Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
JCI Insight ; 7(10)2022 05 23.
Article in English | MEDLINE | ID: mdl-35389892

ABSTRACT

Erythropoietin (EPO) has multiple nonerythropoietic functions, including immune modulation, but EPO's effects in transplantation remain incompletely understood. We tested the mechanisms linking EPO administration to prolongation of murine heterotopic heart transplantation using WT and conditional EPO receptor-knockout (EPOR-knockout) mice as recipients. In WT controls, peritransplant administration of EPO synergized with CTLA4-Ig to prolong allograft survival (P < 0.001), reduce frequencies of donor-reactive effector CD8+ T cells in the spleen (P < 0.001) and in the graft (P < 0.05), and increase frequencies and total numbers of donor-reactive Tregs (P < 0.01 for each) versus CTLA4-Ig alone. Studies performed in conditional EPOR-knockout recipients showed that each of these differences required EPOR expression in myeloid cells but not in T cells. Analysis of mRNA isolated from spleen monocytes showed that EPO/EPOR ligation upregulated macrophage-expressed, antiinflammatory, regulatory, and pro-efferocytosis genes and downregulated selected proinflammatory genes. Taken together, the data support the conclusion that EPO promotes Treg-dependent murine cardiac allograft survival by crucially altering the phenotype and function of macrophages. Coupled with our previous documentation that EPO promotes Treg expansion in humans, the data support the need for testing the addition of EPO to costimulatory blockade-containing immunosuppression regimens in an effort to prolong human transplant survival.


Subject(s)
Erythropoietin , T-Lymphocytes, Regulatory , Abatacept , Allografts , Animals , Epoetin Alfa , Erythropoietin/genetics , Erythropoietin/metabolism , Mice , Myeloid Cells
2.
JCI Insight ; 6(24)2021 12 22.
Article in English | MEDLINE | ID: mdl-34752416

ABSTRACT

TNF ligation of TNF receptor 1 (TNFR1) promotes either inflammation and cell survival by (a) inhibiting RIPK1's death-signaling function and activating NF-κB or (b) causing RIPK1 to associate with the death-inducing signaling complex to initiate apoptosis or necroptosis. The cellular source of TNF that results in RIPK1-dependent cell death remains unclear. To address this, we employed in vitro systems and murine models of T cell-dependent transplant or tumor rejection in which target cell susceptibility to RIPK1-dependent cell death could be genetically altered. We show that TNF released by T cells is necessary and sufficient to activate RIPK1-dependent cell death in target cells and thereby mediate target cell cytolysis independently of T cell frequency. Activation of the RIPK1-dependent cell death program in target cells by T cell-derived TNF accelerates murine cardiac allograft rejection and synergizes with anti-PD1 administration to destroy checkpoint blockade-resistant murine melanoma. Together, the findings uncover a distinct immunological role for TNF released by cytotoxic effector T cells following cognate interactions with their antigenic targets. Manipulating T cell TNF and/or target cell susceptibility to RIPK1-dependent cell death can be exploited to either mitigate or augment T cell-dependent destruction of allografts and malignancies to improve outcomes.


Subject(s)
Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , TCF Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Death , Humans , Mice
3.
PLoS One ; 14(12): e0225857, 2019.
Article in English | MEDLINE | ID: mdl-31790488

