Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Maria M. Aleman is active.

Publication


Featured researches published by Maria M. Aleman.


Anesthesia & Analgesia | 2012

Procoagulant Activity in Hemostasis and Thrombosis: Virchow’s Triad Revisited

Alisa S. Wolberg; Maria M. Aleman; Karin Leiderman; Kellie R. Machlus

Virchows triad is traditionally invoked to explain pathophysiologic mechanisms leading to thrombosis, alleging concerted roles for abnormalities in blood composition, vessel wall components, and blood flow in the development of arterial and venous thrombosis. Given the tissue-specific bleeding observed in hemophilia patients, it may be instructive to consider the principles of Virchows triad when investigating mechanisms operant in hemostatic disorders as well. Blood composition (the function of circulating blood cells and plasma proteins) is the most well studied component of the triad. For example, increased levels of plasma procoagulant proteins such as prothrombin and fibrinogen are established risk factors for thrombosis, whereas deficiencies in plasma factors VIII and IX result in bleeding (hemophilia A and B, respectively). Vessel wall (cellular) components contribute adhesion molecules that recruit circulating leukocytes and platelets to sites of vascular damage, tissue factor, which provides a procoagulant signal of vascular breach, and a surface upon which coagulation complexes are assembled. Blood flow is often characterized by 2 key variables: shear rate and shear stress. Shear rate affects several aspects of coagulation, including transport rates of platelets and plasma proteins to and from the injury site, platelet activation, and the kinetics of fibrin monomer formation and polymerization. Shear stress modulates adhesion rates of platelets and expression of adhesion molecules and procoagulant activity on endothelial cells lining the blood vessels. That no one abnormality in any component of Virchows triad fully predicts coagulopathy a priori suggests coagulopathies are complex, multifactorial, and interactive. In this review, we focus on contributions of blood composition, vascular cells, and blood flow to hemostasis and thrombosis, and suggest that cross-talk among the 3 components of Virchows triad is necessary for hemostasis and determines propensity for thrombosis or bleeding. Investigative models that permit interplay among these components are necessary to understand the operant pathophysiology, and effectively treat and prevent thrombotic and bleeding disorders.


Journal of Thrombosis and Haemostasis | 2011

Differential Contributions of Monocyte- and Platelet-derived Microparticles towards Thrombin Generation and Fibrin Formation and Stability

Maria M. Aleman; Chris Gardiner; Paul Harrison; Alisa S. Wolberg

Summary.  Background: Microparticles (MPs) are sub‐micron vesicles shed by activated or apoptotic cells, including platelets and monocytes. Increased circulating MPs are associated with thrombosis; however, their role in thrombogenesis is poorly understood. Objective: To determine how MPs promote thrombin generation and modulate fibrin density and stability. Methods: Platelets and monocytes were isolated from healthy donors. Platelets were stimulated with calcium ionophore, thrombin receptor agonist peptide (TRAP) or TRAP/convulxin. Monocytes and human monocytic THP‐1 cells were stimulated with lipopolysaccharide (LPS). MPs were isolated, washed by high‐speed centrifugation and assessed using the following: transmission electron microscopy (TEM), Nanoparticle Tracking Analysis (NTA), flow cytometry, tissue factor (TF) activity, prothrombinase activity, thrombin generation, and clot formation, density and stability. Results: MPs from monocytes (M‐MPs) and platelets (PMPs) had similar shapes and diameters (100–300 nm). M‐MPs had TF activity (16.7 ± 2.4 pm TF per 106 MP), supported prothrombinase activity and triggered shorter thrombin generation lag times than buffer controls (5.4 ± 0.5 vs. 84.2 ± 4.8 min, respectively). Compared with controls, M‐MPs supported faster fibrin formation (0.24 ± 0.24 vs. 76.7 ± 15.1 mOD min−1, respectively), 38% higher fibrin network density and higher clot stability (3.8‐fold higher turbidity in the presence of tissue plasminogen activator). In contrast, PMPs did not have TF activity and supported 2.8‐fold lower prothrombinase activity than M‐MPs. PMPs supported contact‐dependent thrombin generation, but did not independently increase fibrin network density or stability. Interestingly, PMPs increased rates of thrombin generation and fibrin formation (1.7‐ and 1.3‐fold, respectively) when mixed with THP‐1‐derived MPs. Conclusion: MPs from platelets and monocytes differentially modulate clot formation, structure and stability, suggesting unique contributions to thrombosis.


