1
|
Ku CJ, Yu X, Zhao QY, Grzegorski SJ, Daniel JG, Ferguson AC, Shavit JA. Loss of protein C vs protein S results in discrepant thrombotic phenotypes. Blood Adv 2025; 9:545-557. [PMID: 39657127 PMCID: PMC11821410 DOI: 10.1182/bloodadvances.2024013237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/25/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
ABSTRACT Venous thrombosis is a leading cause of morbidity/mortality and associated with deficiencies of the anticoagulant protein C (PC; PROC) and its cofactor, protein S (PS; PROS1). Heterozygous mutations increase the risk of adult-onset thrombosis, whereas homozygous mutations result in pre/neonatal lethal thrombosis. Phenotypes of patients with PC and PS deficiency are generally considered clinically indistinguishable. Here, we generate proc (zebrafish PROC ortholog) and pros1 knockouts through genome editing in zebrafish and uncover partially discordant phenotypes. proc-/- mutants exhibited ∼70% lethality at 1 year of age, whereas pros1-/- survival was unaffected. Induced venous endothelial injury in both mutants revealed reduced occlusive thrombus formation. This is consistent with the consumptive coagulopathy of zebrafish antithrombin 3 knockouts, which also results in spontaneous venous thrombosis. However, proc and pros1 mutants revealed a discrepancy. Although both mutants demonstrated spontaneous thrombosis, proc-/- was localized to the cardiac and venous systems, whereas pros1-/- was intracardiac. Aside from coagulation, PC has been shown to have PS-independent roles in inflammation. proc mutants displayed altered inflammatory markers and defects in neutrophil migration independent of pros1. Transcriptomic analysis and gene knockdown identified novel proc genetic interactions with adgrf7, a G protein-coupled receptor (GPCR) not previously known to be involved in coagulation. In summary, our data reveal differences between PC- and PS-deficient thrombosis, with cardiovascular tissue-specific phenotypes and survival differences, suggesting the possibility of underlying clinical differences in affected patients. This model of complete proc-/- deficiency in an accessible organism will facilitate further in vivo study of these distinctions, as well as PS-dependent and -independent functions of PC.
Collapse
Affiliation(s)
- Chia-Jui Ku
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Xinge Yu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Queena Y. Zhao
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | | | | | | | - Jordan A. Shavit
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| |
Collapse
|
2
|
Yu X, Zhao QY, Yaman M, Emly SM, Lee JK, Su H, Ferguson AC, Nagaswami C, Chaturantabut S, Goessling W, Weisel JW, Auchus RJ, Shavit JA. Hormone-induced thrombosis is mediated through non-canonical fibrin(ogen) aggregation and a novel estrogen target in zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623199. [PMID: 39605542 PMCID: PMC11601434 DOI: 10.1101/2024.11.13.623199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Venous thrombosis is a well-known complication of sex hormone therapy, with onset typically within weeks to months after initiation. Worldwide, more than 100 million pre-menopausal women use combined oral contraceptives, with tens to hundreds of thousands developing thrombosis annually, resulting in significant morbidity and mortality. Although it is known that estrogens can alter expression of coagulation factors, the pathways and mechanisms that connect the two systems, as well as the proteins involved in progression to thrombosis, are poorly understood. Identification of these mediators are central to any comprehensive understanding of hormone-induced pathophysiology, could help ascertain patients at higher risk for thrombosis, and may also pinpoint future therapeutic targets. The zebrafish is a powerful genetic model in which the hemostatic system is almost entirely conserved with humans. Its external development, ability to generate thousands of offspring at low cost, and optical transparency all make it a powerful tool to study the genetics of coagulation disorders. We previously produced a transgenic line (fgb-egfp) that generates GFP-tagged fibrinogen that labels induced and spontaneous fibrin-rich thrombi. Here we show rapid onset of thrombosis after exposure to various estrogens, but not progestins or testosterone. Thrombi are localized to the venous system, develop broadly along the posterior cardinal vein, and show evidence for clot contraction. Thrombosis is only partially impeded by anticoagulants, occurs in the absence of factor X and prothrombin, but is completely blocked in the absence of fibrinogen. Furthermore, although an estrogen receptor antagonist is partially inhibitory, targeted knockout of all known estrogen receptors does not eliminate thrombosis. These data suggest that zebrafish can be used to model human estrogen-induced thrombosis, although the lack of dependence on the canonical coagulation cascade is surprising. The inability to completely inhibit thrombosis through genetic/pharmacologic anticoagulation or estrogen receptor disruption suggests that the mechanisms may be multifactorial. We hypothesize that thrombi are composed of fibrin(ogen) aggregates rather than purely fibrin. Results of further studies could lead to novel therapeutic targets and ascertain patients at higher risk for thrombosis.
Collapse
Affiliation(s)
- Xinge Yu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Queena Y. Zhao
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Murat Yaman
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Sylvia M. Emly
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | | | - Hongyu Su
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| | | | - Chandrasekaran Nagaswami
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA
| | | | - Wolfram Goessling
- Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Division of Health Sciences and Technology, Harvard-MIT, Cambridge, MA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA
| | - John W. Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Richard J. Auchus
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Jordan A. Shavit
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| |
Collapse
|
3
|
Ranjan G, Sehgal P, Scaria V, Sivasubbu S. SCAR-6 elncRNA locus epigenetically regulates PROZ and modulates coagulation and vascular function. EMBO Rep 2024; 25:4950-4978. [PMID: 39358551 PMCID: PMC11549340 DOI: 10.1038/s44319-024-00272-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/03/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
In this study, we characterize a novel lncRNA-producing gene locus that we name Syntenic Cardiovascular Conserved Region-Associated lncRNA-6 (scar-6) and functionally validate its role in coagulation and cardiovascular function. A 12-bp deletion of the scar-6 locus in zebrafish (scar-6gib007Δ12/Δ12) results in cranial hemorrhage and vascular permeability. Overexpression, knockdown and rescue with the scar-6 lncRNA modulates hemostasis in zebrafish. Molecular investigation reveals that the scar-6 lncRNA acts as an enhancer lncRNA (elncRNA), and controls the expression of prozb, an inhibitor of factor Xa, through an enhancer element in the scar-6 locus. The scar-6 locus suppresses loop formation between prozb and scar-6 sequences, which might be facilitated by the methylation of CpG islands via the prdm14-PRC2 complex whose binding to the locus might be stabilized by the scar-6 elncRNA transcript. Binding of prdm14 to the scar-6 locus is impaired in scar-6gib007Δ12/Δ12 zebrafish. Finally, activation of the PAR2 receptor in scar-6gib007Δ12/Δ12 zebrafish triggers NF-κB-mediated endothelial cell activation, leading to vascular dysfunction and hemorrhage. We present evidence that the scar-6 locus plays a role in regulating the expression of the coagulation cascade gene prozb and maintains vascular homeostasis.
Collapse
Affiliation(s)
- Gyan Ranjan
- CSIR Institute of Genomics and Integrative Biology, Mathura Road, Delhi, 110024, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Paras Sehgal
- CSIR Institute of Genomics and Integrative Biology, Mathura Road, Delhi, 110024, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vinod Scaria
- CSIR Institute of Genomics and Integrative Biology, Mathura Road, Delhi, 110024, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
- Vishwanath Cancer Care Foundation, Mumbai, India.
- Dr. D. Y Patil Medical College, Hospital and Research Centre, Pune, India.
| | - Sridhar Sivasubbu
- CSIR Institute of Genomics and Integrative Biology, Mathura Road, Delhi, 110024, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
- Vishwanath Cancer Care Foundation, Mumbai, India.
- Dr. D. Y Patil Medical College, Hospital and Research Centre, Pune, India.
| |
Collapse
|
4
|
Pontius MHH, Ku CJ, Osmond MJ, Disharoon D, Liu Y, Warnock M, Lawrence DA, Marr DWM, Neeves KB, Shavit JA. Magnetically powered microwheel thrombolysis of occlusive thrombi in zebrafish. Proc Natl Acad Sci U S A 2024; 121:e2315083121. [PMID: 38408253 PMCID: PMC10927521 DOI: 10.1073/pnas.2315083121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/09/2024] [Indexed: 02/28/2024] Open
Abstract
Tissue plasminogen activator (tPA) is the only FDA-approved treatment for ischemic stroke but carries significant risks, including major hemorrhage. Additional options are needed, especially in small vessel thrombi which account for ~25% of ischemic strokes. We have previously shown that tPA-functionalized colloidal microparticles can be assembled into microwheels (µwheels) and manipulated under the control of applied magnetic fields to enable rapid thrombolysis of fibrin gels in microfluidic models of thrombosis. Transparent zebrafish larvae have a highly conserved coagulation cascade that enables studies of hemostasis and thrombosis in the context of intact vasculature, clotting factors, and blood cells. Here, we show that tPA-functionalized µwheels can perform rapid and targeted recanalization in vivo. This effect requires both tPA and µwheels, as minimal to no recanalization is achieved with tPA alone, µwheels alone, or tPA-functionalized microparticles in the absence of a magnetic field. We evaluated tPA-functionalized µwheels in CRISPR-generated plasminogen (plg) heterozygous and homozygous mutants and confirmed that tPA-functionalized µwheels are dose-dependent on plasminogen for lysis. We have found that magnetically powered µwheels as a targeted tPA delivery system are dramatically more efficient at plasmin-mediated thrombolysis than systemic delivery in vivo. Further development of this system in fish and mammalian models could enable a less invasive strategy for alleviating ischemia that is safer than directed thrombectomy or systemic infusion of tPA.
Collapse
Affiliation(s)
| | - Chia-Jui Ku
- Department of Pediatrics, University of Michigan, Ann Arbor, MI48109
| | - Matthew J. Osmond
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO80401
| | - Dante Disharoon
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO80401
| | - Yang Liu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI48109
| | - Mark Warnock
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI48109
| | - Daniel A. Lawrence
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI48109
| | - David W. M. Marr
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO80401
| | - Keith B. Neeves
- Department of Bioengineering, University of Colorado, Denver, Aurora, CO80045
- Department of Pediatrics, University of Colorado, Denver, Aurora, CO80045
| | - Jordan A. Shavit
- Department of Pediatrics, University of Michigan, Ann Arbor, MI48109
- Department of Human Genetics, University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
5
|
Richter CE, Raghunath A, Griffin MS, Yaman M, Arruda VR, Samelson-Jones BJ, Shavit JA. Loss of factor VIII in zebrafish rebalances antithrombin deficiency but has a limited bleeding diathesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582609. [PMID: 39896458 PMCID: PMC11785011 DOI: 10.1101/2024.02.28.582609] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Deficiencies in coagulation factor VIII (FVIII, F8) result in the bleeding disorder hemophilia A. An emerging novel therapeutic strategy for bleeding disorders is to enhance hemostasis by limiting natural anticoagulants, such as antithrombin (AT3). To study pro/anticoagulant hemostatic balance in an in vivo model, we used genome editing to create null alleles for f8 and von Willebrand factor (vwf) in zebrafish, a model organism with a high degree of homology to the mammalian hemostatic system and unique attributes, including external development and optical transparency. f8 homozygous mutant larvae surprisingly formed normal thrombi when subjected to laser-mediated endothelial injury, had no overt signs of hemorrhage, but had a modest increase in mortality. We have previously shown that at3 -/- larvae develop disseminated intravascular coagulation (DIC), with spontaneous thrombosis and fibrinogen consumption, resulting in bleeding phenotype marked by secondary lack of induced thrombus formation upon endothelial injury. We found that with loss of FVIII (f8 -/-;at3 -/-), larvae no longer developed spontaneous fibrin thrombi and did produce clots in response to endothelial injury. However, homozygous loss of zebrafish Vwf failed to rescue the at3 DIC phenotype. These studies demonstrate an altered balance of natural anticoagulants that mitigates FVIII deficiency in zebrafish, similar to human clinical pipeline products. The data also suggest that zebrafish FVIII might circulate independently of Vwf. Further study of this unique balance could provide new insights for management of hemophilia A and von Willebrand disease.
Collapse
Affiliation(s)
- Catherine E. Richter
- Department of Pediatrics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Azhwar Raghunath
- Department of Pediatrics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Megan S. Griffin
- Department of Pediatrics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Murat Yaman
- Department of Pediatrics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Valder R. Arruda
- Division of Hematology and Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Benjamin J. Samelson-Jones
- Division of Hematology and Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jordan A. Shavit
- Department of Pediatrics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
- Department of Human Genetics, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| |
Collapse
|
6
|
Pontius MHH, Ku CJ, Osmond M, Disharoon D, Liu Y, Marr DW, Neeves KB, Shavit JA. Magnetically Powered Microwheel Thrombolysis of Occlusive Thrombi in Zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.11.557256. [PMID: 37745422 PMCID: PMC10515822 DOI: 10.1101/2023.09.11.557256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Tissue plasminogen activator (tPA) is the only FDA approved treatment for ischemic stroke but carries significant risks, including major hemorrhage. Additional options are needed, especially in small vessel thrombi which account for ~25% of ischemic strokes. We have previously shown that tPA-functionalized colloidal microparticles can be assembled into microwheels (µwheels) and manipulated under the control of applied magnetic fields to enable rapid thrombolysis of fibrin gels in microfluidic models of thrombosis. Providing a living microfluidic analog, transparent zebrafish larvae have a highly conserved coagulation cascade that enables studies of hemostasis and thrombosis in the context of intact vasculature, clotting factors, and blood cells. Here we show that tPA-functionalized µwheels can perform rapid and targeted recanalization in vivo. This effect requires both tPA and µwheels, as minimal to no recanalization is achieved with tPA alone, µwheels alone, or tPA-functionalized microparticles in the absence of a magnetic field. We evaluated tPA-µwheels in CRISPR-generated plasminogen (plg) heterozygous and homozygous mutants and confirmed that tPA-µwheels are dose-dependent on plasminogen for lysis. We have found that magnetically powered µwheels as a targeted tPA delivery system are dramatically more efficient at plasmin-mediated thrombolysis than systemic delivery in vivo. Further development of this system in fish and mammalian models could enable a less invasive strategy for alleviating ischemia that is safer than directed thrombectomy or systemic infusion of tPA.
Collapse
Affiliation(s)
| | - Chia-Jui Ku
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Matthew Osmond
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO
| | - Dante Disharoon
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO
| | - Yang Liu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - David W.M. Marr
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO
| | - Keith B. Neeves
- Departments of Bioengineering and Pediatrics, University of Colorado, Denver | Anschutz Medical Campus, Aurora, CO
| | - Jordan A. Shavit
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| |
Collapse
|
7
|
Wang HL, Ruan DD, Wu M, Ji YY, Hu XX, Wu QY, Zhang YP, Lin B, Hu YN, Wang H, Tang Y, Fang ZT, Luo JW, Liao LS, Gao MZ. Identification and characterization of two SERPINC1 mutations causing congenital antithrombin deficiency. Thromb J 2023; 21:3. [PMID: 36624481 PMCID: PMC9830717 DOI: 10.1186/s12959-022-00443-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Antithrombin (AT) is the main physiological anticoagulant involved in hemostasis. Hereditary AT deficiency is a rare autosomal dominant thrombotic disease mainly caused by mutations in SERPINC1, which was usually manifested as venous thrombosis and pulmonary embolism. In this study, we analyzed the clinical characteristics and screened for mutant genes in two pedigrees with hereditary AT deficiency, and the functional effects of the pathogenic mutations were evaluated. METHODS Candidate gene variants were analyzed by next-generation sequencing to screen pathogenic mutations in probands, followed by segregation analysis in families by Sanger sequencing. Mutant and wild-type plasmids were constructed and transfected into HEK293T cells to observe protein expression and cellular localization of SERPINC1. The structure and function of the mutations were analyzed by bioinformatic analyses. RESULTS The proband of pedigree A with AT deficiency carried a heterozygous frameshift mutation c.1377delC (p.Asn460Thrfs*20) in SERPINC1 (NM000488.3), a 1377C base deletion in exon 7 resulting in a backward shift of the open reading frame, with termination after translation of 20 residues, and a different residue sequence translated after the frameshift. Bioinformatics analysis suggests that the missing amino acid sequence caused by the frameshift mutation might disrupt the disulfide bond between Cys279 and Cys462 and affect the structural function of the protein. This newly discovered variant is not currently included in the ClinVar and HGMD databases. p.Arg229* resulted in a premature stop codon in exon 4, and bioinformatics analysis suggests that the truncated protein structure lost its domain of interaction with factor IX (Ala414 site) after the deletion of nonsense mutations. However, considering the AT truncation protein resulting from the p.Arg229* variant loss a great proportion of the molecule, we speculate the variant may affect two functional domains HBS and RCL and lack of the corresponding function. The thrombophilia and decreased-AT-activity phenotypes of the two pedigrees were separated from their genetic variants. After lentiviral plasmid transfection into HEK293T cells, the expression level of AT protein decreased in the constructed c.1377delC mutant cells compared to that in the wild-type, which was not only reduced in c.685C > T mutant cells but also showed a significant band at 35 kDa, suggesting a truncated protein. Immunofluorescence localization showed no significant differences in protein localization before and after the mutation. CONCLUSIONS The p.Asn460Thrfs*20 and p.Arg229* variants of SERPINC1 were responsible for the two hereditary AT deficiency pedigrees, which led to AT deficiency by different mechanisms. The p.Asn460Thrfs*20 variant is reported for the first time.
Collapse
Affiliation(s)
- Han-lu Wang
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China
| | - Dan-dan Ruan
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China
| | - Min Wu
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China
| | - Yuan-yuan Ji
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China
| | - Xing-xing Hu
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China ,grid.415108.90000 0004 1757 9178Department of Cardiovascular Medicine, Fujian Provincial Hospital, Fuzhou, 350001 China
| | - Qiu-yan Wu
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China
| | - Yan-ping Zhang
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China
| | - Bin Lin
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China
| | - Ya-nan Hu
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China
| | - Hang Wang
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China ,grid.415108.90000 0004 1757 9178Department of Cardiovascular Surgery, Fujian Provincial Hospital, Fuzhou, 350001 China
| | - Yi Tang
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China ,grid.415108.90000 0004 1757 9178Department of Interventional Radiology, Fujian Provincial Hospital, Fuzhou, 350001 China
| | - Zhu-ting Fang
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China ,grid.415108.90000 0004 1757 9178Department of Interventional Radiology, Fujian Provincial Hospital, Fuzhou, 350001 China
| | - Jie-wei Luo
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China ,grid.415108.90000 0004 1757 9178Department of Traditional Chinese Medicine, Fujian Provincial Hospital, Fuzhou, 350001 China
| | - Li-sheng Liao
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China ,grid.415108.90000 0004 1757 9178Department of Hematology, Fujian Provincial Hospital, Fuzhou, 350001 China
| | - Mei-zhu Gao
- grid.415108.90000 0004 1757 9178Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001 China ,grid.415108.90000 0004 1757 9178Department of Nephrology, Fujian Provincial Hospital, Fuzhou, 350001 China
| |
Collapse
|
8
|
Lopes TJS, Rios RA, Rios TN, Alencar BM, Ferreira MV, Morishita E. Computational analyses reveal fundamental properties of the AT structure related to thrombosis. BIOINFORMATICS ADVANCES 2022; 3:vbac098. [PMID: 36698764 PMCID: PMC9838315 DOI: 10.1093/bioadv/vbac098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/28/2022] [Accepted: 12/22/2022] [Indexed: 12/25/2022]
Abstract
Summary Blood coagulation is a vital process for humans and other species. Following an injury to a blood vessel, a cascade of molecular signals is transmitted, inhibiting and activating more than a dozen coagulation factors and resulting in the formation of a fibrin clot that ceases the bleeding. In this process, antithrombin (AT), encoded by the SERPINC1 gene is a key player regulating the clotting activity and ensuring that it stops at the right time. In this sense, mutations to this factor often result in thrombosis-the excessive coagulation that leads to the potentially fatal formation of blood clots that obstruct veins. Although this process is well known, it is still unclear why even single residue substitutions to AT lead to drastically different phenotypes. In this study, to understand the effect of mutations throughout the AT structure, we created a detailed network map of this protein, where each node is an amino acid, and two amino acids are connected if they are in close proximity in the three-dimensional structure. With this simple and intuitive representation and a machine-learning framework trained using genetic information from more than 130 patients, we found that different types of thrombosis have emerging patterns that are readily identifiable. Together, these results demonstrate how clinical features, genetic data and in silico analysis are converging to enhance the diagnosis and treatment of coagulation disorders. Supplementary information Supplementary data are available at Bioinformatics Advances online.
Collapse
Affiliation(s)
| | - Ricardo A Rios
- Institute of Computing, Federal University of Bahia, Salvador 40170-110, Brazil
| | - Tatiane N Rios
- Institute of Computing, Federal University of Bahia, Salvador 40170-110, Brazil
| | - Brenno M Alencar
- Institute of Computing, Federal University of Bahia, Salvador 40170-110, Brazil
| | - Marcos V Ferreira
- Institute of Computing, Federal University of Bahia, Salvador 40170-110, Brazil
| | | |
Collapse
|
9
|
Genetic duplication of tissue factor reveals subfunctionalization in venous and arterial hemostasis. PLoS Genet 2022; 18:e1010534. [PMID: 36449521 PMCID: PMC9744294 DOI: 10.1371/journal.pgen.1010534] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 12/12/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022] Open
Abstract
Tissue factor (TF) is an evolutionarily conserved protein necessary for initiation of hemostasis. Zebrafish have two copies of the tissue factor gene (f3a and f3b) as the result of an ancestral teleost fish duplication event (so called ohnologs). In vivo physiologic studies of TF function have been difficult given early lethality of TF knockout in the mouse. We used genome editing to produce knockouts of both f3a and f3b in zebrafish. Since ohnologs arose through sub- or neofunctionalization, they can unmask unknown functions of non-teleost genes and could reveal whether mammalian TF has developmental functions distinct from coagulation. Here we show that a single copy of either f3a or f3b is necessary and sufficient for normal lifespan. Complete loss of TF results in lethal hemorrhage by 2-4 months despite normal embryonic and vascular development. Larval vascular endothelial injury reveals predominant roles for TFa in venous circulation and TFb in arterial circulation. Finally, we demonstrate that loss of TF predisposes to a stress-induced cardiac tamponade independent of its role in fibrin formation. Overall, our data suggest partial subfunctionalization of TFa and TFb. This multigenic zebrafish model has the potential to facilitate study of the role of TF in different vascular beds.
Collapse
|
10
|
Raghunath A, Ferguson AC, Shavit JA. Fishing for answers to hemostatic and thrombotic disease: Genome editing in zebrafish. Res Pract Thromb Haemost 2022; 6:e12759. [PMID: 35949884 PMCID: PMC9354590 DOI: 10.1002/rth2.12759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/27/2022] [Accepted: 06/15/2022] [Indexed: 01/22/2023] Open
Abstract
Over the past two decades, the teleost vertebrate Danio rerio (zebrafish) has emerged as a model for hemostasis and thrombosis. At genomic and functional levels, there is a high degree of conservation of the hemostatic system with that of mammals. Numerous features of the fish model offer unique advantages for investigating hemostasis and thrombosis. These include high fecundity, rapid and external development, optical transparency, and extensive functional homology with mammalian hemostasis and thrombosis. Zebrafish are particularly suited to genome-wide mutagenesis experiments for the study of modifier genes. They are also amenable to whole-organism small-molecule screens, a feature that is exceptionally relevant to hemostasis and thrombosis. Zebrafish coagulation factor knockouts that are in utero or neonatal lethal in mammals survive into adulthood before succumbing to hemorrhage or thrombosis, enabling studies not possible in mammals. In this illustrated review, we outline how zebrafish have been employed for the study of hemostasis and thrombosis using modern genome editing techniques, coagulation assays in larvae, and in vivo evaluation of patient-specific variants to infer causality and demonstrate pathogenicity. Zebrafish hemostasis and thrombosis models will continue to serve as a clinically directed basic research tool and powerful alternative to mammals for the development of new diagnostic markers and novel therapeutics for coagulation disorders through high-throughput genetic and small-molecule studies.
Collapse
Affiliation(s)
- Azhwar Raghunath
- Department of PediatricsUniversity of Michigan School of MedicineAnn ArborMichiganUSA
| | - Allison C. Ferguson
- Department of PediatricsUniversity of Michigan School of MedicineAnn ArborMichiganUSA
| | - Jordan A. Shavit
- Department of PediatricsUniversity of Michigan School of MedicineAnn ArborMichiganUSA
- Department of Human GeneticsUniversity of Michigan School of MedicineAnn ArborMichiganUSA
| |
Collapse
|
11
|
Grover SP, Mackman N. Anticoagulant SERPINs: Endogenous Regulators of Hemostasis and Thrombosis. Front Cardiovasc Med 2022; 9:878199. [PMID: 35592395 PMCID: PMC9110684 DOI: 10.3389/fcvm.2022.878199] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/29/2022] [Indexed: 12/17/2022] Open
Abstract
Appropriate activation of coagulation requires a balance between procoagulant and anticoagulant proteins in blood. Loss in this balance leads to hemorrhage and thrombosis. A number of endogenous anticoagulant proteins, such as antithrombin and heparin cofactor II, are members of the serine protease inhibitor (SERPIN) family. These SERPIN anticoagulants function by forming irreversible inhibitory complexes with target coagulation proteases. Mutations in SERPIN family members, such as antithrombin, can cause hereditary thrombophilias. In addition, low plasma levels of SERPINs have been associated with an increased risk of thrombosis. Here, we review the biological activities of the different anticoagulant SERPINs. We further consider the clinical consequences of SERPIN deficiencies and insights gained from preclinical disease models. Finally, we discuss the potential utility of engineered SERPINs as novel therapies for the treatment of thrombotic pathologies.
Collapse
|
12
|
Tran VLT, Hortle E, Britton WJ, Oehlers SH. Common anti-haemostatic medications increase the severity of systemic infection by uropathogenic Escherichia coli. Microbiol Res 2021; 254:126918. [PMID: 34798538 DOI: 10.1016/j.micres.2021.126918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/16/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022]
Abstract
Uropathogenic Escherichia coli (UPEC) causes urinary tract infections that can result in sepsis. The haemostatic system is protective in the pyelonephritis stage of ascending UPEC infection, but the role of the haemostatic system has not been investigated during sepsis. Here we utilize a zebrafish-UPEC systemic infection model to visualize infection-induced coagulation and examine the effects of commonly prescribed anti-haemostatic medications on the infection severity. Treatment of systemically infected zebrafish with warfarin, aspirin, or ticagrelor reduced host survival, while stabilization of clots with aminocaproic acid increased host survival. Anti-haemostatic drug treatment increased UPEC burden. Our findings provide evidence that commonly prescribed anti-haemostatic medications may worsen the outcome of severe UPEC infection.
Collapse
Affiliation(s)
- Vi L T Tran
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Elinor Hortle
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia; The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, NSW, Australia.
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia; Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050 Australia
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia; The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, NSW, Australia.
| |
Collapse
|
13
|
de la Morena-Barrio ME, Gindele R, Bravo-Pérez C, Ilonczai P, Zuazu I, Speker M, Oláh Z, Rodríguez-Sevilla JJ, Entrena L, Infante MS, de la Morena-Barrio B, García JM, Schlammadinger Á, Cifuentes-Riquelme R, Mora-Casado A, Miñano A, Padilla J, Vicente V, Corral J, Bereczky Z. High penetrance of inferior vena cava system atresia in severe thrombophilia caused by homozygous antithrombin Budapest 3 variant: Description of a new syndrome. Am J Hematol 2021; 96:1363-1373. [PMID: 34324211 DOI: 10.1002/ajh.26304] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 11/07/2022]
Abstract
Atresia of inferior vena cava (IVC) is a rare congenital malformation associated with high risk of venous thrombosis that still has unknown etiology, although intrauterine IVC thrombosis has been suggested to be involved. The identification of IVC atresia in a case with early idiopathic venous thrombosis and antithrombin deficiency caused by the homozygous SERPINC1 c.391C > T variant (p.Leu131Phe; antithrombin Budapest 3) encouraged us to evaluate the role of this severe thrombophilia in this vascular abnormality. We have done a cross-sectional study in previously identified cohorts of patients homozygous for the Budapest 3 variant (N = 61) selected from 1118 patients with congenital antithrombin deficiency identified in two different populations: Spain (N = 692) and Hungary (N = 426). Image analysis included computed tomography and phlebography. Atresia of the IVC system was observed in 17/24 cases (70.8%, 95% confidence interval [CI]: 48.9%-87.3%) homozygous for antithrombin Budapest 3 with available computed tomography (5/8 and 12/16 in the Spanish and Hungarian cohorts, respectively), 16 had an absence of infrarenal IVC and one had atresia of the left common iliac vein. All cases with vascular defects had compensatory mechanisms, azygos-hemiazygos continuation or double IVC, and seven also had other congenital anomalies. Short tandem repeat analysis supported the specific association of the IVC system atresia with SERPINC1. We show the first evidence of the association of a severe thrombophilia with IVC system atresia, supporting the possibility that a thrombosis in the developing fetal vessels is the reason for this anomaly. Our hypothesis-generating results encourage further studies to investigate severe thrombophilic states in patients with atresia of IVC.
Collapse
Affiliation(s)
- María E de la Morena-Barrio
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - Réka Gindele
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Carlos Bravo-Pérez
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - Péter Ilonczai
- Jósa Teaching Hospital of University of Debrecen, Nyíregyháza, Hungary
| | - Isabel Zuazu
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - Marianna Speker
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Oláh
- Department of Anaesthesiology and Intensive Care, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Laura Entrena
- Servicio de Hematología, Hospital Virgen de las Nieves, Granada, Spain
| | - Maria S Infante
- Servicio de Hematología y Unidad Central de Radiodiagnóstico, Hospital Infanta Leonor, Madrid, Spain
| | - Belén de la Morena-Barrio
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - José M García
- Servicio de Hematología y Unidad Central de Radiodiagnóstico, Hospital Infanta Leonor, Madrid, Spain
| | - Ágota Schlammadinger
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Rosa Cifuentes-Riquelme
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | | | - Antonia Miñano
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - Jose Padilla
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - Vicente Vicente
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - Javier Corral
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - Zsuzsanna Bereczky
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
14
|
Ding Y, Li Y, Zhao Z, Cliff Zhang Q, Liu F. The chromatin-remodeling enzyme Smarca5 regulates erythrocyte aggregation via Keap1-Nrf2 signaling. eLife 2021; 10:72557. [PMID: 34698638 PMCID: PMC8594921 DOI: 10.7554/elife.72557] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/23/2021] [Indexed: 12/30/2022] Open
Abstract
Although thrombosis has been extensively studied using various animal models, our understanding of the underlying mechanism remains elusive. Here, using zebrafish model, we demonstrated that smarca5-deficient red blood cells (RBCs) formed blood clots in the caudal vein plexus. We further used the anti-thrombosis drugs to treat smarca5zko1049a embryos and found that a thrombin inhibitor, argatroban, partially prevented blood clot formation in smarca5zko1049a. To explore the regulatory mechanism of smarca5 in RBC homeostasis, we profiled the chromatin accessibility landscape and transcriptome features in RBCs from smarca5zko1049a and their siblings and found that both the chromatin accessibility at the keap1a promoter and expression of keap1a were decreased. Keap1 is a suppressor protein of Nrf2, which is a major regulator of oxidative responses. We further identified that the expression of hmox1a, a downstream target of Keap1-Nrf2 signaling pathway, was markedly increased upon smarca5 deletion. Importantly, overexpression of keap1a or knockdown of hmox1a partially rescued the blood clot formation, suggesting that the disrupted Keap1-Nrf2 signaling is responsible for the RBC aggregation in smarca5 mutants. Together, our study using zebrafish smarca5 mutants characterizes a novel role for smarca5 in RBC aggregation, which may provide a new venous thrombosis animal model to support drug screening and pre-clinical therapeutic assessments to treat thrombosis.
Collapse
Affiliation(s)
- Yanyan Ding
- The Max-Planck Center for Tissue Stem Cell Research and Regenerative Medicine, Bioland Laboratory, Guangzhou, China.,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuzhe Li
- MOE Key Laboratory of Bioinformatics, Beijing Advanced Innovation Center for Structural Biology & Frontier Research Center for Biological Structure, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ziqian Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qiangfeng Cliff Zhang
- MOE Key Laboratory of Bioinformatics, Beijing Advanced Innovation Center for Structural Biology & Frontier Research Center for Biological Structure, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
15
|
A genetic modifier of venous thrombosis in zebrafish reveals a functional role for fibrinogen AαE in early hemostasis. Blood Adv 2021; 4:5480-5491. [PMID: 33166405 DOI: 10.1182/bloodadvances.2020001472] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 10/02/2020] [Indexed: 12/30/2022] Open
Abstract
Plasma fibrinogen molecules comprise 2 copies of Aα, Bβ, and γ chains folded into a hexameric protein. A minor fibrinogen isoform with an extended Aα chain (AαE) is more abundant in newborn human blood than in adults. Larval zebrafish produce predominantly AαE-containing fibrinogen, but its functional significance is unclear. In 3-day-old zebrafish, when hemostasis is reliant on fibrinogen and erythrocyte-rich clotting but is largely thrombocyte-independent, we measured the time to occlusion (TTO) in a laser-induced venous thrombosis assay in 3 zebrafish strains (AB, TU, and AB × TL hybrids). AB larvae showed delayed TTO compared with the TU and AB × TL strains. Mating AB with TU or TL produced larvae with a TU-like TTO. In contrast to TU, AB larvae failed to produce fibrinogen AαE, due to a mutation in the AαE-specific coding region of fibrinogen α-chain gene (fga). We investigated whether the lack of AαE explained the delayed AB TTO. Transgenic expression of AαE, but not Aα, shortened the AB TTO to that of TU. AαE rescued venous occlusion in fibrinogen mutants or larvae with morpholino-targeted fibrinogen α-chain messenger RNA, but Aα was less effective. In 5-day-old larvae, circulating thrombocytes contribute to hemostasis, as visualized in Tg(itga2b:EGFP) transgenics. Laser-induced venous thrombocyte adhesion and aggregation is reduced in fibrinogen mutants, but transgenic expression of Aα or AαE restored similar thrombocyte accumulation at the injury site. Our data demonstrate a genetic modifier of venous thrombosis and a role for fibrinogen AαE in early developmental blood coagulation, and suggest a link between differentially expressed fibrinogen isoforms and the cell types available for clotting.
Collapse
|
16
|
Fish RJ, Freire C, Di Sanza C, Neerman-Arbez M. Venous Thrombosis and Thrombocyte Activity in Zebrafish Models of Quantitative and Qualitative Fibrinogen Disorders. Int J Mol Sci 2021; 22:E655. [PMID: 33440782 PMCID: PMC7826895 DOI: 10.3390/ijms22020655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/28/2020] [Accepted: 01/04/2021] [Indexed: 12/18/2022] Open
Abstract
Venous thrombosis occurs in patients with quantitative and qualitative fibrinogen disorders. Injury-induced thrombosis in zebrafish larvae has been used to model human coagulopathies. We aimed to determine whether zebrafish models of afibrinogenemia and dysfibrinogenemia have different thrombotic phenotypes. Laser injuries were used to induce venous thrombosis and the time-to-occlusion (TTO) and the binding and aggregation of fluorescent Tg(itga2b:EGFP) thrombocytes measured. The fga-/- larvae failed to support occlusive venous thrombosis and showed reduced thrombocyte binding and aggregation at injury sites. The fga+/- larvae were largely unaffected. When genome editing zebrafish to produce fibrinogen Aα R28C, equivalent to the human Aα R35C dysfibrinogenemia mutation, we detected in-frame skipping of exon 2 in the fga mRNA, thereby encoding AαΔ19-56. This mutation is similar to Fibrinogen Montpellier II which causes hypodysfibrinogenemia. Aα+/Δ19-56 fish had prolonged TTO and reduced thrombocyte activity, a dominant effect of the mutation. Finally, we used transgenic expression of fga R28C cDNA in fga knock-down or fga-/- mutants to model thrombosis in dysfibrinogenemia. Aα R28C expression had similar effects on TTO and thrombocyte activity as Aα+/Δ19-56. We conclude that thrombosis assays in larval zebrafish can distinguish between quantitative and qualitative fibrinogen disorder models and may assist in anticipating a thrombotic phenotype of novel fibrinogen mutations.
Collapse
Affiliation(s)
| | | | | | - Marguerite Neerman-Arbez
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; (R.J.F.); (C.F.); (C.D.S.)
| |
Collapse
|
17
|
Cholan PM, Han A, Woodie BR, Watchon M, Kurz AR, Laird AS, Britton WJ, Ye L, Holmes ZC, McCann JR, David LA, Rawls JF, Oehlers SH. Conserved anti-inflammatory effects and sensing of butyrate in zebrafish. Gut Microbes 2020; 12:1-11. [PMID: 33064972 PMCID: PMC7575005 DOI: 10.1080/19490976.2020.1824563] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Short-chain fatty acids (SCFAs) are produced by microbial fermentation of dietary fiber in the gut. Butyrate is a particularly important SCFA with anti-inflammatory properties and is generally present at lower levels in inflammatory diseases associated with gut microbiota dysbiosis in mammals. We aimed to determine if SCFAs are produced by the zebrafish microbiome and if SCFAs exert conserved effects on zebrafish immunity as an example of the non-mammalian vertebrate immune system. We demonstrate that bacterial communities from adult zebrafish intestines synthesize all three main SCFA in vitro, although SCFA were below our detectable limits in zebrafish intestines in vivo. Immersion in butyrate, but not acetate or propionate, reduced the recruitment of neutrophils and M1-type pro-inflammatory macrophages to wounds. We found conservation of butyrate sensing by neutrophils via orthologs of the hydroxycarboxylic acid receptor 1 (hcar1) gene. Neutrophils from Hcar1-depleted embryos were no longer responsive to the anti-inflammatory effects of butyrate, while macrophage sensitivity to butyrate was independent of Hcar1. Our data demonstrate conservation of anti-inflammatory butyrate effects and identify the presence of a conserved molecular receptor in fish.
Collapse
Affiliation(s)
- Pradeep Manuneedhi Cholan
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney , Camperdown, Australia
| | - Alvin Han
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - Brad R Woodie
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney , Camperdown, Australia.,Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - Maxinne Watchon
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University , Macquarie Park, Australia.,Sydney Medical School, The University of Sydney , Camperdown, Australia
| | - Angela Rm Kurz
- Centenary Imaging and Sydney Cytometry at the Centenary Institute, The University of Sydney , Camperdown, Australia
| | - Angela S Laird
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University , Macquarie Park, Australia
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney , Camperdown, Australia.,The University of Sydney, Discipline of Infectious Diseases and Immunology, Faculty of Medicine and Health, And Marie Bashir Institute , Camperdown, Australia.,Department of Clinical Immunology, Royal Prince Alfred Hospital , Camperdown, Australia
| | - Lihua Ye
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - Zachary C Holmes
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - Jessica R McCann
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - Lawrence A David
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney , Camperdown, Australia.,The University of Sydney, Discipline of Infectious Diseases and Immunology, Faculty of Medicine and Health, And Marie Bashir Institute , Camperdown, Australia
| |
Collapse
|
18
|
Shirozu K, Karashima Y, Yamaura K. Effect of antithrombin in fresh frozen plasma on hemostasis after cardiopulmonary bypass surgery. Perfusion 2020; 36:395-400. [PMID: 32781884 DOI: 10.1177/0267659120948435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Supplementation of fresh frozen plasma immediately after cardiopulmonary bypass is an effective method to enhance clotting ability as coagulation factors are consumed in the extracorporeal circuit during cardiopulmonary bypass. On the other hand, the anticoagulation factors in fresh frozen plasma can also deter the clotting ability. This study investigated the effect of fresh frozen plasma administration on the comprehensive clotting ability following cardiopulmonary bypass. METHODS This prospective observational study included 22 patients scheduled for cardiac surgery. Clotting times and maximum clot firmness were evaluated using the types of rotational thromboelastometry, intrinsic rotational thromboelastometry, and heparinase thromboelastography preoperatively, immediately after cardiopulmonary bypass, and 1 hour after cardiopulmonary bypass. Activated clotting time, antithrombin activity, and heparin concentration were also measured at these time-points. RESULTS Antithrombin activity (62.9 ± 7.2% vs. 51.1 ± 7.4%, p < 0.0001) and activated clotting time (132.6 ± 9.6% vs. 120.0 ± 9.0%, p < 0.001) were significantly higher 1 hour after cardiopulmonary bypass compared to measurements taken immediately after cardiopulmonary bypass. Heparin concentration 1 hour after cardiopulmonary bypass was significantly decreased compared to that immediately after cardiopulmonary bypass. On the other hand, maximum clot firmness determined via intrinsic rotational thromboelastometry was significantly greater 1 hour after cardiopulmonary bypass (53.8 ± 4.8 mm) than that immediately after cardiopulmonary bypass (49.5 ± 4.8 mm). Clotting time determined via intrinsic rotational thromboelastometry and heparinase thromboelastography was also significantly shorter 1 hour after cardiopulmonary bypass than that immediately after cardiopulmonary bypass. CONCLUSION Fresh frozen plasma administration increased antithrombin activity and caused activated clotting time prolongation, but then increased clotting ability. Thus, testing by rotational thromboelastometry after cardiopulmonary bypass could be valuable in the detection of comprehensive clotting ability.
Collapse
Affiliation(s)
- Kazuhiro Shirozu
- Department of Anesthesiology and Critical Care Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Yuji Karashima
- Operating Rooms, Kyushu University Hospital, Fukuoka, Japan
| | - Ken Yamaura
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
19
|
Bravo-Pérez C, de la Morena-Barrio ME, Palomo A, Entrena L, de la Morena-Barrio B, Padilla J, Miñano A, Navarro E, Cifuentes R, Corral J, Vicente V. Genotype-phenotype gradient of SERPINC1 variants in a single family reveals a severe compound antithrombin deficiency in a dead embryo. Br J Haematol 2020; 191:e32-e35. [PMID: 32686144 DOI: 10.1111/bjh.16963] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 06/18/2020] [Indexed: 11/27/2022]
Affiliation(s)
- C Bravo-Pérez
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - M E de la Morena-Barrio
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - A Palomo
- Servicio de Hematología y Hemoterapia del centro Materno-Infantil del Hospital Regional Universitario Carlos de Haya, Málaga, Spain
| | - L Entrena
- Servicio de Hematología, Hospital Virgen de las Nieves, Granada, Spain
| | - B de la Morena-Barrio
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - J Padilla
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - A Miñano
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - E Navarro
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - R Cifuentes
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - J Corral
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| | - V Vicente
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, University of Murcia, IMIB-Arrixaca, CIBERER, Murcia, Spain
| |
Collapse
|
20
|
Analysis of factor V in zebrafish demonstrates minimal levels needed for early hemostasis. Blood Adv 2020; 3:1670-1680. [PMID: 31167819 DOI: 10.1182/bloodadvances.2018029066] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 04/17/2019] [Indexed: 12/28/2022] Open
Abstract
In humans, coagulation factor V (FV) deficiency is a rare, clinically heterogeneous bleeding disorder, suggesting that genetic modifiers may contribute to disease expressivity. Zebrafish possess many distinct advantages including high fecundity, optical clarity, external development, and homology with the mammalian hemostatic system, features that make it ideal for genetic studies. Our aim was to study the role of FV in zebrafish through targeted mutagenesis and apply the model to the study of human F5 variants. CRISPR-mediated genome editing of the zebrafish f5 locus was performed, generating mutants homozygous for a 49 base pair deletion in exon 4. Thrombus formation secondary to vascular endothelial injury was absent in f5 -/- mutant embryos and larvae. Despite this severe hemostatic defect, homozygous mutants survived before succumbing to severe hemorrhage in adulthood. Human F5 variants of uncertain significance from patients with FV deficiency were evaluated, and the causative mutations identified and stratified by their ability to restore thrombus formation in larvae. Analysis of these novel mutations demonstrates variable residual FV function, with minimal activity being required to restore hemostasis in response to laser-induced endothelial injury. This in vivo evaluation may be beneficial for patients whose factor activity levels lack correlation with bleeding symptomatology, although limitations exist. Furthermore, homozygous mutant embryos tolerate what is a severe and lethal defect in mammals, suggesting the possibility of species-specific factors enabling survival, and allowing further study not possible in the mouse. Identification of these factors or other genetic modifiers could lead to novel therapeutic modalities.
Collapse
|
21
|
Zapilko V, Fish RJ, Garcia A, Reny JL, Dunoyer-Geindre S, Lecompte T, Neerman-Arbez M, Fontana P. MicroRNA-126 is a regulator of platelet-supported thrombin generation. Platelets 2020; 31:746-755. [DOI: 10.1080/09537104.2020.1775804] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Veronika Zapilko
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Richard J. Fish
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Alix Garcia
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jean-Luc Reny
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of General Internal Medicine, Geneva University Hospitals, Geneva, Switzerland
| | | | - Thomas Lecompte
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Pierre Fontana
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Angiology and Haemostasis, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
22
|
Grzegorski SJ, Hu Z, Liu Y, Yu X, Ferguson AC, Madarati H, Friedmann AP, Reyon D, Kim PY, Kretz CA, Joung JK, Shavit JA. Disruption of the kringle 1 domain of prothrombin leads to late onset mortality in zebrafish. Sci Rep 2020; 10:4049. [PMID: 32132579 PMCID: PMC7055286 DOI: 10.1038/s41598-020-60840-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/22/2020] [Indexed: 02/04/2023] Open
Abstract
The ability to prevent blood loss in response to injury is a conserved function of all vertebrates. Complete deficiency of the central clotting enzyme prothrombin has never been observed in humans and is incompatible with postnatal life in mice, thus limiting the ability to study its role in vivo. Zebrafish are able to tolerate severe hemostatic deficiencies that are lethal in mammals. We have generated a targeted genetic deletion in the kringle 1 domain of zebrafish prothrombin. Homozygous mutant embryos develop normally into the mid-juvenile stage but demonstrate complete mortality by 2 months of age primarily due to internal hemorrhage. Mutants are unable to form occlusive venous and arterial thrombi in response to endothelial injury, a defect that was phenocopied using direct oral anticoagulants. Human prothrombin engineered with the equivalent mutation exhibits a severe reduction in secretion, thrombin generation, and fibrinogen cleavage. Together, these data demonstrate the conserved function of thrombin in zebrafish and provide insight into the role of kringle 1 in prothrombin maturation and activity. Understanding how zebrafish are able to develop normally and survive into early adulthood without thrombin activity will provide important insight into its pleiotropic functions as well as the management of patients with bleeding disorders.
Collapse
Affiliation(s)
| | - Zhilian Hu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
- Oxford University, Oxford, UK
| | - Yang Liu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
- Molecular Innovations, Inc., Novi, MI, USA
| | - Xinge Yu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | | | - Hasam Madarati
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Thromosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - Alexander P Friedmann
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Thromosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - Deepak Reyon
- Molecular Pathology Unit, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
- Editas Medicine Inc., Cambridge, MA, USA
| | - Paul Y Kim
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Thromosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - Colin A Kretz
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Thromosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - J Keith Joung
- Molecular Pathology Unit, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Jordan A Shavit
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
23
|
Wu TS, Lin YT, Huang YT, Yu FY, Liu BH. Ochratoxin A triggered intracerebral hemorrhage in embryonic zebrafish: Involvement of microRNA-731 and prolactin receptor. CHEMOSPHERE 2020; 242:125143. [PMID: 31675585 DOI: 10.1016/j.chemosphere.2019.125143] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 06/10/2023]
Abstract
Ochratoxin A (OTA), a mycotoxin widely found in foodstuffs, reportedly damages multiple brain regions in developing rodents, but the corresponding mechanisms have not been elucidated. In this study, zebrafish embryos at 6 h post fertilization (hpf) were exposed to various concentrations of OTA and the phenomenon associated with intracerebral hemorrhage was observed at 72 hpf. Exposure of embryos to OTA significantly increased their hemorrhagic rate in a dose-dependent manner. Large numbers of extravagated erythrocytes were observed in the midbrain/hindbrain areas of Tg(fli-1a:EGFP; gata1:DsRed) embryos following exposure to OTA. OTA also disrupted the vascular patterning, especially the arch-shaped central arteries (CtAs), in treated embryos. Histological analysis revealed a cavity-like pattern in their hindbrain ventricles, implying the possibility of cerebral edema. OTA-induced intracerebral hemorrhage and CtA vessel defects were partially reversed by the presence of miR-731 antagomir or the overexpression of prolactin receptor a (prlra); prlra is a downstream target of miR-731. These results suggest that exposure to OTA has a negative effect on cerebral vasculature development by interfering with the miR-731/PRLR axis in zebrafish.
Collapse
Affiliation(s)
- Ting-Shuan Wu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Ting Lin
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ying-Tzu Huang
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Feng-Yih Yu
- Department of Biomedical Sciences, Chung Shan Medical University, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Biing-Hui Liu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
24
|
Deller RC, Richardson T, Richardson R, Bevan L, Zampetakis I, Scarpa F, Perriman AW. Artificial cell membrane binding thrombin constructs drive in situ fibrin hydrogel formation. Nat Commun 2019; 10:1887. [PMID: 31015421 PMCID: PMC6478844 DOI: 10.1038/s41467-019-09763-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 03/21/2019] [Indexed: 12/13/2022] Open
Abstract
Cell membrane re-engineering is emerging as a powerful tool for the development of next generation cell therapies, as it allows the user to augment therapeutic cells to provide additional functionalities, such as homing, adhesion or hypoxia resistance. To date, however, there are few examples where the plasma membrane is re-engineered to display active enzymes that promote extracellular matrix protein assembly. Here, we report on a self-contained matrix-forming system where the membrane of human mesenchymal stem cells is modified to display a novel thrombin construct, giving rise to spontaneous fibrin hydrogel nucleation and growth at near human plasma concentrations of fibrinogen. The cell membrane modification process is realised through the synthesis of a membrane-binding supercationic thrombin-polymer surfactant complex. Significantly, the resulting robust cellular fibrin hydrogel constructs can be differentiated down osteogenic and adipogenic lineages, giving rise to self-supporting monoliths that exhibit Young's moduli that reflect their respective extracellular matrix compositions.
Collapse
Affiliation(s)
- Robert C Deller
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- School of Engineering, University of Liverpool, Liverpool, L69 3GH, UK
| | - Thomas Richardson
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- Bristol Centre for Functional Nanomaterials, University of Bristol, Bristol, BS8 1FD, UK
| | - Rebecca Richardson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Laura Bevan
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Ioannis Zampetakis
- Bristol Composites Institute (ACCIS), University of Bristol, Bristol, BS8 1TR, UK
| | - Fabrizio Scarpa
- Bristol Composites Institute (ACCIS), University of Bristol, Bristol, BS8 1TR, UK
| | - Adam W Perriman
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
25
|
Hu Z, Lavik KI, Liu Y, Vo AH, Richter CE, Di Paola J, Shavit JA. Loss of fibrinogen in zebrafish results in an asymptomatic embryonic hemostatic defect and synthetic lethality with thrombocytopenia. J Thromb Haemost 2019; 17:607-617. [PMID: 30663848 PMCID: PMC6443434 DOI: 10.1111/jth.14391] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Indexed: 12/17/2022]
Abstract
Essentials Loss of fibrinogen in zebrafish has been previously shown to result in adult onset hemorrhage Hemostatic defects were discovered in early fga-/- embryos but well tolerated until adulthood Afibrinogenemia and thrombocytopenia results in synthetic lethality in zebrafish. Testing human FGA variants of uncertain significance in zebrafish identified causative mutations SUMMARY: Background Mutations in the alpha chain of fibrinogen (FGA), such as deficiencies in other fibrinogen subunits, lead to rare inherited autosomal recessive hemostatic disorders. These range from asymptomatic to catastrophic life-threatening bleeds and the molecular basis of inherited fibrinogen deficiencies is only partially understood. Zinc finger nucleases have been used to produce mutations in zebrafish fga, resulting in overt adult-onset hemorrhage and reduced survival. Objectives To determine the age of onset of hemostatic defects in afibrinogenemic zebrafish and model human fibrinogen deficiencies. Methods TALEN genome editing (transcription activator-like effector nucleases) was used to generate a zebrafish fga mutant. Hemostatic defects were assessed through survival, gross anatomical and histological observation and laser-induced endothelial injury. Human FGA variants with unknown pathologies were engineered into the orthologous positions in zebrafish fga. Results Loss of Fga decreased survival and resulted in synthetic lethality when combined with thrombocytopenia. Zebrafish fga mutants exhibit a severe hemostatic defect by 3 days of life, but without visible hemorrhage. Induced thrombus formation through venous endothelial injury was completely absent in mutant embryos and larvae. This hemostatic defect was restored by microinjection of wild-type fga cDNA plasmid or purified human fibrinogen. This system was used to determine whether unknown human variants were pathological by engineering them into fga. Conclusions These studies confirm that loss of fibrinogen in zebrafish results in the absence of hemostasis from the embryonic period through adulthood. When combined with thrombocytopenia, zebrafish exhibit synthetic lethality, demonstrating that thrombocytes are necessary for survival in response to hemorrhage.
Collapse
Affiliation(s)
- Zhilian Hu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Kari I Lavik
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Yang Liu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Andy H Vo
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | | | - Jorge Di Paola
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jordan A Shavit
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
26
|
Rost MS, Shestopalov I, Liu Y, Vo AH, Richter CE, Emly SM, Barrett FG, Stachura DL, Holinstat M, Zon LI, Shavit JA. Nfe2 is dispensable for early but required for adult thrombocyte formation and function in zebrafish. Blood Adv 2018; 2:3418-3427. [PMID: 30504234 PMCID: PMC6290098 DOI: 10.1182/bloodadvances.2018021865] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/31/2018] [Indexed: 12/20/2022] Open
Abstract
The NFE2 transcription factor is expressed in multiple hematopoietic lineages with a well-defined role in regulating megakaryocyte biogenesis and platelet production in mammals. Mice deficient in NFE2 develop severe thrombocytopenia with lethality resulting from neonatal hemorrhage. Recent data in mammals reveal potential differences in embryonic and adult thrombopoiesis. Multiple studies in zebrafish have revealed mechanistic insights into hematopoiesis, although thrombopoiesis has been less studied. Rather than platelets, zebrafish possess thrombocytes, which are nucleated cells with similar functional properties. Using transcription activator-like effector nucleases to generate mutations in nfe2, we show that unlike mammals, zebrafish survive to adulthood in the absence of Nfe2. Despite developing severe thrombocytopenia, homozygous mutants do not display overt hemorrhage or reduced survival. Surprisingly, quantification of circulating thrombocytes in mutant 6-day-old larvae revealed no significant differences from wild-type siblings. Both wild-type and nfe2 null larvae formed thrombocyte-rich clots in response to endothelial injury. In addition, ex vivo thrombocytic colony formation was intact in nfe2 mutants, and adult kidney marrow displayed expansion of hematopoietic progenitors. These data suggest that loss of Nfe2 results in a late block in adult thrombopoiesis, with secondary expansion of precursors: features consistent with mammals. Overall, our data suggest parallels with erythropoiesis, including distinct primitive and definitive pathways of development and potential for a previously unknown Nfe2-independent pathway of embryonic thrombopoiesis. Long-term homozygous mutant survival will facilitate in-depth study of Nfe2 deficiency in vivo, and further investigation could lead to alternative methodologies for the enhancement of platelet production.
Collapse
Affiliation(s)
- Megan S Rost
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| | - Ilya Shestopalov
- Boston Children's Hospital and Harvard Medical School, Boston, MA
| | - Yang Liu
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| | - Andy H Vo
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| | - Catherine E Richter
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| | - Sylvia M Emly
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| | | | - David L Stachura
- Department of Biological Sciences, California State University Chico, Chico, CA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan, Ann Arbor, MI; and
| | - Leonard I Zon
- Boston Children's Hospital and Harvard Medical School, Boston, MA
- Stem Cell Program and Division of Hematology/Oncology, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Dana-Farber Cancer Institute and Howard Hughes Medical Institute, Boston, MA
| | - Jordan A Shavit
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| |
Collapse
|
27
|
Rebl A, Goldammer T. Under control: The innate immunity of fish from the inhibitors' perspective. FISH & SHELLFISH IMMUNOLOGY 2018; 77:328-349. [PMID: 29631025 DOI: 10.1016/j.fsi.2018.04.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 06/08/2023]
Abstract
The innate immune response involves a concerted network of induced gene products, preformed immune effectors, biochemical signalling cascades and specialised cells. However, the multifaceted activation of these defensive measures can derail or overshoot and, if left unchecked, overwhelm the host. A plenty of regulatory devices therefore mediate the fragile equilibrium between pathogen defence and pathophysiological manifestations. Over the past decade in particular, an almost complete set of teleostean sequences orthologous to mammalian immunoregulatory factors has been identified in various fish species, which prove the remarkable conservation of innate immune-control concepts among vertebrates. This review will present the current knowledge on more than 50 teleostean regulatory factors (plus additional fish-specific paralogs) that are of paramount importance for controlling the clotting cascade, the complement system, pattern-recognition pathways and cytokine-signalling networks. A special focus lies on those immunoregulatory features that have emerged as potential biomarker genes in transcriptome-wide research studies. Moreover, we report on the latest progress in elucidating control elements that act directly with immune-gene-encoding nucleic acids, such as transcription factors, hormone receptors and micro- and long noncoding RNAs. Investigations into the function of teleostean inhibitory factors are still mainly based on gene-expression profiling or overexpression studies. However, in support of structural and in-vitro analyses, evidence from in-vivo trials is also available and revealed many biochemical details on piscine immune regulation. The presence of multiple gene copies in fish adds a degree of complexity, as it is so far hardly understood if they might play distinct roles during inflammation. The present review addresses this and other open questions that should be tackled by fish immunologists in future.
Collapse
Affiliation(s)
- Alexander Rebl
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Fish Genetics Unit, Dummerstorf, Germany.
| | - Tom Goldammer
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Fish Genetics Unit, Dummerstorf, Germany
| |
Collapse
|
28
|
Richard B, Swanson R, Izaguirre G, Olson ST. Cooperative Interactions of Three Hotspot Heparin Binding Residues Are Critical for Allosteric Activation of Antithrombin by Heparin. Biochemistry 2018; 57:2211-2226. [PMID: 29561141 DOI: 10.1021/acs.biochem.8b00216] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Heparin allosterically activates the anticoagulant serpin, antithrombin, by binding through a sequence-specific pentasaccharide and inducing activating conformational changes in the protein. Three basic residues of antithrombin, Lys114, Lys125, and Arg129, have been shown to be hotspots for binding the pentasaccharide, but the molecular basis for such hotspot binding has been unclear. To determine whether this results from cooperative interactions, we analyzed the effects of single, double, and triple mutations of the hotspot residues on pentasaccharide binding and activation of antithrombin. Double-mutant cycles revealed that the contribution of each residue to pentasaccharide binding energy was progressively reduced when one or both of the other residues were mutated, indicating strong coupling between each pair of residues that was dependent on the third residue and reflective of the three residues acting as a cooperative unit. Rapid kinetic studies showed that the hotspot residue mutations progressively abrogated the ability of the pentasaccharide to bind productively to native antithrombin and to conformationally activate the serpin by engaging the hotspot residues in an induced-fit interaction. Examination of the antithrombin-pentasaccharide complex structure revealed that the hotspot residues form two adjoining binding pockets for critical sulfates of the pentasaccharide that structurally link these residues. Together, these findings demonstrate that cooperative interactions of Lys114, Lys125, and Arg129 are critical for the productive induced-fit binding of the heparin pentasaccharide to antithrombin that allosterically activates the anticoagulant function of the serpin.
Collapse
Affiliation(s)
- Benjamin Richard
- Center for Molecular Biology of Oral Diseases and Department of Periodontics , University of Illinois at Chicago , Chicago , Illinois 60612 , United States
| | - Richard Swanson
- Center for Molecular Biology of Oral Diseases and Department of Periodontics , University of Illinois at Chicago , Chicago , Illinois 60612 , United States
| | - Gonzalo Izaguirre
- Center for Molecular Biology of Oral Diseases and Department of Periodontics , University of Illinois at Chicago , Chicago , Illinois 60612 , United States
| | - Steven T Olson
- Center for Molecular Biology of Oral Diseases and Department of Periodontics , University of Illinois at Chicago , Chicago , Illinois 60612 , United States
| |
Collapse
|
29
|
Neerman-Arbez M, Casini A. Clinical Consequences and Molecular Bases of Low Fibrinogen Levels. Int J Mol Sci 2018; 19:E192. [PMID: 29316703 PMCID: PMC5796141 DOI: 10.3390/ijms19010192] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 12/22/2017] [Accepted: 12/22/2017] [Indexed: 12/19/2022] Open
Abstract
The study of inherited fibrinogen disorders, characterized by extensive allelic heterogeneity, allows the association of defined mutations with specific defects providing significant insight into the location of functionally important sites in fibrinogen and fibrin. