1
|
Xu W, Tan X, Li ML, Xu H, Villegas J, Fu H. Von Willebrand factor and hematogenous cancer metastasis under flow. Front Cell Dev Biol 2024; 12:1435718. [PMID: 39282473 PMCID: PMC11401050 DOI: 10.3389/fcell.2024.1435718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/05/2024] [Indexed: 09/19/2024] Open
Abstract
Hematogenous metastasis involves cancer cell migration to different locations from the primary tumor through the blood circulation. Von Willebrand factor (VWF) has been shown to play an important role in tumor cell adhesion to and extravasation from the endothelial cell lining of blood vessel walls during cancer metastasis. VWF may contribute to this process by interacting with tumor cells, endothelial cells, and platelets through various cell membrane receptors, such as platelet glycoprotein (GP)Ibα, P-selectin, ανβ3 and αIIbβ3 integrins, and glycocalyx. Blood flow can mechanically extend and activate VWF to bind platelets and associate intermolecularly with other VWF molecules in plasma or on the surface of endothelial cells, cancer cells, or platelets. This suggests a mechanoregulatory role of VWF in mediating the interactions between VWF and these cells to promote cancer cell adhesion to blood vessels. In this review, we will summarize the current knowledge of VWF function and the role of hydrodynamic forces in hematogenous cancer metastasis.
Collapse
Affiliation(s)
- Wenxuan Xu
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Xi Tan
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Morgan L Li
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Hanzhi Xu
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Jasmine Villegas
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Hongxia Fu
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
- Department of Bioengineering, University of Washington, Seattle, WA, United States
- Bloodworks Research Institute, Seattle, WA, United States
| |
Collapse
|
2
|
Regling K, Sidonio RF. Factor VIII stimulants and other novel therapies for the treatment of von Willebrand disease: what's new on the horizon? Expert Opin Pharmacother 2024; 25:1427-1438. [PMID: 39155445 DOI: 10.1080/14656566.2024.2391526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
INTRODUCTION Von Willebrand disease (VWD) is the most common inherited bleeding disorder, affecting about 0.6% to 1.3% of the population, and is characterized primarily by mucocutaneous bleeding secondary to defective platelet adhesion and aggregation. Current therapeutic options for those with severe disease are limited and require frequent intravenous infusions. AREAS COVERED This review discusses the current and recently completed clinical trials involving pathways to FVIII augmentation for the treatment of VWD. Clinical trials registered on clinicaltrials.gov and published data via PubMed searches through June 2024 were included. EXPERT OPINION Available treatment options to those with VWD are limited in part due to limited clinical trials, the complexity of VWD types, and the pharmacokinetics of current treatment options. The development of therapeutic options that reduce treatment burden is necessary to improve quality of life and reduce bleeding complications and in recent years there has been an increased interest from industry to apply novel therapeutics for VWD. The FVIII mimetic, emicizumab, has demonstrated early success in patients with severe VWD and is a promising treatment option for those who require prophylaxis. Furthermore, products like efanesoctocog alfa (Altuviiio®) and BT200 have achieved enhanced VWF/FVIII half-life extension could expand the current treatment landscape while concurrently minimizing treatment burden.
Collapse
Affiliation(s)
- Katherine Regling
- Pediatric Hematology Oncology, Children's Hospital of Michigan, Detroit, MI, USA
- Department of Pediatrics, Central Michigan University College of Medicine, Mount Pleasant, MI, USA
| | - Robert F Sidonio
- Pediatric Hematology Oncology, Emory University and Aflac Cancer and Blood Disorders, Atlanta, GA, USA
| |
Collapse
|
3
|
Byrnes JR, Lee T, Sharaby S, Campbell RA, Dobson DA, Holle LA, Luo M, Kangro K, Homeister JW, Aleman MM, Luyendyk JP, Kerlin BA, Dumond JB, Wolberg AS. Reciprocal stabilization of coagulation factor XIII-A and -B subunits is a determinant of plasma FXIII concentration. Blood 2024; 143:444-455. [PMID: 37883802 PMCID: PMC10862369 DOI: 10.1182/blood.2023022042] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/28/2023] [Accepted: 10/15/2023] [Indexed: 10/28/2023] Open
Abstract
ABSTRACT Transglutaminase factor XIII (FXIII) is essential for hemostasis, wound healing, and pregnancy maintenance. Plasma FXIII is composed of A and B subunit dimers synthesized in cells of hematopoietic origin and hepatocytes, respectively. The subunits associate tightly in circulation as FXIII-A2B2. FXIII-B2 stabilizes the (pro)active site-containing FXIII-A subunits. Interestingly, people with genetic FXIII-A deficiency have decreased FXIII-B2, and therapeutic infusion of recombinant FXIII-A2 (rFXIII-A2) increases FXIII-B2, suggesting FXIII-A regulates FXIII-B secretion, production, and/or clearance. We analyzed humans and mice with genetic FXIII-A deficiency and developed a mouse model of rFXIII-A2 infusion to define mechanisms mediating plasma FXIII-B levels. Like humans with FXIII-A deficiency, mice with genetic FXIII-A deficiency had reduced circulating FXIII-B2, and infusion of FXIII-A2 increased FXIII-B2. FXIII-A-deficient mice had normal hepatic function and did not store FXIII-B in liver, indicating FXIII-A does not mediate FXIII-B secretion. Transcriptional analysis and polysome profiling indicated similar F13b levels and ribosome occupancy in FXIII-A-sufficient and -deficient mice and in FXIII-A-deficient mice infused with rFXIII-A2, indicating FXIII-A does not induce de novo FXIII-B synthesis. Unexpectedly, pharmacokinetic/pharmacodynamic modeling of FXIII-B antigen after rFXIII-A2 infusion in humans and mice suggested FXIII-A2 slows FXIII-B2 loss from plasma. Accordingly, comparison of free FXIII-B2 vs FXIII-A2-complexed FXIII-B2 (FXIII-A2B2) infused into mice revealed faster clearance of free FXIII-B2. These data show FXIII-A2 prevents FXIII-B2 loss from circulation and establish the mechanism underlying FXIII-B2 behavior in FXIII-A deficiency and during rFXIII-A2 therapy. Our findings reveal a unique, reciprocal relationship between independently synthesized subunits that mediate an essential hemostatic protein in circulation. This trial was registered at www.ClinicalTrials.com as #NCT00978380.
Collapse
Affiliation(s)
- James R. Byrnes
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Taek Lee
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Sherif Sharaby
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Robert A. Campbell
- Molecular Medicine Program, Department of Internal Medicine, The University of Utah, Salt Lake City, UT
| | - Dre’Von A. Dobson
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Lori A. Holle
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Michelle Luo
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Kadri Kangro
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jonathon W. Homeister
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Maria M. Aleman
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - James P. Luyendyk
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI
| | - Bryce A. Kerlin
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
- Division of Pediatric Hematology/Oncology/Blood & Marrow Transplantation, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Julie B. Dumond
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Alisa S. Wolberg
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
4
|
Nguyen GN, Lindgren JR, Seleme MC, Kafle S, Zander CB, Zheng XL, Sabatino DE. Altered cleavage of human factor VIII at the B-domain and acidic region 3 interface enhances expression after gene therapy in hemophilia A mice. J Thromb Haemost 2023; 21:2101-2113. [PMID: 37080538 PMCID: PMC11157168 DOI: 10.1016/j.jtha.2023.04.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/17/2023] [Accepted: 04/04/2023] [Indexed: 04/22/2023]
Abstract
BACKGROUND Variants of human factor VIII (hFVIII) have been developed to further understand the structure and function of hFVIII and improve gene-based therapeutics. We have previously characterized several hFVIII variants of the furin cleavage site (1645-1648) with improved secretion. We have also identified a second cleavage site in the acidic region 3 (a3) (1657-1658) that becomes the primary hFVIII intracellular cleavage position in the absence of the furin site. We tested a hypothesis that modification of this site may confer additional functional advantages to hFVIII. OBJECTIVES The aim of this study was to conduct the biochemical and functional characterization of hFVIII variants of the furin cleavage site, the a3 cleavage site, or in combination, both in vitro and in vivo after AAV mediated gene therapy. METHODS Recombinant hFVIII variants of the furin cleavage site (hFVIII-Δ3), the a3 cleavage site (hFVIII-S1657P/D1658E [SP/DE]), or in combination (hFVIII-Δ3-SP/DE) were purified and characterized in vitro and in vivo. RESULTS Recombinant hFVIII-Δ3, hFVIII-SP/DE, and hFVIII-Δ3-SP/DE variants all had comparable specific activity to B-domain deleted (BDD) hFVIII. Hemophilia A mice tolerant to hFVIII did not develop immune responses to hFVIII after protein challenge with these variants or after adeno-associated virus (AAV) delivery. Following AAV delivery, hFVIII-Δ3-SP/DE resulted in expression levels that were 2- to 5-fold higher than those with hFVIII-BDD in hemophilia A mice. CONCLUSION The novel hFVIII-Δ3-SP/DE variant of the furin and a3 cleavage sites significantly improved secretion compared with hFVIII-BDD. This key feature of the Δ3-SP/DE variant provides a unique strategy that can be combined with other approaches to further improve factor VIII expression to achieve superior efficacy in AAV-based gene therapy for hemophilia A.
Collapse
Affiliation(s)
- Giang N Nguyen
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jonathan R Lindgren
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Maria C Seleme
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Samita Kafle
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Catherine B Zander
- Department of Pathology, University of Alabama at Birmingham School of Medicine, University of Alabama, Birmingham, Alabama, USA
| | - X Long Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA; Institute of Reproductive Medicine and Developmental Science, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Denise E Sabatino
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Pediatrics, Division of Hematology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
5
|
Du P, Bergamasco A, Moride Y, Truong Berthoz F, Özen G, Tzivelekis S. Von Willebrand Disease Epidemiology, Burden of Illness and Management: A Systematic Review. J Blood Med 2023; 14:189-208. [PMID: 36891166 PMCID: PMC9987238 DOI: 10.2147/jbm.s389241] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/06/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Although hereditary von Willebrand disease (VWD) is the most common bleeding disorder, its epidemiology is not well understood. A systematic review (PROSPERO CRD42020197674/CRD42021244374) on the epidemiology/burden of illness of VWD was conducted to better understand patients' unmet needs. Methods Observational studies (published January 1, 2010 to April 14, 2021) were identified in MEDLINE and Embase databases, using free-text keywords and thesaurus terms for VWD and outcomes of interest. Pragmatic web-based searches of the gray literature, including conference abstracts, were performed, and reference lists of retained publications were manually searched for additional sources. Case reports and clinical trials (phase 1-3) were excluded. Outcomes of interest were incidence, prevalence, mortality, patient characteristics, burden of illness, and therapeutic management/treatments currently used for VWD. Results Of the 3095 identified sources, 168 were included in this systematic review. Reported VWD prevalence (22 sources) ranged from 108.9 to 2200 per 100,000 in population-based studies and from 0.3 to 16.5 per 100,000 in referral-based studies. Reported times between first symptom onset and diagnosis (two sources; mean 669 days; median 3 years) highlighted gaps in timely VWD diagnosis. Bleeding events reported in 72-94% of the patients with VWD (all types; 27 sources) were mostly mucocutaneous including epistaxis, menorrhagia, and oral/gum bleeding. Poorer health-related quality of life (three sources) and greater health care resource utilization (three sources) were reported for patients with VWD than in general populations. Conclusion Available data suggest that patients with VWD experience high disease burden in terms of bleeding, poor quality of life, and health care resource utilization.
Collapse
Affiliation(s)
- Ping Du
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | | | - Yola Moride
- YOLARX Consultants, Paris, France
- Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | | | - Gülden Özen
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | | |
Collapse
|
6
|
Childers KC, Peters SC, Spiegel PC. Structural insights into blood coagulation factor VIII: Procoagulant complexes, membrane binding, and antibody inhibition. J Thromb Haemost 2022; 20:1957-1970. [PMID: 35722946 DOI: 10.1111/jth.15793] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/10/2022] [Accepted: 06/16/2022] [Indexed: 11/28/2022]
Abstract
Advances in structural studies of blood coagulation factor VIII (FVIII) have provided unique insight into FVIII biochemistry. Atomic detail models of the B domain-deleted FVIII structure alone and in complex with its circulatory partner, von Willebrand factor (VWF), provide a structure-based rationale for hemophilia A-associated mutations which impair FVIII stability and increase FVIII clearance rates. In this review, we discuss the findings from these studies and their implications toward the design of a recombinant FVIII with improved circulatory half-life. Additionally, we highlight recent structural studies of FVIII bound to inhibitory antibodies that have refined our understanding of FVIII binding to activated platelet membranes and formation of the intrinsic tenase complex. The combination of bioengineering and structural efforts to understand FVIII biochemistry will improve therapeutics for treating hemophilia A, either through FVIII replacement therapeutics, immune tolerance induction, or gene therapy approaches.
Collapse
Affiliation(s)
- Kenneth C Childers
- Chemistry Department, Western Washington University, Bellingham, Washington, USA
| | - Shaun C Peters
- Chemistry Department, Western Washington University, Bellingham, Washington, USA
| | - Paul Clint Spiegel
- Chemistry Department, Western Washington University, Bellingham, Washington, USA
| |
Collapse
|
7
|
Eriksson ANM, Rigaud C, Rokka A, Skaugen M, Lihavainen JH, Vehniäinen ER. Changes in cardiac proteome and metabolome following exposure to the PAHs retene and fluoranthene and their mixture in developing rainbow trout alevins. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 830:154846. [PMID: 35351515 DOI: 10.1016/j.scitotenv.2022.154846] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/11/2022] [Accepted: 03/23/2022] [Indexed: 06/14/2023]
Abstract
Exposure to polycyclic aromatic hydrocarbons (PAHs) is known to affect developing organisms. Utilization of different omics-based technologies and approaches could therefore provide a base for the discovery of novel mechanisms of PAH induced development of toxicity. To this aim, we investigated how exposure towards two PAHs with different toxicity mechanisms: retene (an aryl hydrocarbon receptor 2 (Ahr2) agonist), and fluoranthene (a weak Ahr2 agonist and cytochrome P450 inhibitor (Cyp1a)), either alone or as a mixture, affected the cardiac proteome and metabolome in newly hatched rainbow trout alevins (Oncorhynchus mykiss). In total, we identified 65 and 82 differently expressed proteins (DEPs) across all treatments compared to control (DMSO) after 7 and 14 days of exposure. Exposure to fluoranthene altered the expression of 11 and 19 proteins, retene 29 and 23, while the mixture affected 44 and 82 DEPs by Days 7 and 14, respectively. In contrast, only 5 significantly affected metabolites were identified. Pathway over-representation analysis identified exposure-specific activation of phase II metabolic processes, which were accompanied with exposure-specific body burden profiles. The proteomic data highlights that exposure to the mixture increased oxidative stress, altered iron metabolism and impaired coagulation capacity. Additionally, depletion of several mini-chromosome maintenance components, in combination with depletion of several intermediate filaments and microtubules, among alevins exposed to the mixture, suggests compromised cellular integrity and reduced rate of mitosis, whereby affecting heart growth and development. Furthermore, the combination of proteomic and metabolomic data indicates altered energy metabolism, as per amino acid catabolism among mixture exposed alevins; plausibly compensatory mechanisms as to counteract reduced absorption and consumption of yolk. When considered as a whole, proteomic and metabolomic data, in relation to apical effects on the whole organism, provides additional insight into PAH toxicity and the effects of exposure on heart structure and molecular processes.