Blood | 2012

Tumor-derived tissue factor activates coagulation and enhances thrombosis in a mouse xenograft model of human pancreatic cancer.

Jianguo Wang; Julia E. Geddings; Maria M. Aleman; Jessica C. Cardenas; Pichika Chantrathammachart; Julie C. Williams; Daniel Kirchhofer; Vladimir Y. Bogdanov; Ronald R. Bach; Janusz Rak; Frank C. Church; Alisa S. Wolberg; Rafal Pawlinski; Nigel S. Key; Jen Jen Yeh; Nigel Mackman

Cancer patients often have an activated clotting system and are at increased risk for venous thrombosis. In the present study, we analyzed tissue factor (TF) expression in 4 different human pancreatic tumor cell lines for the purpose of producing derivative tumors in vivo. We found that 2 of the lines expressed TF and released TF-positive microparticles (MPs) into the culture medium. The majority of TF protein in the culture medium was associated with MPs. Only TF-positive cell lines activated coagulation in nude mice, and this activation was abolished by an anti-human TF Ab. Of the 2 TF-positive lines, only one produced detectable levels of human MP TF activity in the plasma when grown orthotopically in nude mice. Surprisingly, < 5% of human TF protein in plasma from tumor-bearing mice was associated with MPs. Mice with TF-positive tumors and elevated levels of circulating TF-positive MPs had increased thrombosis in a saphenous vein model. In contrast, we observed no difference in thrombus weight between tumor-bearing and control mice in an inferior vena cava stenosis model. The results of the present study using a xenograft mouse model suggest that tumor TF activates coagulation, whereas TF on circulating MPs may trigger venous thrombosis.


Journal of Clinical Investigation | 2014

Factor XIII activity mediates red blood cell retention in venous thrombi

Maria M. Aleman; James R. Byrnes; Jianguo Wang; Reginald Tran; Wilbur A. Lam; Jorge Di Paola; Nigel Mackman; Jay L. Degen; Matthew J. Flick; Alisa S. Wolberg

Venous thrombi, fibrin- and rbc-rich clots triggered by inflammation and blood stasis, underlie devastating, and sometimes fatal, occlusive events. During intravascular fibrin deposition, rbc are thought to become passively trapped in thrombi and therefore have not been considered a modifiable thrombus component. In the present study, we determined that activity of the transglutaminase factor XIII (FXIII) is critical for rbc retention within clots and directly affects thrombus size. Compared with WT mice, mice carrying a homozygous mutation in the fibrinogen γ chain (Fibγ390-396A) had a striking 50% reduction in thrombus weight due to reduced rbc content. Fibrinogen from mice harboring the Fibγ390-396A mutation exhibited reduced binding to FXIII, and plasma from these mice exhibited delayed FXIII activation and fibrin crosslinking, indicating these residues mediate FXIII binding and activation. FXIII-deficient mice phenocopied mice carrying Fibγ390-396A and produced smaller thrombi with fewer rbc than WT mice. Importantly, FXIII-deficient human clots also exhibited reduced rbc retention. The addition of FXIII to FXIII-deficient clots increased rbc retention, while inhibition of FXIII activity in normal blood reduced rbc retention and produced smaller clots. These findings establish the FXIII-fibrinogen axis as a central determinant in venous thrombogenesis and identify FXIII as a potential therapeutic target for limiting venous thrombosis.


