes involving complement, myeloperoxidase, NETs or oxidant stress.


Antioxidants/pharmacology , Complement Activation/drug effects , Complement Inactivating Agents/pharmacology , Extracellular Traps/drug effects , Peptides/pharmacology , Peroxidase/antagonists & inhibitors , Sarcosine/pharmacology , Amino Acid Sequence , Antioxidants/chemistry , Complement Inactivating Agents/chemistry , Humans , Oxidation-Reduction/drug effects , Peptides/chemistry , Sarcosine/chemistry , Solubility , Water/chemistry
8.
Front Immunol ; 9: 558, 2018.
Article En | MEDLINE | ID: mdl-29632531

Two major aspects of systemic lupus erythematosus (SLE) pathogenesis that have yet to be targeted therapeutically are immune complex-initiated complement activation and neutrophil extracellular trap (NET) formation by neutrophils. Here, we report in vitro testing of peptide inhibitor of complement C1 (PIC1) in assays of immune complex-mediated complement activation in human sera and assays for NET formation by human neutrophils. The lead PIC1 derivative, PA-dPEG24, was able to dose-dependently inhibit complement activation initiated by multiple types of immune complexes (IC), including C1-anti-C1q IC, limiting the generation of pro-inflammatory complement effectors, including C5a and membrane attack complex (sC5b-9). In several instances, PA-dPEG24 achieved complete inhibition with complement effector levels equivalent to background. PA-dPEG24 was also able to dose-dependently inhibit NET formation by human neutrophils stimulated by PMA, MPO, or immune complex activated human sera. In several instances PA-dPEG24 achieved complete inhibition with NETosis with quantitation equivalent to background levels. These results suggest that PA-dPEG24 inhibition of NETs occurs by blocking the MPO pathway of NET formation. Together these results demonstrate that PA-dPEG24 can inhibit immune complex activation of the complement system and NET formation. This provides proof of concept that peptides can potentially be developed to inhibit these two important contributors to rheumatologic pathology that are currently untargeted by available therapies.


Antigen-Antibody Complex/immunology , Complement Activation/immunology , Complement C1 Inhibitor Protein/immunology , Extracellular Traps/immunology , Neutrophils/immunology , Peptides/immunology , Antigen-Antibody Complex/blood , Antigen-Antibody Complex/metabolism , Complement C1/immunology , Complement C1/metabolism , Complement C1 Inhibitor Protein/metabolism , Complement C1q/immunology , Extracellular Traps/metabolism , Humans , Microscopy, Fluorescence , Neutrophils/metabolism , Peptides/metabolism
9.
PLoS One ; 13(3): e0193931, 2018.
Article En | MEDLINE | ID: mdl-29499069

Reactive oxygen species (ROS) are natural byproducts of oxidative respiration that are toxic to organs and tissues. To mitigate ROS damage, organisms have evolved a variety of antioxidant systems to counteract these harmful molecules, however in certain pathological conditions these protective mechanisms can be overwhelmed. We have recently demonstrated that Peptide Inhibitor of Complement C1 (PIC1) mitigates peroxidase activity of the heme bearing proteins myeloperoxidase, hemoglobin, and myoglobin through a reversible process. To determine if this property of PIC1 was antioxidant in nature, we tested PIC1 in a number of well-established antioxidant assays. PIC1 showed dose-dependent antioxidant activity in a total antioxidant (TAC) assay, hydroxyl radical antioxidant capacity (HORAC) assay, oxygen radical antioxidant capacity (ORAC) assay as well as the thiobarbituric acid reactive substances (TBARS) assay to screen for PIC1 antioxidant activity in human plasma. The antioxidant activity of PIC1 in the TAC assay, as well as the HORAC/ORAC assay demonstrated that this peptide acts via the single electron transport (SET) and hydrogen atom transfer (HAT) mechanisms, respectively. Consistent with this mechanism of action, PIC1 did not show activity in a metal chelating activity (MCA) assay. PIC1 contains two vicinal cysteine residues and displayed similar antioxidant activity to the well characterized cysteine-containing tripeptide antioxidant molecule glutathione (GSH). Consistent with the role of the cysteine residues in the antioxidant activity of PIC1, oxidation of these residues significantly abrogated antioxidant activity. These results demonstrate that in addition to its described complement inhibiting activity, PIC1 displays in vitro antioxidant activity.