Collapse
Affiliation(s)
- Andreas N M Eriksson
- Department of Biological and Environmental Science, University of Jyväskylä, P.O. Box 35, FI-40014, Finland.
| | - Cyril Rigaud
- Department of Biological and Environmental Science, University of Jyväskylä, P.O. Box 35, FI-40014, Finland.
| | - Anne Rokka
- Turku Proteomics Facility, Turku University, Tykistökatu 6, 20520 Turku, Finland.
| | - Morten Skaugen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Campus Ås, Universitetstunet 3, 1430 Ås, Norway.
| | - Jenna H Lihavainen
- Umeå Plant Science Centre, Umeå University, KB. K3 (Fys. Bot.), Artedigränd 7, Fysiologisk botanik, UPSC, KB. K3 (B3.44.45) Umeå universitet, 901 87 Umeå, Sweden.
| | - Eeva-Riikka Vehniäinen
- Department of Biological and Environmental Science, University of Jyväskylä, P.O. Box 35, FI-40014, Finland.
| |
Collapse
|
8
|
Demers M, Aleman MM, Kistanova E, Peters R, Salas J, Seth Chhabra E. Efanesoctocog alfa elicits functional clot formation that is indistinguishable to that of recombinant factor VIII. J Thromb Haemost 2022; 20:1674-1683. [PMID: 35466511 PMCID: PMC9320793 DOI: 10.1111/jth.15741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/12/2022] [Accepted: 04/21/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Factor VIII (FVIII) binding to endogenous von Willebrand factor (VWF) has constrained half-life extension of recombinant FVIII (rFVIII) products for hemophilia A. Efanesoctocog alfa (rFVIIIFc-VWF-XTEN; BIVV001) is a novel fusion protein designed to decouple FVIII from VWF in circulation and maximize half-life prolongation by XTEN® polypeptides and Fc fusion. FVIII, VWF, and platelets interact to achieve normal hemostasis. Thus, bioengineered FVIII replacement products, such as efanesoctocog alfa, require comprehensive assessment of their hemostatic potential. OBJECTIVES We compared functional clot formation and injury-induced platelet accumulation between efanesoctocog alfa and rFVIII. PATIENTS/METHODS The hemostatic potential of efanesoctocog alfa and rFVIII were assessed by measuring their dose-dependent effects on in vitro fibrin generation in hemophilic plasma and in vivo injury-induced platelet accumulation using intravital microscopy and repeat saphenous vein laser-induced injuries in hemophilia A mice. RESULTS Equal concentrations of efanesoctocog alfa or rFVIII (up to 1 IU/ml) added to plasma from patients with hemophilia A elicited similar kinetics for dose-dependent fibrin polymerization between factor products. In the presence of tissue plasminogen activator (tPA), clots formed had similar stability between products. Single intravenous doses (50, 100, or 150 IU/kg) of efanesoctocog alfa or rFVIII shortly before repeat saphenous vein laser-induced injuries increased platelet accumulation over time in a dose-dependent manner in hemophilia A mice. Platelet deposition kinetics were similar between products. CONCLUSIONS Equivalent doses of efanesoctocog alfa and rFVIII had similar efficacy in promoting fibrin clot formation and injury-induced platelet accumulation. The hemostatic potential of efanesoctocog alfa was indistinguishable from that of rFVIII.
Collapse
|
9
|
Abstract
INTRODUCTION Hemophilia A (HA) or B (HB) is an X-linked recessive disorder caused by a defect in the factor VIII (FVIII) or factor IX (FIX) gene which leads to the dysfunction of blood coagulation. Protein replacement therapy (PRT) uses recombinant proteins and plasma-derived products, which incurs high cost and inconvenience requiring routine intravenous infusions and life-time treatment. Understanding of detailed molecular mechanisms on FVIII gene function could provide innovative solutions to amend this disorder. In recent decades, gene therapeutics have advanced rapidly and a one-time cure solution has been proposed. AREAS COVERED This review summarizes current understanding of molecular pathways involved in blood coagulation, with emphasis on FVIII's functional role. The existing knowledge and challenges on FVIII gene expression, from transcription, translation, post-translational modification including glycosylation to protein processing and secretion, and co-factor interactions are deciphered and potential molecular interventions discussed. EXPERT OPINION This article reviews the potential treatment targets for HA and HB, including antibodies, small molecules and gene therapeutics, based on molecular mechanisms of FVIII biosynthesis, and further, assessing the pros and cons of these various treatment strategies. Understanding detailed FVIII protein synthesis and secretory pathways could provide exciting opportunities in identifying novel therapeutics to ameliorate hemophilia state.
Collapse
Affiliation(s)
- Jie Gong
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China
| | - Hao-Lin Wang
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China
| | - Lung-Ji Chang
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China.,Geno-Immune Medical Institute, Shenzhen, China
| |
Collapse
|
10
|
Gebetsberger J, Schirmer M, Wurzer WJ, Streif W. Low Bone Mineral Density in Hemophiliacs. Front Med (Lausanne) 2022; 9:794456. [PMID: 35186990 PMCID: PMC8849249 DOI: 10.3389/fmed.2022.794456] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022] Open
Abstract
Objective To review the current knowledge on bone health in patients with hemophilia A and the underlying pathogenetic mechanisms. Data Sources Original research articles, meta-analyses, and scientific reviews. Data Synthesis Already in childhood, patients with hemophilia A are prone to low bone mineral density, leading to osteopenia and/or osteoporosis. Initially associated with the life style of hemophilia, today we are faced with accumulating evidence that coagulation factor VIII is involved directly or indirectly in bone physiology. Conclusion Understanding the role of factor VIII and the mechanisms of decreased bone mineral density in hemophilia A is critically important, especially as non-factor replacement therapies are available, and treatment decisions potentially impact bone health.
Collapse
Affiliation(s)
| | - Michael Schirmer
- Department of Internal Medicine, Clinic II, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Werner Streif
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
11
|
Gong J, Chung TH, Zheng J, Zheng H, Chang LJ. Transduction of modified factor VIII gene improves lentiviral gene therapy efficacy for hemophilia A. J Biol Chem 2021; 297:101397. [PMID: 34774524 PMCID: PMC8649223 DOI: 10.1016/j.jbc.2021.101397] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/18/2022] Open
Abstract
Hemophilia A (HA) is a bleeding disorder caused by deficiency of the coagulation factor VIII (F8). F8 replacement is standard of care, whereas gene therapy (F8 gene) for HA is an attractive investigational approach. However, the large size of the F8 gene and the immunogenicity of the product present challenges in development of the F8 gene therapy. To resolve these problems, we synthesized a shortened F8 gene (F8-BDD) and cloned it into a lentiviral vector (LV). The F8-BDD produced mainly short cleaved inactive products in LV-transduced cells. To improve F8 functionality, we designed two novel F8-BDD genes, one with an insertion of eight specific N-glycosylation sites (F8-N8) and another which restored all N-glycosylation sites (F8-299) in the B domain. Although the overall protein expression was reduced, high coagulation activity (>100-fold) was detected in the supernatants of LV-F8-N8- and LV-F8-299-transduced cells. Protein analysis of F8 and the procoagulation cofactor, von Willebrand Factor, showed enhanced interaction after restoration of B domain glycosylation using F8-299. HA mouse hematopoietic stem cell transplantation studies illustrated that the bleeding phenotype was corrected after LV-F8-N8 or -299 gene transfer into the hematopoietic stem cells. Importantly, the F8-299 modification markedly reduced immunogenicity of the F8 protein in these HA mice. In conclusion, the modified F8-299 gene could be efficiently packaged into LV and, although with reduced expression, produced highly stable and functional F8 protein that corrected the bleeding phenotype without inhibitory immunogenicity. We anticipate that these results will be beneficial in the development of gene therapies against HA.
Collapse
Affiliation(s)
- Jie Gong
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China
| | - Tsai-Hua Chung
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China; Shenzhen Geno-Immune Medical Institute, Shenzhen, China
| | - Jie Zheng
- Hematology Center, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Huyong Zheng
- Hematology Center, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Lung-Ji Chang
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China; Shenzhen Geno-Immune Medical Institute, Shenzhen, China; Hematology Center, Beijing Children's Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
12
|
Cooper S, Wilmarth PA, Cunliffe JM, Klimek J, Pang J, Tassi Yunga S, Minnier J, Reddy A, David L, Aslan JE. Platelet proteome dynamics in hibernating 13-lined ground squirrels. Physiol Genomics 2021; 53:473-485. [PMID: 34677084 PMCID: PMC8616595 DOI: 10.1152/physiolgenomics.00078.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/21/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022] Open
Abstract
Hibernating mammals undergo a dramatic drop in temperature and blood flow during torpor, yet avoid stasis blood clotting through mechanisms that remain unspecified. The effects of hibernation on hemostasis are especially complex, as cold temperatures generally activate platelets, resulting in platelet clearance and cold storage lesions in the context of blood transfusion. With a hibernating body temperature of 4°C-8°C, 13-lined ground squirrels (Ictidomys tridecemlineatus) provide a model to study hemostasis as well as platelet cold storage lesion resistance during hibernation. Here, we quantified and systematically compared proteomes of platelets collected from ground squirrels at summer (active), fall (entrance), and winter (topor) to elucidate how molecular-level changes in platelets may support hemostatic adaptations in torpor. Platelets were isolated from a total of 11 squirrels in June, October, and January. Platelet lysates from each animal were digested with trypsin prior to 11-plex tandem mass tag (TMT) labeling, followed by LC-MS/MS analysis for relative protein quantification. We measured >700 proteins with significant variations in abundance in platelets over the course of entrance, torpor, and activity-including systems of proteins regulating translation, secretion, metabolism, complement, and coagulation cascades. We also noted species-specific differences in levels of hemostatic, secretory, and inflammatory regulators in ground squirrel platelets relative to human platelets. Altogether, we provide the first ever proteomic characterization of platelets from hibernating animals, where systematic changes in metabolic, hemostatic, and other proteins may account for physiological adaptations in torpor and also inform translational effort to improve cold storage of human platelets for transfusion.
Collapse
Affiliation(s)
- Scott Cooper
- Biology Department, University of Wisconsin-La Crosse, La Crosse, Wisconsin
| | - Phillip A Wilmarth
- Proteomics Shared Resource, Oregon Health & Science University, Portland, Oregon
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Jennifer M Cunliffe
- Proteomics Shared Resource, Oregon Health & Science University, Portland, Oregon
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - John Klimek
- Proteomics Shared Resource, Oregon Health & Science University, Portland, Oregon
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Jiaqing Pang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Samuel Tassi Yunga
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, Oregon
| | - Jessica Minnier
- Division of Cardiology, Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Ashok Reddy
- Proteomics Shared Resource, Oregon Health & Science University, Portland, Oregon
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Larry David
- Proteomics Shared Resource, Oregon Health & Science University, Portland, Oregon
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Joseph E Aslan
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon
- Division of Cardiology, Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
13
|
Thomazini CM, Sachetto ATA, de Albuquerque CZ, de Moura Mattaraia VG, de Oliveira AK, Serrano SMDT, Lebrun I, Barbaro KC, Santoro ML. Involvement of von Willebrand factor and botrocetin in the thrombocytopenia induced by Bothrops jararaca snake venom. PLoS Negl Trop Dis 2021; 15:e0009715. [PMID: 34478462 PMCID: PMC8445451 DOI: 10.1371/journal.pntd.0009715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/16/2021] [Accepted: 08/06/2021] [Indexed: 12/20/2022] Open
Abstract
Patients bitten by snakes consistently manifest a bleeding tendency, in which thrombocytopenia, consumption coagulopathy, mucous bleeding, and, more rarely, thrombotic microangiopathy, are observed. Von Willebrand factor (VWF) is required for primary hemostasis, and some venom proteins, such as botrocetin (a C-type lectin-like protein) and snake venom metalloproteinases (SVMP), disturb the normal interaction between platelets and VWF, possibly contributing to snakebite-induced bleedings. To understand the relationship among plasma VWF, platelets, botrocetin and SVMP from Bothrops jararaca snake venom (BjV) in the development of thrombocytopenia, we used (a) Wistar rats injected s.c. with BjV preincubated with anti-botrocetin antibodies (ABA) and/or Na2-EDTA (a SVMP inhibitor), and (b) VWF knockout mice (Vwf-/-) injected with BjV. Under all conditions, BjV induced a rapid and intense thrombocytopenia. In rats, BjV alone reduced the levels of VWF:Ag, VWF:CB, high molecular weight multimers of VWF, ADAMTS13 activity, and factor VIII. Moreover, VWF:Ag levels in rats that received BjV preincubated with Na2-EDTA and/or ABA tended to recover faster. In mice, BjV caused thrombocytopenia in both Vwf-/- and C57BL/6 (background control) strains, and VWF:Ag levels tended to decrease in C57BL/6, demonstrating that thrombocytopenia was independent of the presence of plasma VWF. These findings showed that botrocetin present in BjV failed to affect the extent or the time course of thrombocytopenia induced by envenomation, but it contributed to decrease the levels and function of plasma VWF. Thus, VWF alterations during B. jararaca envenomation are an ancillary event, and not the main mechanism leading to decreased platelet counts.
Collapse
Affiliation(s)
- Camila Martos Thomazini
- Laboratório de Fisiopatologia, Instituto Butantan, São Paulo-SP, Brazil
- Programa de Pós-Graduação em Clínica Médica, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo-SP, Brazil
| | - Ana Teresa Azevedo Sachetto
- Laboratório de Fisiopatologia, Instituto Butantan, São Paulo-SP, Brazil
- Programa de Pós-Graduação em Clínica Médica, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo-SP, Brazil
| | | | | | - Ana Karina de Oliveira
- Laboratório de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo-SP, Brazil
| | - Solange Maria de Toledo Serrano
- Laboratório de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo-SP, Brazil
| | - Ivo Lebrun
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo-SP, Brazil
| | | | - Marcelo Larami Santoro
- Laboratório de Fisiopatologia, Instituto Butantan, São Paulo-SP, Brazil
- Programa de Pós-Graduação em Clínica Médica, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo-SP, Brazil
- * E-mail: ,
| |
Collapse
|
14
|
Baronciani L, Peake I, Schneppenheim R, Goodeve A, Ahmadinejad M, Badiee Z, Baghaipour MR, Benitez O, Bodó I, Budde U, Cairo A, Castaman G, Eshghi P, Goudemand J, Hassenpflug W, Hoorfar H, Karimi M, Keikhaei B, Lassila R, Leebeek FWG, Lopez Fernandez MF, Mannucci PM, Marino R, Nikšić N, Oyen F, Santoro C, Tiede A, Toogeh G, Tosetto A, Trossaert M, Zetterberg EMK, Eikenboom J, Federici AB, Peyvandi F. Genotypes of European and Iranian patients with type 3 von Willebrand disease enrolled in 3WINTERS-IPS. Blood Adv 2021; 5:2987-3001. [PMID: 34351388 PMCID: PMC8361454 DOI: 10.1182/bloodadvances.2020003397] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Type 3 von Willebrand disease (VWD3) is a rare and severe bleeding disorder characterized by often undetectable von Willebrand factor (VWF) plasma levels, a recessive inheritance pattern, and heterogeneous genotype. The objective of this study was to identify the VWF defects in 265 European and Iranian patients with VWD3 enrolled in 3WINTERS-IPS (Type 3 Von Willebrand International Registries Inhibitor Prospective Study). All analyses were performed in centralized laboratories. The VWF genotype was studied in 231 patients with available DNA (121 [115 families] from Europe [EU], and 110 [91 families] from Iran [IR]). Among 206 unrelated patients, 134 were homozygous (EU/IR = 57/77) and 50 were compound heterozygous (EU/IR = 43/7) for VWF variants. In 22 patients, no or only one variant was found. A total of 154 different VWF variants (EU/IR = 101/58 [5 shared]) were identified among the 379 affected alleles (EU/IR = 210/169), of which 48 (EU/IR = 18/30) were novel. The variants p.Arg1659*, p.Arg1853*, p.Arg2535*, p.Cys275Ser, and delEx1_Ex5 were found in both European and Iranian VWD3 patients. Sixty variants were identified only in a single allele (EU/IR = 50/10), whereas 18 were recurrent (≥3 patients) within 144 affected alleles. Nine large deletions and one large insertion were found. Although most variants predicted null alleles, 21% of patients carried at least 1 missense variant. VWD3 genotype was more heterogeneous in the European population than in the Iranian population, with nearly twice as many different variants. A higher number of novel variants were found in the Iranian VWD3 patients.
Collapse
Affiliation(s)
- Luciano Baronciani
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center and Fondazione Luigi Villa, Milan, Italy
| | - Ian Peake
- Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom
| | - Reinhard Schneppenheim
- Department of Pediatric Hematology and Oncology, University Medical Centre, Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Goodeve
- Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom
| | - Minoo Ahmadinejad
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
- Pediatric Congenital Hematologic Disorders Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Zahra Badiee
- Hemophilia-Thalassemia Center, Mashhad University of Medical Science, Mashad, Islamic Republic of Iran
| | | | - Olga Benitez
- Hemophilia Unit, University Vall d'Hebron Hospital, Barcelona, Spain
| | - Imre Bodó
- Department of Internal Medicine and Hematology-Semmelweis University, Budapest, Hungary
| | - Ulrich Budde
- Hemostaseology Medilys Laborgesellschaft mbH, Hamburg, Germany
| | - Andrea Cairo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center and Fondazione Luigi Villa, Milan, Italy
| | - Giancarlo Castaman
- Center for Bleeding Disorders and Coagulation, Careggi University Hospital, Florence, Italy
| | - Peyman Eshghi
- Pediatric Congenital Hematologic Disorders Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Jenny Goudemand
- Univ. Lille, CHU Lille, Haematology and Transfusion, Lille, France
| | - Wolf Hassenpflug
- Department of Pediatric Hematology and Oncology, University Medical Centre, Hamburg-Eppendorf, Hamburg, Germany
| | - Hamid Hoorfar
- Hemophilia Center, Esfahan University of Medical Science, Esfahan, Islamic Republic of Iran
| | - Mehran Karimi
- Hematology Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Bijan Keikhaei
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Riitta Lassila
- Research Program Unit in Oncology, University of Helsinki, Helsinki University Central Hospital, Coagulation Disorders, Helsinki, Finland
| | - Frank W G Leebeek
- Department of Hematology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Pier Mannuccio Mannucci
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center and Fondazione Luigi Villa, Milan, Italy
| | - Renato Marino
- Hemophilia and Thrombosis Centre, University Hospital Policlinico, Bari, Italy
| | - Nikolas Nikšić
- Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom
| | - Florian Oyen
- Department of Pediatric Hematology and Oncology, University Medical Centre, Hamburg-Eppendorf, Hamburg, Germany
| | - Cristina Santoro
- Hematology, Hemophilia and Thrombosis Center, University Hospital Policlinico Umberto I, Rome, Italy
| | - Andreas Tiede
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Gholamreza Toogeh
- Thrombosis Hemostasis Research Center-Vali-Asr Hospital-Emam Khameini Complex Hospital, Tehran University of Medical Science, Tehran, Islamic Republic of Iran
| | - Alberto Tosetto
- Hemophilia and Thrombosis Center, Hematology Department, San Bortolo Hospital, Vicenza, Italy
| | - Marc Trossaert
- Centre Régional de Traitement de l'Hémophilie-Laboratoire d'Hématologie, Nantes, France
| | | | - Jeroen Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Augusto B Federici
- Hematology and Transfusion Medicine, L. Sacco University Hospital, Department of Oncology and Oncohematology, University of Milan, Milan, Italy; and
| | - Flora Peyvandi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center and Fondazione Luigi Villa, Milan, Italy
- Department of Pathophysiology and Transplantation and Università degli Studi di Milano and Milan and Italy
| |
Collapse
|
15
|
Meszaros M, Kunos L, Tarnoki AD, Tarnoki DL, Lazar Z, Bikov A. The Role of Soluble Low-Density Lipoprotein Receptor-Related Protein-1 in Obstructive Sleep Apnoea. J Clin Med 2021; 10:jcm10071494. [PMID: 33916750 PMCID: PMC8038392 DOI: 10.3390/jcm10071494] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/25/2021] [Accepted: 03/28/2021] [Indexed: 12/13/2022] Open
Abstract
Intermittent hypoxia in obstructive sleep apnoea (OSA) is related to inflammation and metabolic abnormalities. Soluble low-density lipoprotein receptor-related protein-1 (sLRP-1) is involved in anti-inflammatory and metabolic processes. However, its ligand, calreticulin (CALR) promotes pro-inflammatory responses and apoptosis. Our aim was to analyse the levels of these biomarkers in OSA. We recruited 46 patients with OSA and 30 control subjects. Inpatient sleep study was performed and fasting plasma samples were collected. Triglyceride glucose index (TyG) and atherogenic index of plasma (AIP) were calculated. Plasma sLRP-1 levels were significantly lower in the OSA group compared to the controls (1.67 (0.90–2.11) mg/L vs. 1.99 (1.53–3.51) mg/L; p = 0.04) after adjustment for age, gender, BMI and lipid profile. Plasma sLRP-1 concentrations were inversely related to age (r = −0.29), BMI (r = −0.35), cigarette pack years (r = −0.31), LDL-C (r = −0.34) and triglyceride levels (r = −0.27), TyG (r = −0.37) and AIP (r = −0.27) as well as to the oxygen desaturation index (ODI, r = −0.24; all p < 0.05). BMI (p = 0.01) and ODI (p = 0.04) were independent predictors for low sLRP-1 levels. CALR did not differ significantly between the two groups (0.23 (0.17–0.34) ng/mL vs. 0.24 (0.20–0.36) ng/mL p = 0.76). We detected lower sLRP-1 levels in subjects with OSA which could contribute to metabolic abnormalities associated with this disease.
Collapse
Affiliation(s)
- Martina Meszaros
- Department of Pulmonology, Semmelweis University, 1083 Budapest, Hungary; (M.M.); (L.K.); (Z.L.)
| | - Laszlo Kunos
- Department of Pulmonology, Semmelweis University, 1083 Budapest, Hungary; (M.M.); (L.K.); (Z.L.)
| | - Adam Domonkos Tarnoki
- Medical Imaging Centre, Semmelweis University, 1082 Budapest, Hungary; (A.D.T.); (D.L.T.)
| | - David Laszlo Tarnoki
- Medical Imaging Centre, Semmelweis University, 1082 Budapest, Hungary; (A.D.T.); (D.L.T.)
| | - Zsofia Lazar
- Department of Pulmonology, Semmelweis University, 1083 Budapest, Hungary; (M.M.); (L.K.); (Z.L.)
| | - Andras Bikov
- North West Lung Centre, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester M13 9NT, UK
- Correspondence: ; Tel.: +44-1612912493; Fax: +44-1612915730
| |
Collapse
|
16
|
Bikov A, Meszaros M, Schwarz EI. Coagulation and Fibrinolysis in Obstructive Sleep Apnoea. Int J Mol Sci 2021; 22:ijms22062834. [PMID: 33799528 PMCID: PMC8000922 DOI: 10.3390/ijms22062834] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 12/29/2022] Open
Abstract
Obstructive sleep apnoea (OSA) is a common disease which is characterised by repetitive collapse of the upper airways during sleep resulting in chronic intermittent hypoxaemia and frequent microarousals, consequently leading to sympathetic overflow, enhanced oxidative stress, systemic inflammation, and metabolic disturbances. OSA is associated with increased risk for cardiovascular morbidity and mortality, and accelerated coagulation, platelet activation, and impaired fibrinolysis serve the link between OSA and cardiovascular disease. In this article we briefly describe physiological coagulation and fibrinolysis focusing on processes which could be altered in OSA. Then, we discuss how OSA-associated disturbances, such as hypoxaemia, sympathetic system activation, and systemic inflammation, affect these processes. Finally, we critically review the literature on OSA-related changes in markers of coagulation and fibrinolysis, discuss potential reasons for discrepancies, and comment on the clinical implications and future research needs.
Collapse
Affiliation(s)
- Andras Bikov
- North West Lung Centre, Manchester University NHS Foundation Trust, Manchester M23 9LT, UK
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester M13 9MT, UK
- Correspondence: ; Tel.: +44-161-291-2493; Fax: +44-161-291-5730
| | - Martina Meszaros
- Department of Pulmonology, Semmelweis University, 1083 Budapest, Hungary;
- Department of Pulmonology and Sleep Disorders Centre, University Hospital Zurich, 8006 Zurich, Switzerland;
| | - Esther Irene Schwarz
- Department of Pulmonology and Sleep Disorders Centre, University Hospital Zurich, 8006 Zurich, Switzerland;
- Centre of Competence Sleep & Health Zurich, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
17
|
Zanetto A, Senzolo M, Campello E, Bulato C, Gavasso S, Shalaby S, Gambato M, Vitale A, Cillo U, Farinati F, Russo FP, Simioni P, Burra P. Influence of Hepatocellular Carcinoma on Platelet Aggregation in Cirrhosis. Cancers (Basel) 2021; 13:cancers13051150. [PMID: 33800224 PMCID: PMC7962527 DOI: 10.3390/cancers13051150] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Platelets are blood cells, the main function of which is to form clots and prevent/stop bleeding. However, it has been shown that platelets may be involved in additional pathophysiological processes, including stimulation of cancer growth and metastasis. In fact, inhibition of platelets in patients with various types of cancer has resulted in lower risks of cancer progression and death. This possibility has not yet been considered in patients with cirrhosis (chronic liver disease) and hepatocellular carcinoma (the most common type of liver cancer) because their platelet function has never been investigated. In this study, we show that hepatocellular carcinoma in patients with cirrhosis is associated with significantly altered (increased) platelet function. This paves the way for further studies to evaluate whether the inhibition of these hyper-functional platelets could be beneficial in patients with cirrhosis and hepatocellular carcinoma. Abstract Hyper-functional platelets are being proposed as a potential therapeutic target in multiple cancers. Whether this can be considered in patients with cirrhosis and hepatocellular carcinoma (HCC) is unknown as their platelet function has not yet been investigated. We evaluated platelet function in cirrhosis patients with HCC. Patients with cirrhosis with and without HCC were prospectively recruited. Platelet aggregation, a marker of platelet function, was assessed by impedance aggregometry with adenosine diphosphate (ADP), arachidonic acid (ASPI), and thrombin (TRAP) stimulation. Plasmatic levels of Von Willebrand factor antigen (VWF) were also determined. One-hundred patients were recruited (50 cirrhotics with and 50 without HCC). Cirrhosis severity by Child class and platelet count were comparable between cirrhotics with and without HCC. Cirrhotics with HCC had higher ADP- (45 vs. 28; p < 0.001), ASPI- (47 vs. 28; p < 0.001), and TRAP- (85 vs. 75; p = 0.01) induced platelet aggregation than cirrhotics without HCC, all indicative of platelet hyper-function. The relatively increased platelet aggregation in patients with HCC was confirmed after adjusting the analysis for platelet count/severity of thrombocytopenia. Levels of VWF were higher in patients with vs. without HCC (348 vs. 267; p = 0.006), particularly in compensated cirrhosis. In patients with cirrhosis, HCC is associated with increased platelet aggregation and higher VWF. The clinical implications of these findings deserve further investigation.