ABSTRACT

Although the high-fat-diet-induced metabolic syndrome (MetS) is a precursor of human cardiac pathology, the myocardial metabolic state in MetS is far from clear. The discrepancies in metabolite handling between human and small animal models and the difficulties inherent in obtaining human tissue complicate the identification of the myocardium-specific metabolic response in patients. Here we use the large animal model of swine that develops the hallmark criteria of human MetS. Our comparative metabolomics together with transcriptomics and computational nonnegative matrix factorization (NMF) interpretation of the data exposes significant decline in metabolites related to the fatty acid oxidation, glycolysis, and pentose phosphate pathway. Behind the reversal lies decreased expression of enzymes that operate in the pathways. We showed that diminished glycogen deposition is a metabolic signature of MetS in the pig myocardium. The depletion of glycogen arises from disbalance in expression of genes that break down and synthesize glycogen. We show robust acetoacetate accumulation and activated expression of key enzymes in ketone body formation, catabolism and transporters, suggesting a shift in fuel utilization in MetS. A contrasting enrichment in O-GlcNAcylated proteins uncovers hexosamine pathway and O-GlcNAcase (OGA) expression involvement in the myocardial response to MetS. Although the hexosamine biosynthetic pathway (HBP) activity and the availability of the UDP-GlcNAc substrate in the MetS myocardium is low, the level of O-GlcNacylated proteins is high as the O-GlcNacase is significantly diminished. Our data support the perception of transcriptionally driven myocardial alterations in expression of standard fatty acids, glucose metabolism, glycogen, and ketone body related enzymes and subsequent paucity of their metabolite products in MetS. This aberrant energy metabolism in the MetS myocardium provide insight into the pathogenesis of CVD in MetS.


Subject(s)
Metabolic Networks and Pathways , Metabolic Syndrome/metabolism , Myocardium/metabolism , Animals , Cholesterol, Dietary/adverse effects , Diet , Glycosylation , Male , Metabolome , Metabolomics , N-Acetylglucosaminyltransferases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Risk Factors , Swine , Unsupervised Machine Learning , beta-N-Acetylhexosaminidases/metabolism
4.
Mol Cell Biochem ; 461(1-2): 57-64, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31352610

ABSTRACT

Metabolic syndrome (MetS) is associated with alterations in coronary vascular smooth muscle and endothelial function. The current study examined the contractile response of the isolated coronary arterioles to serotonin in pigs with and without MetS and investigated the signaling pathways responsible for serotonin-induced vasomotor tone. The MetS pigs (8-weeks old) were fed with a hyper-caloric, fat/cholesterol diet and the control animals (lean) were fed with a regular diet for 12 weeks (n = 6/group). The coronary arterioles (90-180 µm in diameter) were dissected from the harvested pig myocardial tissues and the in vitro coronary arteriolar response to serotonin was measured in the presence of pharmacological inhibitors. The protein expressions of phospholipase A2 (PLA2), TXA2 synthase, and the thromboxane-prostanoid (TP) receptor in the pigs' left ventricular tissue samples were measured using Western blotting. Serotonin (10-9-10-5 M) induced dose-dependent contractions of coronary-resistant arterioles in both non-MetS control (lean) and MetS pigs. This effect was more pronounced in the MetS vessels compared with those of non-MetS controls (lean, P < 0.05]. Serotonin-induced contraction of the MetS vessels was significantly inhibited in the presence of the selective PLA2 inhibitor quinacrine (10-6 M), the COX inhibitor indomethacin (10-5 M), and the TP receptor antagonist SQ29548 (10-6 M), respectively (P < 0.05). MetS exhibited significant increases in tissue levels of TXA2 synthase and TP receptors (P < 0.05 vs. lean), respectively. MetS is associated with increased contractile response of porcine coronary arterioles to serotonin, which is in part via upregulation/activation of PLA2, COX, and subsequent TXA2, suggesting that alteration of vasomotor function may occur at an early stage of MetS and juvenile obesity.


Subject(s)
Arterioles/physiopathology , Coronary Vessels/physiopathology , Metabolic Syndrome/physiopathology , Serotonin/pharmacology , Vasoconstriction/drug effects , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Arterioles/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Coronary Vessels/drug effects , Disease Models, Animal , Fatty Acids, Unsaturated/pharmacology , Hydrazines/pharmacology , Indomethacin/pharmacology , Male , Phospholipases A2/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Quinacrine/pharmacology , Receptors, Thromboxane/metabolism , Swine , Thromboxane A2/metabolism
5.
J Am Heart Assoc ; 7(12)2018 06 12.
Article in English | MEDLINE | ID: mdl-29895586