Antioxidants/metabolism , Complement C1 Inhibitor Protein/metabolism , Electron Transport/physiology , Hydrogen/metabolism , Peptides/metabolism , Complement C1/metabolism , Glutathione/metabolism , Hemoglobins/metabolism , Humans , Hydroxyl Radical/metabolism , Oxidation-Reduction , Peroxidase/metabolism , Reactive Oxygen Species/metabolism
10.
Int J Pept ; 2017: 9454583, 2017.
Article En | MEDLINE | ID: mdl-29081812

Hemoglobin is the natural carrier of oxygen in red blood cells (RBCs). While intracellular hemoglobin provides life-sustaining oxygen transport, extracellular free hemoglobin displays toxicity due to inherent peroxidase activity generating reactive oxygen species that subsequently react with the hemoglobin molecule to produce toxic heme degradation products resulting in free radicals, oxidative stress damage, and lipid peroxidation. We have recently demonstrated that Peptide Inhibitor of Complement C1 (PIC1) inhibits peroxidase activity of the heme-based enzyme myeloperoxidase. To elucidate whether PIC1 could inhibit peroxidase activity of hemoglobin, we evaluated the consequence of PIC1 on RBC lysates, methemoglobin, and myoglobin using tetramethylbenzidine (TMB) as an oxidation target. PIC1 reversibly and dose-dependently prevented TMB oxidation to tetramethylbenzidine diimine by RBC lysates, methemoglobin, and myoglobin, having comparable activity to the inhibitor 4-aminobenzoic acid hydrazide. PIC1 inhibited TMB oxidation of RBC lysates similar to L-cysteine suggesting that the two cysteine residues contained in PIC1 may mediate peroxidase activity. PIC1 also inhibited heme destruction by NaOCl for RBC lysates, hemoglobin, and myoglobin as assayed by preservation of the Soret absorbance peak in the presence of NaOCl and reduction in free iron release. In conclusion, PIC1 inhibits peroxidase activity of hemoglobin and myoglobin likely via an antioxidant mechanism.

11.
PLoS One ; 12(3): e0173257, 2017.
Article En | MEDLINE | ID: mdl-28278205

In cystic fibrosis (CF), lung damage is mediated by a cycle of obstruction, infection, inflammation and tissue destruction. The complement system is a major mediator of inflammation for many diseases with the effectors C5a and C3a often playing important roles. We have previously shown in a small pilot study that CF sputum soluble fraction concentrations of C5a and C3a were associated with clinical measures of CF disease. Here we report a much larger study of 34 CF subjects providing 169 testable sputum samples allowing longitudinal evaluation comparing C5a and C3a with clinical markers. Levels of the strongly pro-inflammatory C5a correlated negatively with FEV1% predicted (P < 0.001), whereas the often anti-inflammatory C3a correlated positively with FEV1% predicted (P = 0.01). C5a concentrations correlated negatively with BMI percentile (P = 0.017), positively with worsening of an acute pulmonary exacerbation score (P = 0.007) and positively with P. aeruginosa growth in sputum (P = 0.002). C5a levels also correlated positively with concentrations of other sputum markers associated with worse CF lung disease including neutrophil elastase (P < 0.001), myeloperoxidase activity (P = 0.006) and DNA concentration (P < 0.001). In contrast to C5a, C3a levels correlated negatively with worse acute pulmonary exacerbation score and correlated negatively with sputum concentrations of neutrophil elastase, myeloperoxidase activity and DNA concentration. In summary, these data suggest that in CF sputum, increased C5a is associated with increased inflammation and poorer clinical measures, whereas increased C3a appears to be associated with less inflammation and improved clinical measures.


Biomarkers/metabolism , Body Fluids/immunology , Complement C3a/metabolism , Complement C5a/metabolism , Cystic Fibrosis/immunology , Inflammation/immunology , Sputum/immunology , Adolescent , Adult , Child , Child, Preschool , Cystic Fibrosis/physiopathology , Female , Humans , Inflammation/physiopathology , Leukocyte Elastase , Longitudinal Studies , Male , Middle Aged , Pilot Projects , Prospective Studies , Severity of Illness Index , Young Adult
12.
PLoS One ; 12(1): e0170500, 2017.
Article En | MEDLINE | ID: mdl-28107529

Diabetic non-healing wounds are a major clinical problem. The mechanisms leading to poor wound healing in diabetes are multifactorial but unresolved inflammation may be a major contributing factor. The complement system (CS) is the most potent inflammatory cascade in humans and contributes to poor wound healing in animal models. Signal transducer and activator of transcription 4 (STAT4) is a transcription factor expressed in immune and adipose cells and contributes to upregulation of some inflammatory chemokines and cytokines. Persistent CS and STAT4 expression in diabetic wounds may thus contribute to chronic inflammation and delayed healing. The purpose of this study was to characterize CS and STAT4 in early diabetic wounds using db/db mice as a diabetic skin wound model. The CS was found to be activated early in the diabetic wounds as demonstrated by increased anaphylatoxin C5a in wound fluid and C3-fragment deposition by immunostaining. These changes were associated with a 76% increase in nucleated cells in the wounds of db/db mice vs. CONTROLS: The novel classical CS inhibitor, Peptide Inhibitor of Complement C1 (PIC1) reduced inflammation when added directly or saturated in an acellular skin scaffold, as reflected by reduced CS components and leukocyte infiltration. A significant increase in expression of STAT4 and the downstream macrophage chemokine CCL2 and its receptor CCR2 were also found in the early wounds of db/db mice compared to non-diabetic controls. These studies provide evidence for two new promising targets to reduce unresolved inflammation and to improve healing of diabetic skin wounds.


Complement Activation/physiology , Diabetes Complications/physiopathology , STAT4 Transcription Factor/physiology , Wounds and Injuries/physiopathology , Animals , Complement C5a/physiology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Wound Healing/physiology , Wounds and Injuries/complications
13.
PLoS One ; 12(1): e0170203, 2017.
Article En | MEDLINE | ID: mdl-28135312

Myeloperoxidase is the major peroxidase enzyme in neutrophil granules and implicated in contributing to inflammatory lung damage in cystic fibrosis. Free myeloperoxidase is present in cystic fibrosis lung fluid and generates hypochlorous acid. Here we report a new inhibitor of myeloperoxidase activity, Peptide Inhibitor of Complement C1 (PIC1). Using TMB as the oxidizing substrate, PIC1 inhibited myeloperoxidase activity in cystic fibrosis sputum soluble fractions by an average of a 3.4-fold decrease (P = 0.02). PIC1 also dose-dependently inhibited myeloperoxidase activity in a neutrophil lysate or purified myeloperoxidase by up to 28-fold (P < 0.001). PIC1 inhibited myeloperoxidase activity similarly, on a molar basis, as the specific myeloperoxidase inhibitor 4-Aminobenzoic acid hydrazide (ABAH) for various oxidizing substrates. PIC1 was able to protect the heme ring of myeloperoxidase from destruction by NaOCl, assayed by spectral analysis. PIC1 incubated with oxidized TMB reversed the oxidation state of TMB, as measured by absorbance at 450 nm, with a 20-fold reduction in oxidized TMB (P = 0.02). This result was consistent with an antioxidant mechanism for PIC1. In summary, PIC1 inhibits the peroxidase activity of myeloperoxidase in CF sputum likely via an antioxidant mechanism.


Complement C1 Inhibitor Protein/metabolism , Cystic Fibrosis/enzymology , Peroxidase/antagonists & inhibitors , Sputum/enzymology , Aniline Compounds/metabolism , Antioxidants/metabolism , Benzidines/metabolism , Heme/metabolism , Humans , Neutrophils/metabolism , Oxidation-Reduction , Peroxidase/metabolism
14.
Transfusion ; 57(3): 517-524, 2017 03.
Article En | MEDLINE | ID: mdl-28000304