Collapse
Affiliation(s)
- Alberto Zanetto
- Gastroenterology, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy; (A.Z.); (M.S.); (S.S.); (F.F.); (F.P.R.)
- Multivisceral Transplant Unit, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy;
| | - Marco Senzolo
- Gastroenterology, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy; (A.Z.); (M.S.); (S.S.); (F.F.); (F.P.R.)
- Multivisceral Transplant Unit, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy;
| | - Elena Campello
- Thrombotic and Hemorrhagic Diseases Unit, General Internal Medicine, Padova University Hospital, 35128 Padova, Italy; (E.C.); (C.B.); (S.G.)
| | - Cristiana Bulato
- Thrombotic and Hemorrhagic Diseases Unit, General Internal Medicine, Padova University Hospital, 35128 Padova, Italy; (E.C.); (C.B.); (S.G.)
| | - Sabrina Gavasso
- Thrombotic and Hemorrhagic Diseases Unit, General Internal Medicine, Padova University Hospital, 35128 Padova, Italy; (E.C.); (C.B.); (S.G.)
| | - Sarah Shalaby
- Gastroenterology, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy; (A.Z.); (M.S.); (S.S.); (F.F.); (F.P.R.)
- Multivisceral Transplant Unit, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy;
| | - Martina Gambato
- Multivisceral Transplant Unit, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy;
| | - Alessandro Vitale
- Hepatobiliary Surgery and Liver Transplantation Center, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy; (A.V.); (U.C.)
| | - Umberto Cillo
- Hepatobiliary Surgery and Liver Transplantation Center, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy; (A.V.); (U.C.)
| | - Fabio Farinati
- Gastroenterology, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy; (A.Z.); (M.S.); (S.S.); (F.F.); (F.P.R.)
| | - Francesco Paolo Russo
- Gastroenterology, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy; (A.Z.); (M.S.); (S.S.); (F.F.); (F.P.R.)
- Multivisceral Transplant Unit, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy;
| | - Paolo Simioni
- Thrombotic and Hemorrhagic Diseases Unit, General Internal Medicine, Padova University Hospital, 35128 Padova, Italy; (E.C.); (C.B.); (S.G.)
- Correspondence: (P.S.); (P.B.); Tel.: +39-049-8212667 (P.S.); +39-049-8212892 (P.B.)
| | - Patrizia Burra
- Gastroenterology, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy; (A.Z.); (M.S.); (S.S.); (F.F.); (F.P.R.)
- Multivisceral Transplant Unit, Department of Surgery, Oncology, and Gastroenterology, Padova University Hospital, 35128 Padova, Italy;
- Correspondence: (P.S.); (P.B.); Tel.: +39-049-8212667 (P.S.); +39-049-8212892 (P.B.)
| |
Collapse
|
18
|
Molecular determinants of the factor VIII/von Willebrand factor complex revealed by BIVV001 cryo-electron microscopy. Blood 2021; 137:2970-2980. [PMID: 33569592 DOI: 10.1182/blood.2020009197] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/01/2021] [Indexed: 12/15/2022] Open
Abstract
Interaction of factor VIII (FVIII) with von Willebrand factor (VWF) is mediated by the VWF D'D3 domains and thrombin-mediated release is essential for hemostasis after vascular injury. VWF-D'D3 mutations resulting in loss of FVIII binding are the underlying cause of von Willebrand disease (VWD) type 2N. Furthermore, the FVIII-VWF interaction has significant implications for the development of therapeutics for bleeding disorders, particularly hemophilia A, in which endogenous VWF clearance imposes a half-life ceiling on replacement FVIII therapy. To understand the structural basis of FVIII engagement by VWF, we solved the structure of BIVV001 by cryo-electron microscopy to 2.9 Å resolution. BIVV001 is a bioengineered clinical-stage FVIII molecule for the treatment of hemophilia A. In BIVV001, VWF-D'D3 is covalently linked to an Fc domain of a B domain-deleted recombinant FVIII (rFVIII) Fc fusion protein, resulting in a stabilized rFVIII/VWF-D'D3 complex. Our rFVIII/VWF structure resolves BIVV001 architecture and provides a detailed spatial understanding of previous biochemical and clinical observations related to FVIII-VWF engagement. Notably, the FVIII acidic a3 peptide region (FVIII-a3), established as a critical determinant of FVIII/VWF complex formation, inserts into a basic groove formed at the VWF-D'/rFVIII interface. Our structure shows direct interaction of sulfated Y1680 in FVIII-a3 and VWF-R816 that, when mutated, leads to severe hemophilia A or VWD type 2N, respectively. These results provide insight on this key coagulation complex, explain the structural basis of many hemophilia A and VWD type 2N mutations, and inform studies to further elucidate how VWF dissociates rapidly from FVIII upon activation.
Collapse
|
19
|
Connell NT, Flood VH, Brignardello-Petersen R, Abdul-Kadir R, Arapshian A, Couper S, Grow JM, Kouides P, Laffan M, Lavin M, Leebeek FWG, O'Brien SH, Ozelo MC, Tosetto A, Weyand AC, James PD, Kalot MA, Husainat N, Mustafa RA. ASH ISTH NHF WFH 2021 guidelines on the management of von Willebrand disease. Blood Adv 2021; 5:301-325. [PMID: 33570647 PMCID: PMC7805326 DOI: 10.1182/bloodadvances.2020003264] [Citation(s) in RCA: 155] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/27/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND von Willebrand disease (VWD) is a common inherited bleeding disorder. Significant variability exists in management options offered to patients. OBJECTIVE These evidence-based guidelines from the American Society of Hematology (ASH), the International Society on Thrombosis and Haemostasis (ISTH), the National Hemophilia Foundation (NHF), and the World Federation of Hemophilia (WFH) are intended to support patients, clinicians, and health care professionals in their decisions about management of VWD. METHODS ASH, ISTH, NHF, and WFH formed a multidisciplinary guideline panel. Three patient representatives were included. The panel was balanced to minimize potential bias from conflicts of interest. The University of Kansas Outcomes and Implementation Research Unit and the McMaster Grading of Recommendations Assessment, Development and Evaluation (GRADE) Centre supported the guideline development process, including performing and updating systematic evidence reviews (through November 2019). The panel prioritized clinical questions and outcomes according to their importance to clinicians and patients. The panel used the GRADE approach, including GRADE Evidence-to-Decision frameworks, to assess evidence and make recommendations, which were subject to public comment. RESULTS The panel agreed on 12 recommendations and outlined future research priorities. CONCLUSIONS These guidelines make key recommendations regarding prophylaxis for frequent recurrent bleeding, desmopressin trials to determine therapy, use of antiplatelet agents and anticoagulant therapy, target VWF and factor VIII activity levels for major surgery, strategies to reduce bleeding during minor surgery or invasive procedures, management options for heavy menstrual bleeding, management of VWD in the context of neuraxial anesthesia during labor and delivery, and management in the postpartum setting.
Collapse
Affiliation(s)
- Nathan T Connell
- Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Veronica H Flood
- Versiti Blood Research Institute, Medical College of Wisconsin, Milwaukee, WI
| | | | - Rezan Abdul-Kadir
- Department of Obstetrics and Gynaecology and Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free Foundation Hospital and Institute for Women's Health, University College London, London, United Kingdom
| | | | | | - Jean M Grow
- Department of Strategic Communication, Marquette University, Milwaukee, WI
| | - Peter Kouides
- Mary M. Gooley Hemophilia Treatment Center, University of Rochester, Rochester, NY
| | - Michael Laffan
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Michelle Lavin
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland and National Coagulation Centre, St James's Hospital, Dublin, Ireland
| | - Frank W G Leebeek
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sarah H O'Brien
- Division of Hematology/Oncology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH
| | | | - Alberto Tosetto
- Hemophilia and Thrombosis Center, Hematology Department, S. Bortolo Hospital, Vicenza, Italy
| | - Angela C Weyand
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI
| | - Paula D James
- Department of Medicine, Queen's University, Kingston, ON, Canada; and
| | - Mohamad A Kalot
- Outcomes and Implementation Research Unit, Division of Nephrology and Hypertension, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Nedaa Husainat
- Outcomes and Implementation Research Unit, Division of Nephrology and Hypertension, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Reem A Mustafa
- Outcomes and Implementation Research Unit, Division of Nephrology and Hypertension, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
20
|
BIVV001, a new class of factor VIII replacement for hemophilia A that is independent of von Willebrand factor in primates and mice. Blood 2020; 135:1484-1496. [PMID: 32078672 DOI: 10.1182/blood.2019001292] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 01/29/2020] [Indexed: 01/19/2023] Open
Abstract
Factor VIII (FVIII) replacement products enable comprehensive care in hemophilia A. Treatment goals in severe hemophilia A are expanding beyond low annualized bleed rates to include long-term outcomes associated with high sustained FVIII levels. Endogenous von Willebrand factor (VWF) stabilizes and protects FVIII from degradation and clearance, but it also subjects FVIII to a half-life ceiling of ∼15 to 19 hours. Increasing recombinant FVIII (rFVIII) half-life further is ultimately dependent upon uncoupling rFVIII from endogenous VWF. We have developed a new class of FVIII replacement, rFVIIIFc-VWF-XTEN (BIVV001), that is physically decoupled from endogenous VWF and has enhanced pharmacokinetic properties compared with all previous FVIII products. BIVV001 was bioengineered as a unique fusion protein consisting of a VWF-D'D3 domain fused to rFVIII via immunoglobulin-G1 Fc domains and 2 XTEN polypeptides (Amunix Pharmaceuticals, Inc, Mountain View, CA). Plasma FVIII half-life after BIVV001 administration in mice and monkeys was 25 to 31 hours and 33 to 34 hours, respectively, representing a three- to fourfold increase in FVIII half-life. Our results showed that multifaceted protein engineering, far beyond a few amino acid substitutions, could significantly improve rFVIII pharmacokinetic properties while maintaining hemostatic function. BIVV001 is the first rFVIII with the potential to significantly change the treatment paradigm for severe hemophilia A by providing optimal protection against all bleed types, with less frequent doses. The protein engineering methods described herein can also be applied to other complex proteins.
Collapse
|
21
|
Huck V, Chen PC, Xu ER, Tischer A, Klemm U, Aponte-Santamaría C, Mess C, Obser T, Kutzki F, König G, Denis CV, Gräter F, Wilmanns M, Auton M, Schneider SW, Schneppenheim R, Hennig J, Brehm MA. Gain-of-Function Variant p.Pro2555Arg of von Willebrand Factor Increases Aggregate Size through Altering Stem Dynamics. Thromb Haemost 2020; 122:226-239. [PMID: 33385180 PMCID: PMC8828397 DOI: 10.1055/a-1344-4405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The multimeric plasma glycoprotein (GP) von Willebrand factor (VWF) is best known for recruiting platelets to sites of injury during primary hemostasis. Generally, mutations in the VWF gene lead to loss of hemostatic activity and thus the bleeding disorder von Willebrand disease. By employing cone and platelet aggregometry and microfluidic assays, we uncovered a platelet GPIIb/IIIa-dependent prothrombotic gain of function (GOF) for variant p.Pro2555Arg, located in the C4 domain, leading to an increase in platelet aggregate size. We performed complementary biophysical and structural investigations using circular dichroism spectra, small-angle X-ray scattering, nuclear magnetic resonance spectroscopy, molecular dynamics simulations on the single C4 domain, and dimeric wild-type and p.Pro2555Arg constructs. C4-p.Pro2555Arg retained the overall structural conformation with minor populations of alternative conformations exhibiting increased hinge flexibility and slow conformational exchange. The dimeric protein becomes disordered and more flexible. Our data suggest that the GOF does not affect the binding affinity of the C4 domain for GPIIb/IIIa. Instead, the increased VWF dimer flexibility enhances temporal accessibility of platelet-binding sites. Using an interdisciplinary approach, we revealed that p.Pro2555Arg is the first VWF variant, which increases platelet aggregate size and shows a shear-dependent function of the VWF stem region, which can become hyperactive through mutations. Prothrombotic GOF variants of VWF are a novel concept of a VWF-associated pathomechanism of thromboembolic events, which is of general interest to vascular health but not yet considered in diagnostics. Thus, awareness should be raised for the risk they pose. Furthermore, our data implicate the C4 domain as a novel antithrombotic drug target.
Collapse
Affiliation(s)
- Volker Huck
- Department of Dermatology and Venereology, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Po-Chia Chen
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Emma-Ruoqi Xu
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Alexander Tischer
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, United States
| | - Ulrike Klemm
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Camilo Aponte-Santamaría
- Max Planck Tandem Group in Computational Biophysics, University of los Andes, Bogotá, Colombia.,Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany
| | - Christian Mess
- Department of Dermatology and Venereology, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Obser
- Department of Dermatology and Venereology, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabian Kutzki
- Molecular Biomechanics Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany.,Institute of Physical Chemistry, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Gesa König
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cécile V Denis
- Laboratory of Hemostasis, Inflammation and Thrombosis, Institut National de la Santé et de la Recherche Médicale UMR_1176, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Frauke Gräter
- Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany.,Molecular Biomechanics Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
| | - Matthias Wilmanns
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany.,University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthew Auton
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, United States
| | - Stefan W Schneider
- Department of Dermatology and Venereology, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Reinhard Schneppenheim
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Janosch Hennig
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Maria A Brehm
- Department of Dermatology and Venereology, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
22
|
Baronciani L, Peyvandi F. How we make an accurate diagnosis of von Willebrand disease. Thromb Res 2020; 196:579-589. [DOI: 10.1016/j.thromres.2019.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/27/2019] [Accepted: 07/14/2019] [Indexed: 11/28/2022]
|
23
|
de Jong A, Dirven RJ, Boender J, Atiq F, Anvar SY, Leebeek FWG, van Vlijmen BJM, Eikenboom J. Ex vivo Improvement of a von Willebrand Disease Type 2A Phenotype Using an Allele-Specific Small-Interfering RNA. Thromb Haemost 2020; 120:1569-1579. [PMID: 32803740 PMCID: PMC7649061 DOI: 10.1055/s-0040-1715442] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Von Willebrand disease (VWD) is the most common inherited bleeding disorder and is mainly caused by dominant-negative mutations in the multimeric protein von Willebrand factor (VWF). These mutations may either result in quantitative or qualitative defects in VWF. VWF is an endothelial protein that is secreted to the circulation upon endothelial activation. Once secreted, VWF multimers bind platelets and chaperone coagulation factor VIII in the circulation. Treatment of VWD focuses on increasing VWF plasma levels, but production and secretion of mutant VWF remain uninterrupted. Presence of circulating mutant VWF might, however, still affect normal hemostasis or functionalities of VWF beyond hemostasis. We hypothesized that inhibition of the production of mutant VWF improves the function of VWF overall and ameliorates VWD phenotypes. We previously proposed the use of allele-specific small-interfering RNAs (siRNAs) that target frequent
VWF
single nucleotide polymorphisms to inhibit mutant
VWF
. The aim of this study is to prove the functionality of these allele-specific siRNAs in endothelial colony-forming cells (ECFCs). We isolated ECFCs from a VWD type 2A patient with an intracellular multimerization defect, reduced VWF collagen binding, and a defective processing of proVWF to VWF. After transfection of an allele-specific siRNA that specifically inhibited expression of mutant VWF, we showed amelioration of the laboratory phenotype, with normalization of the VWF collagen binding, improvement in VWF multimers, and enhanced VWF processing. Altogether, we prove that allele-specific inhibition of the production of mutant VWF by siRNAs is a promising therapeutic strategy to improve VWD phenotypes.
Collapse
Affiliation(s)
- Annika de Jong
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Richard J Dirven
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Johan Boender
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ferdows Atiq
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Seyed Yahya Anvar
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Frank W G Leebeek
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bart J M van Vlijmen
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jeroen Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
24
|
den Exter PL, Beeres SLMA, Eikenboom J, Klok FA, Huisman MV. Anticoagulant treatment and bleeding complications in patients with left ventricular assist devices. Expert Rev Cardiovasc Ther 2020; 18:363-372. [PMID: 32449431 DOI: 10.1080/14779072.2020.1773803] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The treatment options for advanced heart failure patients drastically changed with the introduction of left ventricular assist devices (LVADs), either as bridge to transplant or as destination therapy for patients ineligible for transplant. Despite major benefits in terms of survival, functional status and quality of life, managing patients with LVADs comes with several challenges. The most significant challenge is balancing between the risks of thrombotic and bleeding complications. AREAS COVERED The present review describes the pathophysiological mechanisms explaining the alterations in the hemostatic profile of LVAD patients, and summarizes current evidence to guide clinical decision making with regard to anticoagulant treatment and management of bleeding complications. EXPERT OPINION LVAD patients require life-long anticoagulant therapy to reduce the risk of pump thrombosis. However, exposing LVAD patients to anticoagulant therapy, in combination with common acquired coagulopathies after LVAD implantation such as acquired von Willebrand syndrome, comes with high risks of bleeding. There is a need for randomized controlled trials in LVAD patients to determine the optimal antithrombotic regimen and find the most effective balance between thrombotic and bleeding complications. In addition, strategies to specifically target the acquired von Willebrand syndrome and its associated angiodysplasias need to be evaluated in the LVAD population.
Collapse
Affiliation(s)
- Paul L den Exter
- Department of Thrombosis and Hemostasis, Leiden University Medical Center , Leiden, The Netherlands
| | - Saskia L M A Beeres
- Department of Cardiology, Leiden University Medical Center , Leiden, The Netherlands
| | - Jeroen Eikenboom
- Department of Thrombosis and Hemostasis, Leiden University Medical Center , Leiden, The Netherlands
| | - Frederikus A Klok
- Department of Thrombosis and Hemostasis, Leiden University Medical Center , Leiden, The Netherlands
| | - Menno V Huisman
- Department of Thrombosis and Hemostasis, Leiden University Medical Center , Leiden, The Netherlands
| |
Collapse
|
25
|
Abstract
Von Willebrand factor (VWF) and coagulation factor VIII (FVIII) circulate as a complex in plasma and have a major role in the hemostatic system. VWF has a dual role in hemostasis. It promotes platelet adhesion by anchoring the platelets to the subendothelial matrix of damaged vessels and it protects FVIII from proteolytic degradation. Moreover, VWF is an acute phase protein that has multiple roles in vascular inflammation and is massively secreted from Weibel-Palade bodies upon endothelial cell activation. Activated FVIII on the other hand, together with coagulation factor IX forms the tenase complex, an essential feature of the propagation phase of coagulation on the surface of activated platelets. VWF deficiency, either quantitative or qualitative, results in von Willebrand disease (VWD), the most common bleeding disorder. The deficiency of FVIII is responsible for Hemophilia A, an X-linked bleeding disorder. Here, we provide an overview on the role of the VWF-FVIII interaction in vascular physiology.
Collapse
Affiliation(s)
- Klytaimnistra Kiouptsi
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Mainz, Germany.
| |
Collapse
|
26
|
Abstract
Damoctocog alfa pegol (Jivi®) is approved in the USA, EU, Japan and Canada for the treatment and prophylaxis of previously treated patients aged ≥ 12 years with haemophilia A. Formulated with a 60 kDa polyethylene glycol (PEG) moiety, damoctocog alfa pegol is an intravenously (IV) administered recombinant factor VIII (rFVIII) product with a longer terminal half-life than non-PEGylated FVIII and rFVIII products. In the multinational phase II/III PROTECT VIII trial, prophylaxis with damoctocog alfa pegol reduced the likelihood of bleeding in previously treated patients aged ≥ 12 years with severe haemophilia A, with dosing schedules ranging from twice weekly to once every 7 days. Interim data from the ongoing extension phase indicated that the reduced annualized bleeding rates (ABRs) were maintained for up to 5.2 years of prophylaxis with damoctocog alfa pegol. Damoctocog alfa pegol was also effective in treating bleeding episodes and in providing haemostatic control during surgery. Damoctocog alfa pegol was generally well tolerated in adult and adolescent patients with severe haemophilia A, with most adverse events considered to be unrelated to treatment. There were no new or confirmed cases of FVIII inhibitor development and anti-PEG antibodies, observed in some patients, were of low titre and transient. Damoctocog alfa pegol extends the available treatment options in previously treated adults and adolescents with haemophilia A, offering the possibility of up to once-weekly administration for suitable patients.
Collapse
Affiliation(s)
- Julia Paik
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| | - Emma D Deeks
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand
| |
Collapse
|
27
|
Patsouras MD, Vlachoyiannopoulos PG. Evidence of epigenetic alterations in thrombosis and coagulation: A systematic review. J Autoimmun 2019; 104:102347. [PMID: 31607428 DOI: 10.1016/j.jaut.2019.102347] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
Thrombosis in the context of Cardiovascular disease (CVD) affects mainly the blood vessels supplying the heart, brain and peripheries and it is the leading cause of death worldwide. The pathophysiological thrombotic mechanisms are largely unknown. Heritability contributes to a 30% of the incidence of CVD. The remaining variation can be explained by life style factors such as smoking, dietary and exercise habits, environmental exposure to toxins, and drug usage and other comorbidities. Epigenetic variation can be acquired or inherited and constitutes an interaction between genes and the environment. Epigenetics have been implicated in atherosclerosis, ischemia/reperfusion damage and the cardiovascular response to hypoxia. Epigenetic regulators of gene expression are mainly the methylation of CpG islands, histone post translational modifications (PTMs) and microRNAs (miRNAs). These epigenetic regulators control gene expression either through activation or silencing. Epigenetic control is mostly dynamic and can potentially be manipulated to prevent or reverse the uncontrolled expression of genes, a trait that renders them putative therapeutic targets. In the current review, we systematically studied and present available data on epigenetic alterations implicated in thrombosis derived from human studies. Evidence of epigenetic alterations is observed in several thrombotic diseases such as Coronary Artery Disease and Cerebrovascular Disease, Preeclampsia and Antiphospholipid Syndrome. Differential CpG methylation and specific histone PTMs that control transcription of prothrombotic and proinflammatory genes have also been associated with predisposing factors of thrombosis and CVD, such us smoking, air pollution, hypertriglyceridemia, occupational exposure to particulate matter and comorbidities including cancer, Chronic Obstructive Pulmonary Disease and Chronic Kidney Disease. These clinical observations are further supported by in vitro experiments and indicate that epigenetic regulation affects the pathophysiology of thrombotic disorders with potential diagnostic or therapeutic utility.
Collapse
Affiliation(s)
- M D Patsouras
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - P G Vlachoyiannopoulos
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
28
|
Taves S, Sun J, Livingston EW, Chen X, Amiaud J, Brion R, Hannah WB, Bateman TA, Heymann D, Monahan PE. Hemophilia A and B mice, but not VWF -/-mice, display bone defects in congenital development and remodeling after injury. Sci Rep 2019; 9:14428. [PMID: 31594977 PMCID: PMC6783554 DOI: 10.1038/s41598-019-50787-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 09/19/2019] [Indexed: 12/31/2022] Open
Abstract
While joint damage is the primary co-morbidity of hemophilia, osteoporosis and osteopenia are also observed. Coagulation factor VIII deficient (FVIII−/−) mice develop an osteoporotic phenotype in the absence of induced hemarthrosis that is exacerbated two weeks after an induced joint injury. Here we have compared comprehensively the bone health of clotting factor VIII, factor IX, and Von Willebrand Factor knockout (FVIII−/−, FIX−/−, and VWF−/− respectively) mice both in the absence of joint hemorrhage and following induced joint injury. We found FVIII−/− and FIX−/− mice, but not VWF−/− mice, developmentally have an osteoporotic phenotype. Unilateral induced hemarthrosis causes further bone damage in both FVIII−/− and FIX−/− mice, but has little effect on VWF−/− bone health, indicating that the FVIII.VWF complex is not required for normal bone remodeling in vivo. To further investigate the bone healing following hemarthrosis in hemophilia we examined a two week time course using microCT, serum chemistry, and histological analysis. Elevated ratio of osteoprotegerin (OPG)/receptor activator of nuclear factor-kappa B ligand (RANKL), increased osterix+ osteoblastic cells, and decreased smoothness of the cortical bone surface were evident within several days of injury, indicative of acute heterotopic mineralization along the cortical surface. This was closely followed by increased interleukin-6 (IL-6) levels, increased osteoclast numbers, and significant trabecular bone loss. Uncoupled and disorganized bone formation and resorption continued for the duration of the study resulting in significant deterioration of the joint. Further elucidation of the shared mechanisms underlying abnormal bone homeostasis in the absence of FVIII or FIX is needed to guide evidence-based approaches to the screening and treatment of the prevalent bone defects in hemophilia A and B.
Collapse
Affiliation(s)
- Sarah Taves
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA.,Global Research, Novo Nordisk A/S, Maløv, Denmark
| | - Junjiang Sun
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, USA.,Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Eric W Livingston
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA
| | - Xin Chen
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, USA
| | - Jerome Amiaud
- INSERM, U1238, Faculty of Medicine, Université de Nantes, Nantes, F-44093, France
| | - Regis Brion
- INSERM, U1238, Faculty of Medicine, Université de Nantes, Nantes, F-44093, France
| | - William B Hannah
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, USA
| | - Ted A Bateman
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA.,Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Dominique Heymann
- INSERM, U1232, CRCiNA, Institut de Cancérologie de l'Ouest, Université de Nantes, Université d'Angers, Saint-Herblain, F-44805, France. .,University of Sheffield, INSERM, Associated European Laboratory Sarcoma Research Unit, Department of Oncology and Metabolism, Sheffield, S10 2RX, UK.
| | - Paul E Monahan
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, USA. .,Harold R. Roberts Comprehensive Hemophilia Diagnosis and Treatment Center, University of North Carolina, Chapel Hill, NC, USA. .,Spark Therapeutics, Philadelphia, PA, USA.
| |
Collapse
|
29
|
Genetic determinants of VWF clearance and FVIII binding modify FVIII pharmacokinetics in pediatric hemophilia A patients. Blood 2019; 134:880-891. [DOI: 10.1182/blood.2019000190] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
Abstract
Factor VIII (FVIII) pharmacokinetic (PK) properties show high interpatient variability in hemophilia A patients. Although previous studies have determined that age, body mass index, von Willebrand factor antigen (VWF:Ag) levels, and ABO blood group status can influence FVIII PK, they do not account for all observed variability. In this study, we aim to describe the genetic determinants that modify the FVIII PK profile in a population of 43 pediatric hemophilia A patients. We observed that VWF:Ag and VWF propeptide (VWFpp)/VWF:Ag, but not VWFpp, were associated with FVIII half-life. VWFpp/VWF:Ag negatively correlated with FVIII half-life in patients with non-O blood type, but no correlation was observed for type O patients, suggesting that von Willebrand factor (VWF) half-life, as modified by the ABO blood group, is a strong regulator of FVIII PK. The FVIII-binding activity of VWF positively correlated with FVIII half-life, and the rare or low-frequency nonsynonymous VWF variants p.(Arg826Lys) and p.(Arg852Glu) were identified in patients with reduced VWF:FVIIIB but not VWF:Ag. Common variants at the VWF, CLEC4M, and STAB2 loci, which have been previously associated with plasma levels of VWF and FVIII, were associated with the FVIII PK profile. Together, these studies characterize the mechanistic basis by which VWF clearance and ABO glycosylation modify FVIII PK in a pediatric population. Moreover, this study is the first to identify non-VWF and non-ABO variants that modify FVIII PK in pediatric hemophilia A patients.
Collapse
|
30
|
Abrantes JA, Solms A, Garmann D, Nielsen EI, Jönsson S, Karlsson MO. Relationship between factor VIII activity, bleeds and individual characteristics in severe hemophilia A patients. Haematologica 2019; 105:1443-1453. [PMID: 31371418 PMCID: PMC7193498 DOI: 10.3324/haematol.2019.217133] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 07/23/2019] [Indexed: 01/19/2023] Open
Abstract
Pharmacokinetic-based prophylaxis of replacement factor VIII (FVIII) products has been encouraged in recent years, but the relationship between exposure (factor VIII activity) and response (bleeding frequency) remains unclear. The aim of this study was to characterize the relationship between FVIII dose, plasma FVIII activity, and bleeding patterns and individual characteristics in severe hemophilia A patients. Pooled pharmacokinetic and bleeding data during prophylactic treatment with BAY 81-8973 (octocog alfa) were obtained from the three LEOPOLD trials. The population pharmacokinetics of FVIII activity and longitudinal bleeding frequency, as well as bleeding severity, were described using non-linear mixed effects modeling in NONMEM. In total, 183 patients [median age 22 years (range, 1-61); weight 60 kg (11-124)] contributed with 1,535 plasma FVIII activity observations, 633 bleeds and 11 patient/study characteristics [median observation period 12 months (3.1-13.1)]. A parametric repeated time-to-categorical bleed model, guided by plasma FVIII activity from a 2-compartment population pharmacokinetic model, described the time to the occurrence of bleeds and their severity. Bleeding probability decreased with time of study, and a bleed was not found to affect the time of the next bleed. Several covariate effects were identified, including the bleeding history in the 12-month pre-study period increasing the bleeding hazard. However, unexplained inter-patient variability in the phenotypic bleeding pattern remained large (111%CV). Further studies to translate the model into a tool for dose individualization that considers the individual bleeding risk are required. Research was based on a post-hoc analysis of the LEOPOLD studies registered at clinicaltrials.gov identifiers: 01029340, 01233258 and 01311648.
Collapse
Affiliation(s)
- João A Abrantes
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | | | - Elisabet I Nielsen
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Siv Jönsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Mats O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
31
|
Swystun LL, Notley C, Georgescu I, Lai JD, Nesbitt K, James PD, Lillicrap D. The endothelial lectin clearance receptor CLEC4M binds and internalizes factor VIII in a VWF-dependent and independent manner. J Thromb Haemost 2019; 17:681-694. [PMID: 30740857 PMCID: PMC7083068 DOI: 10.1111/jth.14404] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Indexed: 01/23/2023]
Abstract
Essentials CLEC4M is an endocytic receptor for factor FVIII. CLEC4M interacts with FVIII in a VWF-dependent and independent manner. CLEC4M binds to mannose-containing glycans on FVIII. CLEC4M internalization of FVIII involves clathrin coated pits. SUMMARY: Background von Willebrand factor (VWF) and factor VIII (FVIII) circulate in the plasma as a non-covalent complex, and the majority of FVIII is likely to be cleared by VWF-dependent pathways. Clearance of VWF-free FVIII is rapid and underlies the pathological basis of some quantitative FVIII deficiencies. The receptor pathways that regulate the clearance of VWF-bound and VWF-free FVIII are incompletely uncharacterized. The human liver-expressed endothelial lectin CLEC4M has been previously characterized as a clearance receptor for VWF, although its influence on FVIII is unknown. Objective The interaction between FVIII and CLEC4M was characterized in the presence or absence of VWF. Methods FVIII interactions with CLEC4M were evaluated by in vitro cell-based and solid phase binding assays. Interactions between FVIII and CLEC4M or liver sinusoidal endothelial cells were evaluated in vivo by immunohistochemistry. Results CLEC4M-expressing HEK 293 cells bound and internalized recombinant and plasma-derived FVIII through VWF-dependent and independent mechanisms. CLEC4M binding to recombinant FVIII was dependent on mannose-exposed N-linked glycans. CLEC4M mediated FVIII internalization via a clathrin-coated pit-dependent mechanism, resulting in transport of FVIII from early and late endosomes for catabolism by lysosomes. In vivo hepatic expression of CLEC4M after hydrodynamic liver transfer was associated with a decrease in plasma levels of endogenous murine FVIII:C in normal mice, whereas infused recombinant human FVIII was associated with sinusoidal endothelial cells in the presence or absence of VWF. Conclusions These findings suggest that CLEC4M is a novel clearance receptor that interacts with mannose-exposed glycans on FVIII in the presence or absence of VWF.
Collapse
Affiliation(s)
- Laura L Swystun
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Colleen Notley
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Ilinca Georgescu
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Jesse D Lai
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Kate Nesbitt
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Paula D James
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| |
Collapse
|
32
|
Chang IJ, Byers HM, Ng BG, Merritt JL, Gilmore R, Shrimal S, Wei W, Zhang Y, Blair AB, Freeze HH, Zhang B, Lam C. Factor VIII and vWF deficiency in STT3A-CDG. J Inherit Metab Dis 2019; 42:325-332. [PMID: 30701557 PMCID: PMC6658093 DOI: 10.1002/jimd.12021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/08/2018] [Accepted: 12/06/2018] [Indexed: 11/09/2022]
Abstract
STT3A-CDG (OMIM# 615596) is an autosomal recessive N-linked glycosylation disorder characterized by seizures, developmental delay, intellectual disability, and a type I carbohydrate deficient transferrin pattern. All previously reported cases (n = 6) have been attributed to a homozygous pathogenic missense variant c.1877C>T (p.Val626Ala) in STT3A. We describe a patient with a novel homozygous likely pathogenic missense variant c.1079A>C (p.Tyr360Ser) who presents with chronically low Factor VIII (FVIII) and von Willebrand Factor (vWF) levels and activities in addition to the previously reported symptoms of developmental delay and seizures. VWF in our patient's plasma is present in a mildly hypoglycosylated form. FVIII antigen levels were too low to quantify in our patient. Functional studies with STT3A-/- HEK293 cells showed severely reduced FVIII antigen and activity levels in conditioned media <10% expected, but normal intracellular levels. We also show decreased glycosylation of STT3A-specific acceptors in fibroblasts from our patient, providing a mechanistic explanation for how STT3A deficiency leads to a severe defect in FVIII secretion. Our results suggest that certain STT3A-dependent N-glycans are required for efficient FVIII secretion, and the decreased FVIII level in our patient is a combined effect of both severely impaired FVIII secretion and lower plasma VWF level. Our report expands both the genotype and phenotype of STT3A-CDG; demonstrating, as in most types of CDG, that there are multiple disease-causing variants in STT3A.
Collapse
Affiliation(s)
- Irene J. Chang
- Department of Pediatrics, Division of Medical Genetics, University of Washington, Seattle, Washington
| | - Heather M. Byers
- Department of Pediatrics, Division of Medical Genetics, Stanford University, Stanford, California
| | - Bobby G. Ng
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - John Lawrence Merritt
- Department of Pediatrics, Division of Medical Genetics, University of Washington, Seattle, Washington
| | - Reid Gilmore
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Shiteshu Shrimal
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Wei Wei
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Yuan Zhang
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Amanda B. Blair
- Department of Pediatrics, Division of Hematology-Oncology, University of Washington, Seattle, Washington
| | - Hudson H. Freeze
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Bin Zhang
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Christina Lam
- Department of Pediatrics, Division of Medical Genetics, University of Washington, Seattle, Washington
| |
Collapse
|
33
|
Nakhaei-Nejad M, Farhan M, Mojiri A, Jabbari H, Murray AG, Jahroudi N. Regulation of von Willebrand Factor Gene in Endothelial Cells That Are Programmed to Pluripotency and Differentiated Back to Endothelial Cells. Stem Cells 2019; 37:542-554. [PMID: 30682218 DOI: 10.1002/stem.2978] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/13/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022]
Abstract
Endothelial cells play a central role in physiological function and pathophysiology of blood vessels in health and disease. However, the molecular mechanism that establishes the endothelial phenotype, and contributes to its signature cell type-specific gene expression, is not yet understood. We studied the regulation of a highly endothelial-specific gene, von Willebrand factor (VWF), in induced pluripotent stem cells generated from primary endothelial cells (human umbilical vein endothelial cells [HUVEC] into a pluripotent state [HiPS]) and subsequently differentiated back into endothelial cells. This allowed us to explore how VWF expression is regulated when the endothelial phenotype is revoked (endothelial cells to HiPS), and re-established (HiPS back to endothelial cells [EC-Diff]). HiPS were generated from HUVECs, their pluripotency established, and then differentiated back to endothelial cells. We established phenotypic characteristics and robust angiogenic function of EC-Diff. Gene array analyses, VWF chromatin modifications, and transacting factors binding assays were performed on the three cell types (HUVEC, HiPS, and EC-Diff). The results demonstrated that generally cohorts of transacting factors that function as transcriptional activators, and those that contribute to histone acetylation and DNA demethylation, were significantly decreased in HiPS compared with HUVECs and EC-Diff. In contrast, there were significant increases in the gene expression levels of epigenetic modifiers that function as methyl transferases in HiPS compared with endothelial cells. The results demonstrated that alterations in chromatin modifications of the VWF gene, in addition to expression and binding of transacting factors that specifically function as activators, are responsible for establishing endothelial specific regulation of the VWF gene. Stem Cells 2019;37:542-554.
Collapse
Affiliation(s)
| | - Maikel Farhan
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Anahita Mojiri
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Hosna Jabbari
- Department of Computer Science, University of Vermont, Burlington, Vermont, USA
| | - Allan G Murray
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Nadia Jahroudi
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
34
|
Xu ER, von Bülow S, Chen PC, Lenting PJ, Kolšek K, Aponte-Santamaría C, Simon B, Foot J, Obser T, Schneppenheim R, Gräter F, Denis CV, Wilmanns M, Hennig J. Structure and dynamics of the platelet integrin-binding C4 domain of von Willebrand factor. Blood 2019; 133:366-376. [PMID: 30305279 PMCID: PMC6450055 DOI: 10.1182/blood-2018-04-843615] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 09/24/2018] [Indexed: 12/22/2022] Open
Abstract
Von Willebrand factor (VWF) is a key player in the regulation of hemostasis by promoting recruitment of platelets to sites of vascular injury. An array of 6 C domains forms the dimeric C-terminal VWF stem. Upon shear force activation, the stem adopts an open conformation allowing the adhesion of VWF to platelets and the vessel wall. To understand the underlying molecular mechanism and associated functional perturbations in disease-related variants, knowledge of high-resolution structures and dynamics of C domains is of paramount interest. Here, we present the solution structure of the VWF C4 domain, which binds to the platelet integrin and is therefore crucial for the VWF function. In the structure, we observed 5 intra- and inter-subdomain disulfide bridges, of which 1 is unique in the C4 domain. The structure further revealed an unusually hinged 2-subdomain arrangement. The hinge is confined to a very short segment around V2547 connecting the 2 subdomains. Together with 2 nearby inter-subdomain disulfide bridges, this hinge induces slow conformational changes and positional alternations of both subdomains with respect to each other. Furthermore, the structure demonstrates that a clinical gain-of-function VWF variant (Y2561) is more likely to have an effect on the arrangement of the C4 domain with neighboring domains rather than impairing platelet integrin binding.
Collapse
Affiliation(s)
- Emma-Ruoqi Xu
- Hamburg Unit, European Molecular Biology Laboratory, Hamburg, Germany
| | - Sören von Bülow
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Po-Chia Chen
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Peter J Lenting
- INSERM, UMR_S 1176, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Katra Kolšek
- Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
| | - Camilo Aponte-Santamaría
- Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing, Mathematikon, Heidelberg University, Heidelberg, Germany
- Max Planck Tandem Group in Computational Biophysics, University of Los Andes, Bogotá, Colombia
| | - Bernd Simon
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jaelle Foot
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Tobias Obser
- Department of Pediatric Hematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany; and
| | - Reinhard Schneppenheim
- Department of Pediatric Hematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany; and
| | - Frauke Gräter
- Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
| | - Cécile V Denis
- INSERM, UMR_S 1176, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Matthias Wilmanns
- Hamburg Unit, European Molecular Biology Laboratory, Hamburg, Germany
- University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Janosch Hennig
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
35
|
Riddell A, Vinayagam S, Gomez K, Laffan M, McKinnon T. Evaluation of von Willebrand factor concentrates by platelet adhesion to collagen using an in vitro flow assay. Res Pract Thromb Haemost 2019; 3:126-135. [PMID: 30656286 PMCID: PMC6332833 DOI: 10.1002/rth2.12166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 10/19/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Von Willebrand disease (VWD) results from quantitative or qualitative deficiency of von Willebrand factor (VWF) and is treated using VWF-containing concentrates. Several studies have compared the function of various VWF containing concentrates however this has not been performed using shear based assays. OBJECTIVES To compare the platelet-capture potential of 10 commercially available, plasma-derived VWF concentrates under shear conditions. METHODS VWF containing concentrates were assessed for VWF:Ag, VWF:CB, VWF:RCo, factor VIII:C ADAMTS13 content, VWF multimeric profile and glycan content using lectin binding assays. Free-thiol content of each concentrate was investigated using MPB binding assays. An in vitro flow assay was used to determine the ability of each concentrate to mediate platelet capture to collagen. RESULTS VWF multimeric analysis revealed reduction of high molecular weight (HMW) forms in four of the concentrates (Alphante, Octanate and Haemoctin, and 8Y). The high MW multimer distribution of the remaining six concentrates (Optivate, Wilate, Fandhi, Wilfactin, Haemate P, and Voncento) was similar to the plasma control. Lectin analysis demonstrated that 8Y had increased amount of T-antigen. Although platelet capture after 5 minutes perfusion was similar for all concentrates; Alphante, Octanate, and Haemoctin, demonstrated the lowest levels of platelet capture after 60 seconds of perfusion. Free-thiol content and ADAMTS13 levels varied widely between the concentrates but was not correlated with function. CONCLUSION Alphanate, Octanate, and Haemoctin, lacked HMW multimers and had the lowest initial platelet capture levels suggesting that the presence of VWF HMW multimers are required for initial platelet deposition.
Collapse
Affiliation(s)
- Anne Riddell
- Katharine Dormandy Haemophilia Centre and Thrombosis UnitRoyal Free London NHS Foundation TrustLondonUK
| | - Saravanan Vinayagam
- Katharine Dormandy Haemophilia Centre and Thrombosis UnitRoyal Free London NHS Foundation TrustLondonUK
| | - Keith Gomez
- Katharine Dormandy Haemophilia Centre and Thrombosis UnitRoyal Free London NHS Foundation TrustLondonUK
| | - Mike Laffan
- Department of HaematologyImperial College of Science Technology and MedicineLondonUK
| | - Tom McKinnon
- Department of HaematologyImperial College of Science Technology and MedicineLondonUK
| |
Collapse
|
36
|
A factor VIII–nanobody fusion protein forming an ultrastable complex with VWF: effect on clearance and antibody formation. Blood 2018; 132:1193-1197. [DOI: 10.1182/blood-2018-01-829523] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/25/2018] [Indexed: 01/26/2023] Open
Abstract
Key Points
The fusion between FVIII and anti-VWF nanobodies increases affinity for VWF 25-fold without compromising FVIII activity. Stabilized VWF binding results in a twofold enhanced circulatory survival of FVIII and reduced anti-FVIII antibody formation.
Collapse
|
37
|
Loomans JI, Stokhuijzen E, Peters M, Fijnvandraat K. Administration of DDAVP did not improve the pharmacokinetics of FVIII concentrate in a clinically significant manner. J Clin Transl Res 2018; 3:351-357. [PMID: 30873482 PMCID: PMC6412613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The half-life and mean residence time (MRT) of infused recombinant factor VIII (FVIII) concentrate are associated with pre-infusion levels of von Willebrand factor (VWF) in severely affected hemophilia A patients. It is currently unknown if individual FVIII concentrate half-life and MRT can be extended by increasing endogenous VWF levels. Aim: Our aim was to evaluate the effect of a 1-deamino-8-D-arginine vasopressin (DDAVP)-induced rise in VWF concentration on the pharmacokinetics of infused FVIII in hemophilia A patients. METHODS Four adult hemophilia A patients participated in this cross-over, placebo-controlled study. Each patient received either intravenous DDAVP or placebo, one hour prior to administration of 50 IU/kg plasma-derived immune-affinity purified FVIII concentrate. RESULTS The combined administration of DDAVP and FVIII concentrate was well tolerated. The levels of VWF Antigen (Ag) doubled after DDAVP, whereas they remained stable after placebo infusion. This rise in VWF Ag resulted in a slight modification of the pharmacokinetic parameters of FVIII concentrate. The MRT of FVIII concentrate increased in all patients (mean from 17.6 h to 19.9 h, p < 0.001, 95% CI for MRT change: +4.7 to -0.3 h). However, in vivo recoveries tended to decrease following DDAVP administration. CONCLUSIONS Collectively, these data show that administration of DDAVP did not improve the pharmacokinetics of FVIII concentrate in a clinically significant manner. RELEVANCE FOR PATIENTS Our results indicate that no clinical benefit is to be expected from the modification in FVIII pharmacokinetics resulting from DDAVP-administration prior to infusion of FVIII concentrate in hemophilia A patients.
Collapse
Affiliation(s)
- Janneke I Loomans
- Emma Children's Hospital, Department of Pediatric Hematology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Eva Stokhuijzen
- Emma Children's Hospital, Department of Pediatric Hematology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Plasma Proteins, Sanquin Research, Amsterdam, the Netherlands
| | - Marjolein Peters
- Emma Children's Hospital, Department of Pediatric Hematology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Karin Fijnvandraat
- Emma Children's Hospital, Department of Pediatric Hematology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Plasma Proteins, Sanquin Research, Amsterdam, the Netherlands
| |
Collapse
|
38
|
Chetcuti Zammit S, Koulaouzidis A, Sanders DS, McAlindon ME, Rondonotti E, Yung DE, Sidhu R. Overview of small bowel angioectasias: clinical presentation and treatment options. Expert Rev Gastroenterol Hepatol 2018; 12:125-139. [PMID: 28994309 DOI: 10.1080/17474124.2018.1390429] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Elderly patients with multiple co-morbidities are at an increased risk of developing small bowel angioectasias. Treating these lesions can be both challenging and costly with patients requiring extensive investigations and recurrent admissions for iron infusions and blood transfusions as well as invasive procedures. This review presents treatment options and describes in detail drugs that should be considered whilst taking into account their effectiveness and their safety profile. Areas covered: A PubMed search was carried out using the following keywords: small bowel angiodysplasias, small bowel angioectasias, small bowel bleeding and obscure gastrointestinal bleeding to assess existing evidence. The pathophysiology and risk factors are covered in this review together with appropriate methods of investigation and management. Treatment options discussed are endoscopic measures, surgical options and pharmacotherapy. The role of serum biomarkers is also discussed. Expert commentary: Future work should be directed at alternative drugs with a good safety profile that target biomarkers. Novel pharmacotherapy directed at biomarkers could potentially provide a non-invasive treatment option for angioectasias particularly in the elderly where management can be challenging.
Collapse
Affiliation(s)
| | | | - David S Sanders
- a Gastroenterology Department , Royal Hallamshire Hospital , Sheffield , UK
| | - Mark E McAlindon
- a Gastroenterology Department , Royal Hallamshire Hospital , Sheffield , UK
| | | | - Diana E Yung
- b Endoscopy Unit , the Royal Infirmary of Edinburgh , Edinburgh , UK
| | - Reena Sidhu
- a Gastroenterology Department , Royal Hallamshire Hospital , Sheffield , UK
| |
Collapse
|
39
|
Casonato A, Galletta E, Sarolo L, Daidone V. Type 2N von Willebrand disease: Characterization and diagnostic difficulties. Haemophilia 2017; 24:134-140. [PMID: 29115006 DOI: 10.1111/hae.13366] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2017] [Indexed: 11/30/2022]
Abstract
INTRODUCTION An abnormal factor VIII (FVIII) binding capacity of von Willebrand factor (VWF) identifies type 2N von Willebrand disease (VWD). Type 2N VWD patients are identified by means of the VWF FVIII binding (VWF:FVIIIB) assay, and especially their VWF:FVIIIB/VWF:Ag ratio (VWF:FVIIIB ratio). AIM We report on our 15-year experience of diagnosing type 2N VWD. METHODS We have performed 2178 VWF:FVIIIB assays in bleeders and normal subjects. RESULTS von Willebrand factor (VWF):FVIIIB was reduced in 682, but only 60 had low VWF:FVIIIB ratios (<0.74). Among nine patients who had a VWF:FVIIIB ratio below 0.3, four had normal VWF levels and were homozygotes for the p.R854Q mutation; the other five had low VWF levels due to a quantitative VWF mutation combined with p.R854Q. The VWF:FVIIIB ratio ranged between 0.3 and 0.73 in 51 subjects; 34 of them were heterozygotes for the p.R854Q mutation, while one carried the p.R760C. The heterozygotes for type 2N included subjects with or without bleeding symptoms, the former with significantly lower mean VWF levels than the latter. Among the 116 normal subjects tested, six were heterozygotes for the p.R854Q mutation (all asymptomatic). CONCLUSIONS The prevalence of type 2N in our VWD cohort was 2.5%, and 5.2% of the general population in Northeast Italy was found heterozygous for the p.R854Q mutation. It might be difficult to reveal a type 2N defect using routine tests alone, especially when it is combined with a quantitative VWF mutation. Accordingly, we always recommend VWF:FVIIIB assay in the diagnostic workup of VWD.
Collapse
Affiliation(s)
- A Casonato
- Department of Medicine, Hemorrhagic and Thrombotic Disorders Unit, University of Padua Medical School, Padua, Italy
| | - E Galletta
- Department of Medicine, Hemorrhagic and Thrombotic Disorders Unit, University of Padua Medical School, Padua, Italy
| | - L Sarolo
- Department of Medicine, Hemorrhagic and Thrombotic Disorders Unit, University of Padua Medical School, Padua, Italy
| | - V Daidone
- Department of Medicine, Hemorrhagic and Thrombotic Disorders Unit, University of Padua Medical School, Padua, Italy
| |
Collapse
|
40
|
de Jong A, Eikenboom J. Von Willebrand disease mutation spectrum and associated mutation mechanisms. Thromb Res 2017; 159:65-75. [PMID: 28987708 DOI: 10.1016/j.thromres.2017.09.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/13/2017] [Accepted: 09/22/2017] [Indexed: 01/24/2023]
Abstract
Von Willebrand disease (VWD) is a bleeding disorder that is mainly caused by mutations in the multimeric protein von Willebrand factor (VWF). These mutations may lead to deficiencies in plasma VWF or dysfunctional VWF. VWF is a heterogeneous protein and over the past three decades, hundreds of VWF mutations have been identified. In this review we have organized all reported mutations, spanning a timeline from the late eighties until early 2017. This resulted in an overview of 750 unique mutations that are divided over the VWD types 1, 2A, 2B, 2M, 2N and 3. For many of these mutations the disease-causing effects have been characterized in vitro through expression studies, ex vivo by analysis of patient-derived endothelial cells, as well as in animal or (bio)physical models. Here we describe the mechanisms associated with the VWF mutations per VWD type.
Collapse
Affiliation(s)
- Annika de Jong
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
41
|
Identification of extant vertebrate Myxine glutinosa VWF: evolutionary conservation of primary hemostasis. Blood 2017; 130:2548-2558. [PMID: 28899852 DOI: 10.1182/blood-2017-02-770792] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 08/23/2017] [Indexed: 11/20/2022] Open
Abstract
Hemostasis in vertebrates involves both a cellular and a protein component. Previous studies in jawless vertebrates (cyclostomes) suggest that the protein response, which involves thrombin-catalyzed conversion of a soluble plasma protein, fibrinogen, into a polymeric fibrin clot, is conserved in all vertebrates. However, similar data are lacking for the cellular response, which in gnathostomes is regulated by von Willebrand factor (VWF), a glycoprotein that mediates the adhesion of platelets to the subendothelial matrix of injured blood vessels. To gain evolutionary insights into the cellular phase of coagulation, we asked whether a functional vwf gene is present in the Atlantic hagfish, Myxine glutinosa We found a single vwf transcript that encodes a simpler protein compared with higher vertebrates, the most striking difference being the absence of an A3 domain, which otherwise binds collagen under high-flow conditions. Immunohistochemical analyses of hagfish tissues and blood revealed Vwf expression in endothelial cells and thrombocytes. Electron microscopic studies of hagfish tissues demonstrated the presence of Weibel-Palade bodies in the endothelium. Hagfish Vwf formed high-molecular-weight multimers in hagfish plasma and in stably transfected CHO cells. In functional assays, botrocetin promoted VWF-dependent thrombocyte aggregation. A search for vwf sequences in the genome of sea squirts, the closest invertebrate relatives of hagfish, failed to reveal evidence of an intact vwf gene. Together, our findings suggest that VWF evolved in the ancestral vertebrate following the divergence of the urochordates some 500 million years ago and that it acquired increasing complexity though sequential insertion of functional modules.
Collapse
|
42
|
Roumenina LT, Rayes J, Frimat M, Fremeaux-Bacchi V. Endothelial cells: source, barrier, and target of defensive mediators. Immunol Rev 2017; 274:307-329. [PMID: 27782324 DOI: 10.1111/imr.12479] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endothelium is strategically located at the interface between blood and interstitial tissues, placing thus endothelial cell as a key player in vascular homeostasis. Endothelial cells are in a dynamic equilibrium with their environment and constitute concomitantly a source, a barrier, and a target of defensive mediators. This review will discuss the recent advances in our understanding of the complex crosstalk between the endothelium, the complement system and the hemostasis in health and in disease. The first part will provide a general introduction on endothelial cells heterogeneity and on the physiologic role of the complement and hemostatic systems. The second part will analyze the interplay between complement, hemostasis and endothelial cells in physiological conditions and their alterations in diseases. Particular focus will be made on the prototypes of thrombotic microangiopathic disorders, resulting from complement or hemostasis dysregulation-mediated endothelial damage: atypical hemolytic uremic syndrome and thrombotic thrombocytopenic purpura. Novel aspects of the pathophysiology of the thrombotic microangiopathies will be discussed.
Collapse
Affiliation(s)
- Lubka T Roumenina
- INSERM UMRS 1138, Cordeliers Research Center, Université Pierre et Marie Curie (UPMC-Paris-6) and Université Paris Descartes Sorbonne Paris-Cité, Paris, France.
| | - Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Marie Frimat
- INSERM UMR 995, Lille, France.,Nephrology Department, CHU Lille, Lille, France
| | - Veronique Fremeaux-Bacchi
- INSERM UMRS 1138, Cordeliers Research Center, Université Pierre et Marie Curie (UPMC-Paris-6) and Université Paris Descartes Sorbonne Paris-Cité, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
43
|
Swystun LL, Georgescu I, Mewburn J, Deforest M, Nesbitt K, Hebert K, Dwyer C, Brown C, Notley C, Lillicrap D. Abnormal von Willebrand factor secretion, factor VIII stabilization and thrombus dynamics in type 2N von Willebrand disease mice. J Thromb Haemost 2017; 15:1607-1619. [PMID: 28581694 DOI: 10.1111/jth.13749] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Indexed: 12/20/2022]
Abstract
Essentials Type 2N von Willebrand disease involves impaired von Willebrand factor to factor VIII binding. Type 2N von Willebrand disease mutations exhibit qualitative and mild quantitative deficiencies. Type 2N von Willebrand disease mice exhibit unstable venous hemostatic thrombi. The factor VIII-binding ability of von Willebrand factor regulates arteriole thrombosis dynamics. SUMMARY Background von Willebrand factor (VWF) and factor VIII (FVIII) circulate as a non-covalent complex, with VWF serving as the carrier for FVIII. VWF indirectly influences secondary hemostasis by stabilizing FVIII and transporting it to the site of primary hemostasis. Type 2N von Willebrand disease involves impaired binding of VWF to FVIII, resulting in decreased plasma levels of FVIII. Objectives In these studies, we characterize the impact of three type 2N VWD variants (R763A, R854Q, R816W) on VWF secretion, FVIII stabilization and thrombus formation in a murine model. Methods Type 2N VWD mice were generated by hydrodynamic injections of mutant murine VWF cDNAs and the influence of these variants on VWF secretion and FVIII binding was evaluated. In vivo hemostasis and the dynamics of thrombus formation and embolization were assessed using a murine tail vein transection hemostasis model and an intravital thrombosis model in the cremaster arterioles. Results Type 2N VWD variants were associated with decreased VWF secretion using cell and animal-based models. FVIII-binding to type 2N variants was impaired in vitro and was variably stabilized in vivo by expressed or infused 2N variant VWF protein. Both transgenic type 2N VWD and FVIII knockout (KO) mice demonstrated impaired thrombus formation associated with decreased thrombus stability. Conclusions The type 2N VWD phenotype can be recapitulated in a murine model and is associated with both quantitative and qualitative VWF deficiencies and impaired thrombus formation. Patients with type 2N VWD may have normal primary hemostasis formation but decreased thrombus stability related to ineffective secondary hemostasis.