ABSTRACT

BACKGROUND: Mesenchymal stem cell-derived extracellular vesicles (EVs) are believed to be cardioprotective in myocardial infarct. The objective of this study was to examine the effects of human mesenchymal cell-derived EV injection on cardiac function, myocardial blood flow, and vessel density in the setting of chronic myocardial ischemia. METHODS AND RESULTS: Twenty-three Yorkshire swine underwent placement of an ameroid constrictor on their left circumflex artery. Two weeks later, the animals were split into 2 groups: the control group (CON; n=7) and the EV myocardial injection group (MVM; n=10). The MVM group underwent myocardial injection of 50 µg of EVs in 2 mL 0.9% saline into the ischemic myocardium. Five weeks later, the pigs underwent a harvest procedure, and the left ventricular myocardium was analyzed. Absolute blood flow and the ischemic/nonischemic myocardial perfusion ratio were increased in the ischemic myocardium in the MVM group compared with the CON group. Pigs in the MVM group had increased capillary and arteriolar density in the ischemic myocardial tissue compared with CON pigs. There was an increase in expression of the phospho-mitogen-activated protein kinase/mitogen-activated protein kinase ratio, the phospho-endothelial nitric oxide synthase/endothelial nitric oxide synthase ratio, and total protein kinase B in the MVM group compared with CON. There was an increase in cardiac output and stroke volume in the MVM group compared with CON. CONCLUSIONS: In the setting of chronic myocardial ischemia, myocardial injection of human mesenchymal cell-derived EVs increases blood flow to ischemic myocardial tissue by induction of capillary and arteriolar growth via activation of the protein kinase B/endothelial nitric oxide synthase and mitogen-activated protein kinase signaling pathways resulting in increased cardiac output and stroke volume.


Subject(s)
Coronary Circulation , Extracellular Vesicles/transplantation , Hemodynamics , Mesenchymal Stem Cell Transplantation/methods , Myocardial Ischemia/therapy , Neovascularization, Physiologic , Ventricular Function, Left , Animals , Cells, Cultured , Disease Models, Animal , Extracellular Vesicles/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Recovery of Function , Signal Transduction , Sus scrofa
6.
J Vis Exp ; (122)2017 04 02.
Article in English | MEDLINE | ID: mdl-28448010

ABSTRACT

Ischemic heart disease (IHD), or acute coronary syndrome (ACS), is one of the leading causes of death in the United States. IHD is characterized by reduced blood supply to the heart, resulting in the loss of oxygen to and the ensuing necrosis of the heart muscle. The MI model has gained popularity for its use as a short-term ischemia-reperfusion model and a long-term permanent ligation model. Below, we describe a reliable method for the permanent ligation of the LAD. With mouse genetic engineering technology becoming more advanced, and with an increasing availability of quality murine surgical instruments, the mouse has become a popular model for MI surgeries. Our surgical model incorporates the use of an easily reversible anesthetic for the rapid recovery of the mouse; a minimally invasive endotracheal intubation without involving a tracheotomy; and a thoracentesis through the original thoracotomy site without creating an additional incision in the chest, as is done in some other methods, to effectively remove excess blood and air from the chest cavity. This method is comparatively less invasive than other methods, which dramatically reduces surgical and post-surgical complications and mortality and improves reproducibility.


Subject(s)
Coronary Vessels/surgery , Disease Models, Animal , Myocardial Infarction/etiology , Animals , Ligation/methods , Mice , Myocardial Ischemia , Reproducibility of Results
7.
Kidney Int ; 90(4): 774-82, 2016 10.
Article in English | MEDLINE | ID: mdl-27378476

ABSTRACT

Hemolytic uremic syndrome caused by Shiga toxin-producing Escherichia coli (STEC HUS) is a worldwide endemic problem, and its pathophysiology is not fully elucidated. Here we tested whether the mannose-binding lectin (MBL2), an initiating factor of lectin complement pathway activation, plays a crucial role in STEC HUS. Using novel human MBL2-expressing mice (MBL2 KI) that lack murine Mbls (MBL2(+/+)Mbl1(-/-)Mbl2(-/-)), a novel STEC HUS model consisted of an intraperitoneal injection with Shiga toxin-2 (Stx-2) with or without anti-MBL2 antibody (3F8, intraperitoneal). Stx-2 induced weight loss, anemia, and thrombocytopenia and increased serum creatinine, free serum hemoglobin, and cystatin C levels, but a significantly decreased glomerular filtration rate compared with control/sham mice. Immunohistochemical staining revealed renal C3d deposition and fibrin deposition in glomeruli in Stx-2-injected mice. Treatment with 3F8 completely inhibited serum MBL2 levels and significantly attenuated Stx-2 induced-renal injury, free serum hemoglobin levels, renal C3d, and fibrin deposition and preserved the glomerular filtration rate. Thus, MBL2 inhibition significantly protected against complement activation and renal injury induced by Stx-2. This novel mouse model can be used to study the role of complement, particularly lectin pathway-mediated complement activation, in Stx-2-induced renal injury.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Complement C3d/metabolism , Escherichia coli Infections/drug therapy , Hemolytic-Uremic Syndrome/drug therapy , Immunoglobulin G/therapeutic use , Mannose-Binding Lectin/immunology , Shiga Toxin 2/toxicity , Animals , Antibodies, Monoclonal, Murine-Derived , Complement Activation/drug effects , Disease Models, Animal , Escherichia coli Infections/blood , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Gene Knock-In Techniques , Glomerular Filtration Rate , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/microbiology , Humans , Immunohistochemistry , Kidney/immunology , Male , Mannose-Binding Lectin/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Shiga Toxin 2/immunology , Shiga-Toxigenic Escherichia coli/metabolism
8.
Am J Pathol ; 185(2): 347-55, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25482922

ABSTRACT

Myocardial infarction and coagulation disorders are leading causes of disability and death in the world. An important role of the lectin complement pathway in myocardial infarction and coagulation has been demonstrated in mice genetically deficient in lectin complement pathway proteins. However, these studies are limited to comparisons between wild-type and deficient mice and lack the ability to examine reversal/inhibition of injury after disease establishment. We developed a novel mouse that expresses functional human mannose-binding lectin (MBL) 2 under the control of Mbl1 promoter. Serum MBL2 concentrations averaged approximately 3 µg/mL in MBL2(+/+)Mbl1(-/-)Mbl2(-/-) [MBL2 knock in (KI)] mice. Serum MBL2 level in MBL2 KI mice significantly increased after 7 (8 µg/mL) or 14 (9 µg/mL) days of hyperglycemia compared to normoglycemic mice (P < 0.001). Monoclonal antibody 3F8 inhibited C3 deposition on mannan-coated plates in MBL2 KI, but not wild-type, mice. Myocardial ischemia/reperfusion in MBL2 KI mice revealed that 3F8 preserved cardiac function and decreased infarct size and fibrin deposition in a time-dependent manner. Furthermore, 3F8 prevented ferric chloride-induced occlusive arterial thrombogenesis in vivo. MBL2 KI mice represent a novel animal model that can be used to study the lectin complement pathway in acute and chronic models of human disease. Furthermore, these novel mice demonstrate the therapeutic window for MBL2 inhibition for effective treatment of disease and its complications.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Neutralizing/pharmacology , Disease Models, Animal , Mannose-Binding Lectin/antagonists & inhibitors , Myocardial Infarction/drug therapy , Thrombosis/drug therapy , Animals , Gene Knock-In Techniques , Humans , Mannose-Binding Lectin/blood , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/metabolism , Mice , Mice, Knockout , Myocardial Infarction/blood , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Promoter Regions, Genetic , Thrombosis/blood , Thrombosis/genetics , Thrombosis/pathology
9.
Circulation ; 126(18): 2227-35, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23032324

ABSTRACT

BACKGROUND: Coagulation disorders and reperfusion of ischemic myocardium are major causes of morbidity and mortality. Lectin pathway initiation complexes are composed of multimolecular carbohydrate recognition subcomponents and 3 lectin pathway-specific serine proteases. We have recently shown that the lectin pathway-specific carbohydrate recognition subcomponent mannose-binding lectin plays an essential role in the pathophysiology of thrombosis and ischemia/reperfusion injury. Thus, we hypothesized that the endogenous mannose-binding lectin (MBL)/ficolin-associated protein-1 (MAP-1) that inhibits complement activation in vitro also could be an in vivo regulator by attenuating myocardial schema/reperfusion injury and thrombogenesis when used at pharmacological doses in wild-type mice. METHODS AND RESULTS: In 2 mouse models, MAP-1 preserves cardiac function, decreases infarct size, decreases C3 deposition, inhibits MBL deposition, and prevents thrombogenesis. Furthermore, we demonstrate that MAP-1 displaces MBL/ficolin-associated serine protease (MASP)-1, MASP-2, and MASP-3 from the MBL complex. CONCLUSIONS: Our results suggest that the natural, endogenous inhibitor MAP-1 effectively inhibits lectin pathway activation in vivo. MAP-1 at pharmacological doses represents a novel therapeutic approach for human diseases involving the lectin pathway and its associated MASPs.