BACKGROUND: The agglutination-based cross-matching method is sensitive for antibody binding to red blood cells but is only partially predictive of complement-mediated hemolysis, which is important in many acute hemolytic transfusion reactions. Here, we describe complement hemolysis using human erythrocytes (CHUHE) assays that directly evaluate complement-mediated hemolysis between individual serum-plasma and red blood cell combinations. The CHUHE assay is used to evaluate correlations between agglutination titers and complement-mediated hemolysis as well as the hemolytic potential of plasma from type A blood donors. STUDY DESIGN AND METHODS: Plasma or serum from each type A blood donor was incubated with AB or B red blood cells in the CHUHE assay and measured for free hemoglobin release. RESULTS: CHUHE assays for serum or plasma demonstrate a wide, dynamic range and high sensitivity for complement-mediated hemolysis for individual serum/plasma and red blood cell combinations. CHUHE results suggest that agglutination assays alone are only moderately predictive of complement-mediated hemolysis. CHUHE results also suggest that plasma from particular type A blood donors produce minimal complement-mediated hemolysis, whereas plasma from other type A blood donors produce moderate to high-level complement-mediated hemolysis, depending on the red blood cell donor. CONCLUSION: The current results indicate that the CHUHE assay can be used to assess complement-mediated hemolysis for plasma or serum from a type A blood donor, providing additional risk discrimination over agglutination titers alone.


ABO Blood-Group System/metabolism , Blood Grouping and Crossmatching/methods , Complement System Proteins/metabolism , Erythrocytes/metabolism , Hemolysis , Plasma/metabolism , Female , Humans , Male , Transfusion Reaction/metabolism , Transfusion Reaction/prevention & control
15.
Pediatr Res ; 81(4): 654-662, 2017 Apr.
Article En | MEDLINE | ID: mdl-28002390

BACKGROUND: Therapeutic hypothermia (HT) is the only intervention that improves outcomes in neonatal hypoxic-ischemic encephalopathy (HIE). However, the multifactorial mechanisms by which HT impacts HIE are incompletely understood. The complement system plays a major role in the pathogenesis of ischemia-reperfusion injuries such as HIE. We have previously demonstrated that HT modulates complement activity in vitro. METHODS: Term equivalent rat pups were subjected to unilateral carotid ligation followed by hypoxia (8% O2) for 45 min to simulate HIE. A subset of animals was subjected to HT (31-32°C for 6 h). Plasma and brain levels of C3a and C5a were measured. Receptors for C3a (C3aR) and C5a (C5aR) along with C1q, C3, and C9 were characterized in neurons, astrocytes, and microglia. RESULTS: We found that HT increased systemic expression of C3a and decreased expression of C5a after HIE. In the brain, C3aR and C5aR are predominantly expressed on microglia after HIE. HT increased local expression of C3aR and decreased expression on C5aR after HIE. Furthermore, HT decreased local expression of C1q, C3-products, and C9 in the brain. CONCLUSION: HT is associated with significant alteration of complement effectors and their cognate receptors. Complement modulation may improve outcomes in neonatal HIE.


Brain Diseases/blood , Complement C3a/analysis , Complement C5a/analysis , Hypothermia, Induced , Hypoxia-Ischemia, Brain/blood , Animals , Animals, Newborn , Astrocytes/metabolism , Brain/pathology , Brain Diseases/therapy , Hypoxia , Hypoxia-Ischemia, Brain/therapy , Microglia/metabolism , Neurons/metabolism , Rats , Rats, Wistar , Reperfusion Injury , Temperature , Time Factors
16.
Transfusion ; 56(8): 2133-45, 2016 08.
Article En | MEDLINE | ID: mdl-27282513

BACKGROUND: Acute hemolytic transfusion reactions have a broad clinical presentation from mild and transitory signs and symptoms to shock, disseminated intravascular coagulation, renal failure, and death. We have recently developed a rat model of acute intravascular hemolysis showing that the classical complement pathway mediates antibody-dependent hemolysis. The objective of this study was to evaluate the role of the classical pathway inhibitor peptide inhibitor of complement C1 (PIC1) in this animal model. STUDY DESIGN AND METHODS: Male Wistar rats received a 15% transfusion of human red blood cells (RBCs) and blood was isolated from the animals up to 120 minutes. Animals received PIC1 either 2 minutes before or 0.5 minutes after transfusion. Sham-, vehicle-, and cobra venom factor (CVF)-treated animals were used as control groups with a subset of rats also receiving an equivalent dose of intravenous immunoglobulin (IVIG) before transfusion. Blood was analyzed for transfused RBC survival by flow cytometry and free hemoglobin (Hb) in isolated plasma by spectrophotometry. RESULTS: Vehicle-treated rats showed decreased human RBC survival and increased free Hb as expected. Rats receiving PIC1 before transfusion showed increased human RBC survival and decreased Hb similar to CVF-treated rats. Notably, rats receiving PIC1 after initiation of transfusion showed similar decreases in hemolysis as animals receiving PIC1 before transfusion. Compared to IVIG and saline controls, PIC1-treated animals demonstrated decreased hemolysis and protection from acute kidney injury. CONCLUSIONS: These results demonstrate that PIC1 has efficacy in an animal model of acute intravascular hemolysis in both prevention and rescue scenarios.


Complement C1/antagonists & inhibitors , Hemolysis/drug effects , Peptides/pharmacology , Animals , Blood Group Incompatibility/drug therapy , Erythrocyte Count , Erythrocytes/cytology , Erythrocytes/drug effects , Hemoglobins/metabolism , Humans , Male , Peptides/therapeutic use , Rats , Rats, Wistar , Transfusion Reaction/drug therapy
17.
Transfusion ; 56(7): 1845-8, 2016 07.
Article En | MEDLINE | ID: mdl-26997226

BACKGROUND: A patient with B+ sickle cell disease received 3 units of red blood cells (RBCs) from two O+ donors and developed fever and hypotension after the first unit, consistent with an acute transfusion reaction (ATR). Anti-B titers in plasma from each O+ donor were markedly elevated and nondiscriminatory. In order to evaluate the potential for the transfused units to produce complement-mediated hemolysis of B+ RBCs, hemolytic complement testing was performed. STUDY DESIGN AND METHODS: Plasma from each donor was diluted in veronal buffer and incubated with B+ RBCs, and free hemoglobin was measured by spectrophotometer in the complement hemolysis using human erythrocytes (CHUHE) assay. Peptide inhibitor of complement C1 (PIC1) was used to confirm antibody-initiated complement pathway activation. RESULTS: A 96-fold difference (p = 0.014) in hemolysis was measured between plasma samples from the two O+ donors using the CHUHE assay. The extremely high degree of hemolysis produced by the one plasma was inhibited by PIC1 in a dose-dependent manner. CONCLUSION: These results indicate that hemolytic complement testing with the CHUHE assay can be used to assess the risk of antibody-initiated, complement-mediated hemolysis from a transfusion beyond what can be achieved with antibody titers alone.


ABO Blood-Group System/immunology , Complement Activation/immunology , Hemolysis/immunology , Transfusion Reaction/immunology , Adolescent , Anemia, Sickle Cell/therapy , Antibodies , Blood Group Incompatibility , Female , Humans , Middle Aged , Risk Assessment
18.
Ann Allergy Asthma Immunol ; 116(5): 435-9, 2016 05.
Article En | MEDLINE | ID: mdl-27017558

BACKGROUND: Eosinophilic esophagitis (EoE) is a chronic disease that requires long-term medical management and monitoring. The eosinophil count determined during esophageal biopsy remains the gold standard for diagnosis and monitoring of EoE. Although markers of eosinophil degranulation correlate with symptoms, eosinophil counts do not correlate. Development of a noninvasive, cost-effective biomarker of eosinophil activation for the evaluation of EoE is an unmet medical need. OBJECTIVE: To conduct a proof-of-concept study to evaluate the potential for measuring urinary 3-bromotyrosine (3-BT) levels in creatinine normalized urine for quantifying eosinophil degranulation in EoE disease. METHODS: A mass spectrometry-based method of measuring normalized 3-BT levels, the Eosinophil Quantitated Urine Kinetic (EoQUIK), was developed, and proof-of-concept evaluation was performed for patients with EoE (n = 27), atopic controls (n = 24), and nonatopic controls (n = 24). RESULTS: EoQUIK revealed that median normalized 3-BT levels were increased 93-fold in patients with EoE compared with nonatopic controls (P = .01) and increased 13-fold in patients with EoE compared with atopic controls (P = .01). Cutoff thresholds were selected for EoQUIK that yielded a specificity of 100% and a negative predictive value of 100% for nonatopic controls and a specificity of 79% and a negative predictive value of 90% for atopic controls. In a logistic regression model, a urine 3-BT level greater than 20 pg per 400 mg of creatinine increased the odds of a patient having EoE by 4.8 (95% confidence interval, 1.14-20.5; P = .03) when compared with atopic controls after controlling for race and sex. CONCLUSION: These data provide proof of concept that EoQUIK can potentially be a useful noninvasive clinical tool in the evaluation of possible EoE.


Eosinophilic Esophagitis/urine , Tyrosine/analogs & derivatives , Adolescent , Adult , Biological Assay , Child , Child, Preschool , Eosinophilic Esophagitis/diagnosis , Eosinophilic Esophagitis/immunology , Eosinophils/immunology , Female , Humans , Leukocyte Count , Male , Tyrosine/urine , Young Adult
19.
PLoS One ; 10(12): e0144723, 2015.
Article En | MEDLINE | ID: mdl-26642048

In cystic fibrosis (CF), lung damage is mediated by a cycle of obstruction, infection, and inflammation. Here we explored complement inflammatory effectors in CF lung fluid. In this study soluble fractions (sols) from sputum samples of 15 CF patients were assayed for complement effectors and analyzed with clinical measurements. The pro-inflammatory peptide C5a was increased 4.8-fold (P = 0.04) in CF sols compared with controls. Incubation of CF sols with P. aeruginosa or S. aureus increased C5a concentration 2.3-fold (P = 0.02). A peptide inhibitor of complement C1 (PIC1) completely blocked the increase in C5a concentration from P. aeruginosa in CF sol in vitro (P = 0.001). C5a concentration in CF sol correlated inversely with body mass index (BMI) percentile in children (r = -0.77, P = 0.04). C3a, which has anti-inflammatory effects, correlated positively with FEV1% predicted (rs = 0.63, P = 0.02). These results suggest that complement effectors may significantly impact inflammation in CF lung fluid.


Body Fluids/immunology , Complement C5a/metabolism , Cystic Fibrosis/immunology , Inflammation/immunology , Lung/immunology , Pseudomonas Infections/immunology , Sputum/immunology , Case-Control Studies , Child , Cystic Fibrosis/physiopathology , Follow-Up Studies , Humans , Inflammation/physiopathology , Lung/physiopathology , Prognosis , Pseudomonas Infections/microbiology , Pseudomonas Infections/physiopathology , Staphylococcus aureus/immunology
20.
PLoS One ; 10(7): e0132446, 2015.
Article En | MEDLINE | ID: mdl-26196285

The complement system has been increasingly recognized to play a pivotal role in a variety of inflammatory and autoimmune diseases. Consequently, therapeutic modulators of the classical, lectin and alternative pathways of the complement system are currently in pre-clinical and clinical development. Our laboratory has identified a peptide that specifically inhibits the classical and lectin pathways of complement and is referred to as Peptide Inhibitor of Complement C1 (PIC1). In this study, we determined that the lead PIC1 variant demonstrates a salt-dependent binding to C1q, the initiator molecule of the classical pathway. Additionally, this peptide bound to the lectin pathway initiator molecule MBL as well as the ficolins H, M and L, suggesting a common mechanism of PIC1 inhibitory activity occurs via binding to the collagen-like tails of these collectin molecules. We further analyzed the effect of arginine and glutamic acid residue substitution on the complement inhibitory activity of our lead derivative in a hemolytic assay and found that the original sequence demonstrated superior inhibitory activity. To improve upon the solubility of the lead derivative, a pegylated, water soluble variant was developed, structurally characterized and demonstrated to inhibit complement activation in mouse plasma, as well as rat, non-human primate and human serum in vitro. After intravenous injection in rats, the pegylated derivative inhibited complement activation in the blood by 90% after 30 seconds, demonstrating extremely rapid function. Additionally, no adverse toxicological effects were observed in limited testing. Together these results show that PIC1 rapidly inhibits classical complement activation in vitro and in vivo and is functional for a variety of animal species, suggesting its utility in animal models of classical complement-mediated diseases.


Complement Activation/drug effects , Complement C1q/immunology , Peptides/chemistry , Peptides/pharmacology , Amino Acid Sequence , Animals , Humans , Injections , Lectins/immunology , Macaca fascicularis , Male , Mannose-Binding Lectin/immunology , Mice , Molecular Sequence Data , Peptides/administration & dosage , Peptides/blood , Rats , Rats, Wistar , Sheep , Ficolins
...