Collapse
Affiliation(s)
- L L Swystun
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - I Georgescu
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - J Mewburn
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - M Deforest
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - K Nesbitt
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - K Hebert
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - C Dwyer
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - C Brown
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - C Notley
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - D Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| |
Collapse
|
44
|
Hartholt RB, van Velzen AS, Peyron I, Ten Brinke A, Fijnvandraat K, Voorberg J. To serve and protect: The modulatory role of von Willebrand factor on factor VIII immunogenicity. Blood Rev 2017; 31:339-347. [PMID: 28716211 DOI: 10.1016/j.blre.2017.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 05/26/2017] [Accepted: 07/03/2017] [Indexed: 12/23/2022]
Abstract
Hemophilia A is a bleeding disorder characterized by the absence or dysfunction of blood coagulation factor VIII (FVIII). Patients are treated with regular infusions of FVIII concentrate. In response to treatment, approximately 30% of patients with severe hemophilia A develop inhibitory antibodies targeting FVIII. Both patient and treatment related risk factors for inhibitor development have been described. Multiple studies comparing the immunogenicity of recombinant and plasma-derived FVIII have yielded conflicting results. The randomized controlled SIPPET (Survey of Inhibitors in Plasma-Product Exposed Toddlers) trial demonstrated an increased risk of inhibitor development of recombinant FVIII when compared to von Willebrand factor (VWF)-containing plasma-derived FVIII. Presently, it is unclear which mechanism underlies the reduced immunogenicity of plasma-derived FVIII. In this review we address the potential role of VWF on FVIII immunogenicity and we discuss how VWF affects the immune recognition, processing and presentation of FVIII. We also briefly discuss the potential impact of glycan-composition on FVIII immunogenicity. It is well established that VWF shields the uptake of FVIII by antigen presenting cells. We have recently shown that VWF binds to the surface of dendritic cells. Here, we present a novel model in which surface bound FVIII-VWF complexes regulate the internalization of FVIII. Binding of FVIII to VWF is critically dependent on sulfation of Tyr1699 (HVGS numbering) in the light chain of FVIII. Incomplete sulfation of Tyr1699 has been suggested to occur in several recombinant FVIII products resulting in a loss of VWF binding. We hypothesize that this results in alternative pathways of FVIII internalization by antigen presenting cells which are not regulated by VWF. This hypothetical mechanism may explain the reduced immunogenicity of VWF containing plasma-derived FVIII concentrates as found in the SIPPET study.
Collapse
Affiliation(s)
- Robin B Hartholt
- Department of Plasma Proteins, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| | - Alice S van Velzen
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands.
| | - Ivan Peyron
- Department of Plasma Proteins, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| | - Anja Ten Brinke
- Department of Plasma Proteins, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| | - Karin Fijnvandraat
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands.
| | - Jan Voorberg
- Department of Plasma Proteins, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
45
|
Merlin S, Cannizzo ES, Borroni E, Bruscaggin V, Schinco P, Tulalamba W, Chuah MK, Arruda VR, VandenDriessche T, Prat M, Valente G, Follenzi A. A Novel Platform for Immune Tolerance Induction in Hemophilia A Mice. Mol Ther 2017; 25:1815-1830. [PMID: 28552407 DOI: 10.1016/j.ymthe.2017.04.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 12/14/2022] Open
Abstract
Hemophilia A (HA) is an X-linked bleeding disease caused by factor VIII (FVIII) deficiency. We previously demonstrated that FVIII is produced specifically in liver sinusoid endothelial cells (LSECs) and to some degree in myeloid cells, and thus, in the present work, we seek to restrict the expression of FVIII transgene to these cells using cell-specific promoters. With this approach, we aim to limit immune response in a mouse model by lentiviral vector (LV)-mediated gene therapy encoding FVIII. To increase the target specificity of FVIII expression, we included miRNA target sequences (miRTs) (i.e., miRT-142.3p, miRT-126, and miRT-122) to silence expression in hematopoietic cells, endothelial cells, and hepatocytes, respectively. Notably, we report, for the first time, therapeutic levels of FVIII transgene expression at its natural site of production, which occurred without the formation of neutralizing antibodies (inhibitors). Moreover, inhibitors were eradicated in FVIII pre-immune mice through a regulatory T cell-dependent mechanism. In conclusion, targeting FVIII expression to LSECs and myeloid cells by using LVs with cell-specific promoter minimized off-target expression and immune responses. Therefore, at least for some transgenes, expression at the physiologic site of synthesis can enhance efficacy and safety, resulting in long-term correction of genetic diseases such as HA.
Collapse
Affiliation(s)
- Simone Merlin
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | - Elvira Stefania Cannizzo
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | - Ester Borroni
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | - Valentina Bruscaggin
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | - Piercarla Schinco
- Azienda Ospedaliera Universitaria Città della Salute e della Scienza, 10126 Torino, Italy
| | - Warut Tulalamba
- Department of Gene Therapy & Regenerative Medicine, Free University of Brussels, 1050 Brussels, Belgium; Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Marinee K Chuah
- Department of Gene Therapy & Regenerative Medicine, Free University of Brussels, 1050 Brussels, Belgium; Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Valder R Arruda
- The Children's Hospital of Philadelphia, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thierry VandenDriessche
- Department of Gene Therapy & Regenerative Medicine, Free University of Brussels, 1050 Brussels, Belgium; Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Maria Prat
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | - Guido Valente
- Department of Translational Medicine, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy
| | - Antonia Follenzi
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", 28100 Novara, Italy.
| |
Collapse
|
46
|
Temperature effects on the activity, shape, and storage of platelets from 13-lined ground squirrels. J Comp Physiol B 2017; 187:815-825. [PMID: 28332020 DOI: 10.1007/s00360-017-1081-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/15/2016] [Accepted: 02/26/2017] [Indexed: 01/14/2023]
Abstract
The objective of this study is to determine how a hibernating mammal avoids the formation of blood clots under periods of low blood flow. A microfluidic vascular injury model was performed to differentiate the effects of temperature and shear rate on platelet adhesion to collagen. Human and ground squirrel whole blood was incubated at 15 or 37 °C and then passed through a microfluidic chamber over a 250-µm strip of type I fibrillar collagen at that temperature and the shear rates of 50 or 300 s-1 to simulate torpid and aroused conditions, respectively. At 15 °C, both human and ground squirrel platelets showed a 90-95% decrease in accumulation on collagen independent of shear rate. At 37 °C, human platelet accumulation reduced by 50% at 50 s-1 compared to 300 s-1, while ground squirrel platelet accumulation dropped by 80%. When compared to platelets from non-hibernating animals, platelets from animals collected after arousal from torpor showed a 60% decrease in binding at 37 °C and 300 s-1, but a 2.5-fold increase in binding at 15 °C and 50 s-1. vWF binding in platelets from hibernating ground squirrels was decreased by 50% relative to non-hibernating platelets. The source of the plasma that platelets were stored in did not affect the results indicating that the decreased vWF binding was a property of the platelets. Upon chilling, ground squirrel platelets increase microtubule assembly leading to the formation of long rods. This shape change is concurrent with sequestration of platelets in the liver and not the spleen. In conclusion, it appears that ground squirrel platelets are sequestered in the liver during torpor and have reduced binding capacity for plasma vWF and lower accumulation on collagen at low shear rates and after storage at cold temperatures, while still being activated by external agonists. These adaptations would protect the animals from spontaneous thrombus formation during torpor but allow them to restore normal platelet function upon arousal.
Collapse
|
47
|
Jacquemin M, Vodolazkaia A, Toelen J, Schoeters J, Van Horenbeeck I, Vanlinthout I, Debasse M, Peerlinck K. Measurement of B-domain-deleted ReFacto AF activity with a product-specific standard is affected by choice of reagent and patient-specific factors. Haemophilia 2017; 24:675-682. [DOI: 10.1111/hae.13123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2016] [Indexed: 12/01/2022]
Affiliation(s)
- M. Jacquemin
- Center for Molecular and Vascular Biology; University of Leuven; Leuven Belgium
- Laboratorium Geneeskunde; UZ Leuven; Leuven Belgium
| | | | - J. Toelen
- Laboratorium Geneeskunde; UZ Leuven; Leuven Belgium
| | - J. Schoeters
- Laboratorium Geneeskunde; UZ Leuven; Leuven Belgium
| | | | | | - M. Debasse
- Laboratorium Geneeskunde; UZ Leuven; Leuven Belgium
| | - K. Peerlinck
- Center for Molecular and Vascular Biology; University of Leuven; Leuven Belgium
- Vascular Medicine and Haemostasis; UZ Leuven; Leuven Belgium
| |
Collapse
|
48
|
Nguyen GN, George LA, Siner JI, Davidson RJ, Zander CB, Zheng XL, Arruda VR, Camire RM, Sabatino DE. Novel factor VIII variants with a modified furin cleavage site improve the efficacy of gene therapy for hemophilia A. J Thromb Haemost 2017; 15:110-121. [PMID: 27749002 PMCID: PMC5280213 DOI: 10.1111/jth.13543] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Indexed: 12/26/2022]
Abstract
Essentials Factor (F) VIII is an inefficiently expressed protein. Furin deletion FVIII variants were purified and characterized using in vitro and in vivo assays. These minimally modified novel FVIII variants have enhanced function. These variants provide a strategy for increasing FVIII expression in hemophilia A gene therapy. SUMMARY Background The major challenge for developing gene-based therapies for hemophilia A is that human factor VIII (hFVIII) has intrinsic properties that result in inefficient biosynthesis. During intracellular processing, hFVIII is predominantly cleaved at a paired basic amino acid cleaving enzyme (PACE) or furin cleavage site to yield a heterodimer that is the major form of secreted protein. Previous studies with B-domain-deleted (BDD) canine FVIII and hFVIII-R1645H, both differing from hFVIII by a single amino acid at this site, suggested that these proteins are secreted mainly in a single polypeptide chain (SC) form and exhibit enhanced function. Objective We hypothesized that deletion(s) of the furin site modulates FVIII biology and may enhance its function. Methods A series of recombinant hFVIII-furin deletion variants were introduced into hFVIII-BDD [Δ1645, 1645-46(Δ2), 1645-47(Δ3), 1645-48(Δ4), or Δ1648] and characterized. Results In vitro, recombinant purified Δ3 and Δ4 were primarily SC and, interestingly, had 2-fold higher procoagulant activity compared with FVIII-BDD. In vivo, the variants also have improved hemostatic function. After adeno-associated viral (AAV) vector delivery, the expression of these variants is 2-4-fold higher than hFVIII-BDD. Protein challenges of each variant in mice tolerant to hFVIII-BDD showed no anti-FVIII immune response. Conclusions These data suggest that the furin deletion hFVIII variants are superior to hFVIII-BDD without increased immunogenicity. In the setting of gene-based therapeutics, these novel variants provide a unique strategy to increase FVIII expression, thus lowering the vector dose, a critical factor for hemophilia A gene therapy.
Collapse
Affiliation(s)
- G. N. Nguyen
- The Raymond G. Perelman Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - L. A. George
- The Raymond G. Perelman Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of PediatricsDivision of HematologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - J. I. Siner
- The Raymond G. Perelman Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - R. J. Davidson
- The Raymond G. Perelman Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - C. B. Zander
- Division of Laboratory MedicineDepartment of PathologyUniversity of Alabama at BirminghamBirminghamALUSA
| | - X. L. Zheng
- Division of Laboratory MedicineDepartment of PathologyUniversity of Alabama at BirminghamBirminghamALUSA
| | - V. R. Arruda
- The Raymond G. Perelman Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of PediatricsDivision of HematologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - R. M. Camire
- The Raymond G. Perelman Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of PediatricsDivision of HematologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - D. E. Sabatino
- The Raymond G. Perelman Center for Cellular and Molecular TherapeuticsThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of PediatricsDivision of HematologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| |
Collapse
|
49
|
Miesbach W, Berntorp E. Von Willebrand disease - the ‘Dos’ and ‘Don'ts’ in surgery. Eur J Haematol 2016; 98:121-127. [DOI: 10.1111/ejh.12809] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Wolfgang Miesbach
- Haemophilia Centre; Medical Clinic II; Institute of Transfusion Medicine; Goethe University Hospital; Frankfurt am Main Germany
| | - Erik Berntorp
- Centre for Thrombosis and Haemostasis; Skane University Hospital; Lund University; Malmö Sweden
| |
Collapse
|
50
|
Von Willebrand factor is reversibly decreased during torpor in 13-lined ground squirrels. J Comp Physiol B 2016; 186:131-9. [PMID: 26481634 DOI: 10.1007/s00360-015-0941-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 10/01/2015] [Accepted: 10/11/2015] [Indexed: 12/28/2022]
Abstract
During torpor in a hibernating mammal, decreased blood flow increases the risk of blood clots such as deep vein thrombi (DVT). In other animal models platelets, neutrophils, monocytes and von Willebrand factor (VWF) have been found in DVT. Previous research has shown that hibernating mammals decrease their levels of platelets and clotting factors VIII (FVIII) and IX (FIX), increasing both bleeding time and activated partial thromboplastin time. In this study, FVIII, FIX and VWF activities and mRNA levels were measured in torpid and non-hibernating ground squirrels (Ictidomys tridecemlineatus). Here, we show that VWF high molecular weight multimers, collagen-binding activity, lung mRNA and promoter activity decrease during torpor. The VWF multimers reappear in plasma within 2 h of arousal in the spring. Similarly, FIX activity and liver mRNA both dropped threefold during torpor. In contrast, FVIII liver mRNA levels increased twofold while its activity dropped threefold, consistent with a post-transcriptional decrease in FVIII stability in the plasma due to decreased VWF levels. Finally, both neutrophils and monocytes are decreased eightfold during torpor which could slow the formation of DVT. In addition to providing insight in how blood clotting can be regulated to allow mammals to survive in extreme environments, hibernating ground squirrels provide an interesting model for studying.
Collapse
|