Thrombosis and Haemostasis | 2010

Flow profoundly influences fibrin network structure: Implications for fibrin formation and clot stability in haemostasis

Robert A. Campbell; Maria M. Aleman; Laura D. Gray; Michael R. Falvo; Alisa S. Wolberg

Flow profoundly influences fibrin network structure: Implications for fibrin formation and clot stability in haemostasis -


Thrombosis Research | 2014

Fibrinogen and red blood cells in venous thrombosis

Maria M. Aleman; Bethany L. Walton; James R. Byrnes; Alisa S. Wolberg

Deep vein thrombosis and pulmonary embolism, collectively termed venous thromboembolism (VTE), affect over 1 million Americans each year. VTE is triggered by inflammation and blood stasis leading to the formation of thrombi rich in fibrin and red blood cells (RBCs). However, little is known about mechanisms regulating fibrin and RBC incorporation into venous thrombi, or how these components mediate thrombus size or resolution. Both elevated circulating fibrinogen (hyperfibrinogenemia) and abnormal fibrin(ogen) structure and function, including increased fibrin network density and resistance to fibrinolysis, have been observed in plasmas from patients with VTE. Abnormalities in RBC number and/or function have also been associated with VTE risk. RBC contributions to VTE are thought to stem from their effects on blood viscosity and margination of platelets to the vessel wall. More recent studies suggest RBCs also express phosphatidylserine, support thrombin generation, and decrease fibrinolysis. RBC interactions with fibrin(ogen) and cells, including platelets and endothelial cells, may also promote thrombus formation. The contributions of fibrin(ogen) and RBCs to the pathophysiology of VTE warrants further investigation.


The Journal of Allergy and Clinical Immunology | 2015

IL-1 receptor antagonist reduces endotoxin-induced airway inflammation in healthy volunteers.

Michelle L. Hernandez; Katherine Mills; Martha Almond; Krista Todoric; Maria M. Aleman; Hongtao Zhang; Haibo Zhou; David B. Peden

BACKGROUND Asthma with neutrophil predominance is challenging to treat with corticosteroids. Novel treatment options for asthma include those that target innate immune activity. Recent literature has indicated a significant role for IL-1β in both acute and chronic neutrophilic asthma. OBJECTIVE This study used inhaled endotoxin (LPS) challenge as a model of innate immune activation to (1) assess the safety of the IL-1 receptor antagonist anakinra in conjunction with inhaled LPS and (2) to test the hypothesis that IL-1 blockade will suppress the acute neutrophil response to challenge with inhaled LPS. METHODS In a phase I clinical study 17 healthy volunteers completed a double-blind, placebo-controlled crossover study in which they received 2 daily subcutaneous doses of 1 mg/kg anakinra (maximum dose, 100 mg) or saline (placebo). One hour after the second treatment dose, subjects underwent an inhaled LPS challenge. Induced sputum was assessed for neutrophils 4 hours after inhaled LPS. The effect of anakinra compared with placebo on airway neutrophil counts and airway proinflammatory cytokine levels after LPS challenge was compared by using a linear mixed-model approach. RESULTS Anakinra pretreatment significantly diminished airway neutrophilia compared with placebo. LPS-induced IL-1β, IL-6, and IL-8 levels were significantly reduced during the anakinra treatment period compared with those seen after placebo. Subjects tolerated the anakinra treatment well without an increased frequency of infections attributable to anakinra treatment. CONCLUSIONS Anakinra effectively reduced airway neutrophilic inflammation and resulted in no serious adverse events in a model of inhaled LPS challenge. Anakinra is a potential therapeutic candidate for treatment of asthma with neutrophil predominance in diseased populations.