Subject(s)
Anticoagulants/therapeutic use , Carotid Artery Thrombosis/drug therapy , Complement Pathway, Mannose-Binding Lectin/drug effects , Mannose-Binding Protein-Associated Serine Proteases/antagonists & inhibitors , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/prevention & control , Animals , Anticoagulants/pharmacology , Carotid Artery Thrombosis/chemically induced , Complement C3/analysis , Complement Pathway, Mannose-Binding Lectin/physiology , Depression, Chemical , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Lectins/metabolism , Mannose-Binding Protein-Associated Serine Proteases/deficiency , Mannose-Binding Protein-Associated Serine Proteases/genetics , Mannose-Binding Protein-Associated Serine Proteases/pharmacology , Mannose-Binding Protein-Associated Serine Proteases/physiology , Mannose-Binding Protein-Associated Serine Proteases/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Cardiovascular , Models, Immunological , Molecular Weight , Multiprotein Complexes/drug effects , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/diagnostic imaging , Myocardial Reperfusion Injury/pathology , Protein Binding , Recombinant Fusion Proteins/metabolism , Ultrasonography , Ficolins
10.
Circulation ; 126(12): 1484-94, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22879370

ABSTRACT

BACKGROUND: The involvement of the complement system in brain injury has been scarcely investigated. Here, we document the pivotal role of mannose-binding lectin (MBL), one of the recognition molecules of the lectin complement pathway, in brain ischemic injury. METHODS AND RESULTS: Focal cerebral ischemia was induced in mice (by permanent or transient middle cerebral artery occlusion) and rats (by 3-vessel occlusion). We first observed that MBL is deposited on ischemic vessels up to 48 hours after injury and that functional MBL/MBL-associated serine protease 2 complexes are increased. Next, we demonstrated that (1) MBL(-/-) mice are protected from both transient and permanent ischemic injury; (2) Polyman2, the newly synthesized mannosylated molecule selected for its binding to MBL, improves neurological deficits and infarct volume when given up to 24 hours after ischemia in mice; (3) anti-MBL-A antibody improves neurological deficits and infarct volume when given up to 18 hours after ischemia, as assessed after 28 days in rats. CONCLUSIONS: Our data show an important role for MBL in the pathogenesis of brain ischemic injury and provide a strong support to the concept that MBL inhibition may be a relevant therapeutic target in humans, one with a wide therapeutic window of application.


Subject(s)
Brain Ischemia/physiopathology , Infarction, Middle Cerebral Artery/physiopathology , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Brain Edema/drug therapy , Brain Edema/genetics , Brain Edema/physiopathology , Brain Ischemia/drug therapy , Brain Ischemia/genetics , Disease Models, Animal , Humans , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/genetics , Male , Mannans/metabolism , Mannans/pharmacology , Mannose-Binding Lectin/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Rats , Rats, Inbred Strains
11.
Front Immunol ; 3(15)2012 Feb 12.
Article in English | MEDLINE | ID: mdl-22375142

ABSTRACT

Hyperglycemia, in the absence of type 1 or 2 diabetes, is an independent risk factor for cardiovascular disease. We have previously demonstrated a central role for mannose binding lectin (MBL)-mediated cardiac dysfunction in acute hyperglycemic mice. In this study, we applied whole-genome microarray data analysis to investigate MBL's role in systematic gene expression changes. The data predict possible intracellular events taking place in multiple cellular compartments such as enhanced insulin signaling pathway sensitivity, promoted mitochondrial respiratory function, improved cellular energy expenditure and protein quality control, improved cytoskeleton structure, and facilitated intracellular trafficking, all of which may contribute to the organismal health of MBL null mice against acute hyperglycemia. Our data show a tight association between gene expression profile and tissue function which might be a very useful tool in predicting cellular targets and regulatory networks connected with in vivo observations, providing clues for further mechanistic studies.

12.
Am J Pathol ; 180(1): 104-12, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22079428

ABSTRACT

Diabetes, stress, pharmaceuticals, surgery, and physical trauma can lead to hyperglycemic conditions. A consistent relationship has been found between chronic inflammation and the cardiovascular complications of hyperglycemia. We hypothesized that cardiomyopathy and vasculopathy resulting from acute hyperglycemia are dependent on mannose-binding lectin (MBL) and lectin complement pathway activation. Hyperglycemia was induced in wild-type (WT) C57BL/6 and MBL-null mice after streptozotocin administration. Echocardiographic data and tissue samples were collected after 4, 7, or 14 days of acute hyperglycemia. Hyperglycemic WT mice demonstrated dilated cardiomyopathy with significantly increased short and long axis area measurements during systole and diastole compared to hyperglycemic MBL-null mice. The EC(50) for acetylcholine-induced relaxation of mesenteric arterioles in WT mice after 4 days of hyperglycemia demonstrated a significant loss of nitric oxide-mediated relaxation compared to normoglycemic WT or hyperglycemic MBL-null mice. Myocardial histochemistry and Western blot analysis revealed a significant influx of macrophages, altered morphology, and increased elastin and collagen deposition in hyperglycemic WT hearts compared to MBL-null hearts. Serum transforming growth factor-ß1 levels were significantly lower in hyperglycemic MBL-null compared to WT mice, suggesting decreased profibrotic signaling. Together, these data suggest that MBL and the lectin complement pathway play a significant role in vascular dysfunction and cardiomyopathy after acute hyperglycemia.


Subject(s)
Cardiomyopathy, Dilated/prevention & control , Complement Activation/physiology , Diabetes Mellitus, Type 1/prevention & control , Diabetic Cardiomyopathies/prevention & control , Hyperglycemia/complications , Mannose-Binding Lectin/physiology , Acetylcholine/pharmacology , Acute Disease , Animals , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/pathology , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/pathology , Diabetic Angiopathies/pathology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/pathology , Hyperglycemia/pathology , Mannose-Binding Lectin/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/physiology , Signal Transduction/physiology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
13.
J Immunol ; 188(2): 885-91, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22156595

ABSTRACT

Bleeding disorders and thrombotic complications constitute a major cause of death and disability worldwide. Although it is known that the complement and coagulation systems interact, no studies have investigated the specific role or mechanisms of lectin-mediated coagulation in vivo. FeCl(3) treatment resulted in intra-arterial occlusive thrombogenesis within 10 min in wild-type (WT) and C2/factor B-null mice. In contrast, mannose-binding lectin (MBL)-null and MBL-associated serine protease (MASP)-1/-3 knockout (KO) mice had significantly decreased FeCl(3)-induced thrombogenesis. Reconstitution with recombinant human (rh) MBL restored FeCl(3)-induced thrombogenesis in MBL-null mice to levels comparable to WT mice, suggesting a significant role of the MBL/MASP complex for in vivo coagulation. Additionally, whole blood aggregation demonstrated increased MBL/MASP complex-dependent platelet aggregation. In vitro, MBL/MASP complexes were captured on mannan-coated plates, and cleavage of a chromogenic thrombin substrate (S2238) was measured. We observed no significant differences in S2238 cleavage between WT, C2/factor B-null, MBL-A(-/-), or MBL-C(-/-) sera; however, MBL-null or MASP-1/-3 KO mouse sera demonstrated significantly decreased S2238 cleavage. rhMBL alone failed to cleave S2238, but cleavage was restored when rMASP-1 was added to either MASP-1/-3 KO sera or rhMBL. Taken together, these findings indicate that MBL/MASP complexes, and specifically MASP-1, play a key role in thrombus formation in vitro and in vivo.


Subject(s)
Blood Coagulation , Carotid Artery Thrombosis/enzymology , Complement Pathway, Mannose-Binding Lectin , Mannose-Binding Protein-Associated Serine Proteases/physiology , Animals , Blood Coagulation/immunology , Carotid Artery Thrombosis/chemically induced , Carotid Artery Thrombosis/genetics , Chlorides/toxicity , Complement Pathway, Mannose-Binding Lectin/genetics , Disease Models, Animal , Ferric Compounds/toxicity , Humans , Immunity, Innate/genetics , Mannose-Binding Lectins/deficiency , Mannose-Binding Lectins/genetics , Mannose-Binding Protein-Associated Serine Proteases/adverse effects , Mannose-Binding Protein-Associated Serine Proteases/deficiency , Mannose-Binding Protein-Associated Serine Proteases/genetics , Mice , Thrombin/physiology
14.
Int Immunopharmacol ; 11(12): 2138-43, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21979496

ABSTRACT

Humoral molecules can trigger injury on mechanically stressed and damaged tissue. We have studied the role of complement 3 (C3) in a mouse model of ventilator-induced lung injury (VILI). Compared with sham-treated wild type (WT) mice, ventilated WT mice have reduced total bronchoalveolar lavage (BAL) cells; and elevated activities of thrombin and matrix metalloproteinases (MMPs), such as gelatinase/collagenase in the BAL fluid. In contrast, these parameters in ventilated C3 null mice are not significantly different from sham-treated WT and C3 null mice. In mechanically ventilated mice, thrombin activity and MMPs are lower in C3 null mice than in WT mice and are inversely correlated with total single BAL cells. C3 activation is associated with MMP activation in vitro. Pretreatment of WT mice with humanized cobra venom factor, which inactivates C3, reduces C3 deposition in the lung and increases total BAL cells in VILI. We propose that C3 is involved with VILI and inhibition of complement activation may be a potential therapeutic strategy.


Subject(s)
Complement C3/metabolism , Ventilator-Induced Lung Injury/immunology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Complement Activation , Complement C3/genetics , Male , Matrix Metalloproteinases/analysis , Mice , Mice, Inbred C57BL , Phagocytes/cytology , Phagocytes/metabolism , Thrombin/analysis
15.
Immunobiology ; 216(1-2): 96-102, 2011.
Article in English | MEDLINE | ID: mdl-20399528

ABSTRACT

The first line of host defense is the innate immune system that includes coagulation factors and pattern recognition molecules, one of which is mannose-binding lectin (MBL). Previous studies have demonstrated that MBL deficiency increases susceptibility to infection. Several mechanisms are associated with increased susceptibility to infection, including reduced opsonophagocytic killing and reduced lectin complement pathway activation. In this study, we demonstrate that MBL and MBL-associated serine protease (MASP)-1/3 together mediate coagulation factor-like activities, including thrombin-like activity. MBL and/or MASP-1/3 deficient hosts demonstrate in vivo evidence that MBL and MASP-1/3 are involved with hemostasis following injury. Staphylococcus aureus infected MBL null mice developed disseminated intravascular coagulation (DIC), which was associated with elevated blood IL-6 levels (but not TNF-α and multi-organ inflammatory responses). Infected MBL null mice also develop liver injury. These findings suggest that MBL deficiency may manifest into DIC and organ failure during infectious diseases.


Subject(s)
Disseminated Intravascular Coagulation/immunology , Mannose-Binding Lectin/metabolism , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Adoptive Transfer , Animals , Blood Coagulation/genetics , Complement Pathway, Mannose-Binding Lectin/genetics , Disease Susceptibility , Disseminated Intravascular Coagulation/epidemiology , Disseminated Intravascular Coagulation/genetics , Disseminated Intravascular Coagulation/physiopathology , Interleukin-6/genetics , Interleukin-6/metabolism , Liver/immunology , Liver/metabolism , Liver/microbiology , Liver/pathology , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/immunology , Mannose-Binding Protein-Associated Serine Proteases/genetics , Mannose-Binding Protein-Associated Serine Proteases/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Risk Factors , Staphylococcal Infections/epidemiology , Staphylococcal Infections/genetics , Staphylococcal Infections/physiopathology , Staphylococcus aureus/pathogenicity , Thrombin/immunology , Thrombin/metabolism
16.
Am J Physiol Heart Circ Physiol ; 297(5): H1853-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19749170

ABSTRACT

Complement activation has been shown to play an important role in the inflammation and tissue injury following myocardial ischemia and reperfusion (MI/R). Several recent studies from our laboratory demonstrated the importance of mannose-binding lectin (MBL) as the initiation pathway for complement activation and the resulting pathological effects following MI/R. However, other studies from the past suggest an important role of the classical pathway and perhaps natural antibodies. In the present study, we used newly generated genetically modified mice that lack secreted IgM (sIgM), MBL-A, and MBL-C (sIgM/MBL null) in a plasma reconstitution mouse model of MI/R. Following 30 min of ischemia and 4 h of reperfusion, left ventricular ejection fractions were significantly higher in sIgM/MBL null mice reconstituted with MBL null or sIgM/MBL null plasma compared with reconstitution with wild-type (WT) plasma or WT mice reconstituted with WT plasma following MI/R. Serum troponin I concentration, myocardial polymorphonuclear leukocyte infiltration, and C3 deposition were dependent on the combined presence of sIgM and MBL. These results demonstrate that MI/R-induced complement activation, inflammation, and subsequent tissue injury require both IgM and MBL. Thus MBL-dependent activation of the lectin pathway may not be completely antibody independent in I/R models.


Subject(s)
Complement Activation , Immunoglobulin M/metabolism , Inflammation/immunology , Mannose-Binding Lectin/metabolism , Myocardial Reperfusion Injury/immunology , Myocardium/immunology , Animals , Biomarkers/blood , Complement Activation/genetics , Complement C3/metabolism , Disease Models, Animal , Immunoglobulin M/deficiency , Immunoglobulin M/genetics , Inflammation/diagnostic imaging , Inflammation/genetics , Inflammation/physiopathology , Male , Mannose-Binding Lectin/deficiency , Mannose-Binding Lectin/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/diagnostic imaging , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Neutrophil Infiltration , Peptide Fragments/blood , Stroke Volume , Troponin I/blood , Ultrasonography , Ventricular Function, Left
17.
J Immunol ; 181(7): 4580-9, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18802060

ABSTRACT

Decay-accelerating factor (DAF) is a cell surface regulator that accelerates the dissociation of C3/C5 convertases and thereby prevents the amplification of complement activation on self cells. In the context of transplantation, DAF has been thought to primarily regulate antibody-mediated allograft injury, which is in part serum complement-dependent. Based on our previously delineated link between DAF and CD4 T cell responses, we evaluated the effects of donor Daf1 (the murine homolog of human DAF) deficiency on CD8 T cell-mediated cardiac allograft rejection. MHC-disparate Daf1(-/-) allografts were rejected with accelerated kinetics compared with wild-type grafts. The accelerated rejection predominantly tracked with DAF's absence on bone marrow-derived cells in the graft and required allograft production of C3. Transplantation of Daf1(-/-) hearts into wild-type allogeneic hosts augmented the strength of the anti-donor (direct pathway) T cell response, in part through complement-dependent proliferative and pro-survival effects on alloreactive CD8 T cells. The accelerated allograft rejection of Daf1(-/-) hearts occurred in recipients lacking anti-donor Abs. The results reveal that donor DAF expression, by controlling local complement activation on interacting T cell APC partners, regulates the strength of the direct alloreactive CD8(+) T cell response. The findings provide new insights into links between innate and adaptive immunity that could be exploited to limit T cell-mediated injury to an allograft following transplantation.


Subject(s)
CD55 Antigens/genetics , Graft Rejection/immunology , Heart Transplantation/immunology , T-Lymphocytes/immunology , Animals , CD55 Antigens/biosynthesis , CD55 Antigens/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Complement Activation/genetics , Complement Activation/immunology , Cytotoxicity, Immunologic/genetics , Graft Rejection/genetics , Graft Rejection/metabolism , Graft Rejection/pathology , Heart Transplantation/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Mice, Transgenic , Pilot Projects , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...