EBioMedicine | 2016

Complement Activation in Arterial and Venous Thrombosis is Mediated by Plasmin

Jonathan H. Foley; Bethany L. Walton; Maria M. Aleman; Alice M. O'Byrne; Victor Lei; Micaela Harrasser; Kimberley Foley; Alisa S. Wolberg; Edward M. Conway

Thrombus formation leading to vaso-occlusive events is a major cause of death, and involves complex interactions between coagulation, fibrinolytic and innate immune systems. Leukocyte recruitment is a key step, mediated partly by chemotactic complement activation factors C3a and C5a. However, mechanisms mediating C3a/C5a generation during thrombosis have not been studied. In a murine venous thrombosis model, levels of thrombin–antithrombin complexes poorly correlated with C3a and C5a, excluding a central role for thrombin in C3a/C5a production. However, clot weight strongly correlated with C5a, suggesting processes triggered during thrombosis promote C5a generation. Since thrombosis elicits fibrinolysis, we hypothesized that plasmin activates C5 during thrombosis. In vitro, the catalytic efficiency of plasmin-mediated C5a generation greatly exceeded that of thrombin or factor Xa, but was similar to the recognized complement C5 convertases. Plasmin-activated C5 yielded a functional membrane attack complex (MAC). In an arterial thrombosis model, plasminogen activator administration increased C5a levels. Overall, these findings suggest plasmin bridges thrombosis and the immune response by liberating C5a and inducing MAC assembly. These new insights may lead to the development of strategies to limit thrombus formation and/or enhance resolution.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2013

Elevated Prothrombin Promotes Venous, but Not Arterial, Thrombosis in Mice

Maria M. Aleman; Bethany L. Walton; James R. Byrnes; Jianguo Wang; Matthew J. Heisler; Kellie R. Machlus; Brian C. Cooley; Alisa S. Wolberg

Objective—Individuals with elevated prothrombin, including those with the prothrombin G20210A mutation, have increased risk of venous thrombosis. Although these individuals do not have increased circulating prothrombotic biomarkers, their plasma demonstrates increased tissue factor–dependent thrombin generation in vitro. The objectives of this study were to determine the pathological role of elevated prothrombin in venous and arterial thrombosis in vivo, and distinguish thrombogenic mechanisms in these vessels. Approach and Results—Prothrombin was infused into mice to raise circulating levels. Venous thrombosis was induced by electrolytic stimulus to the femoral vein or inferior vena cava ligation. Arterial thrombosis was induced by electrolytic stimulus or ferric chloride application to the carotid artery. Mice infused with prothrombin demonstrated increased tissue factor–triggered thrombin generation measured ex vivo, but did not have increased circulating prothrombotic biomarkers in the absence of vessel injury. After venous injury, elevated prothrombin increased thrombin generation and the fibrin accumulation rate and total amount of fibrin ≈3-fold, producing extended thrombi with increased mass. However, elevated prothrombin did not accelerate platelet accumulation, increase the fibrin accumulation rate, or shorten the vessel occlusion time after arterial injury. Conclusions—These findings reconcile previously discordant findings on thrombin generation in hyperprothrombinemic individuals measured ex vivo and in vitro, and show elevated prothrombin promotes venous, but not arterial, thrombosis in vivo.


Journal of Thrombosis and Haemostasis | 2014

Strengths and weaknesses of a new mouse model of thrombosis induced by inferior vena cava stenosis: communication from the SSC of the ISTH

Julia E. Geddings; Maria M. Aleman; Alisa S. Wolberg; M-L von Brühl; Steffen Massberg; Nigel Mackman

Deep vein thrombosis (DVT) is a significant cause of morbidity and mortality throughout the world 1. Risk factors for DVT include major surgery, immobilization, trauma, and cancer, among others 1. Many mouse models of thrombosis have been used to study mechanisms of thrombosis, including those involving small and large veins. In this report from the Animal Models Subcommittee of the ISTH, we summarize the strengths and weaknesses of the inferior vena cava (IVC) stenosis mouse model of venous thrombosis.

Collaboration


Dive into the Maria M. Aleman's collaboration.

Top Co-Authors

Avatar

Alisa S. Wolberg

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

David B. Peden

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Michelle L. Hernandez

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jianguo Wang

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Haibo Zhou

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Katherine H. Mills

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Nigel Mackman

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Bethany L. Walton

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Frank C. Church

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Hongtao Zhang

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge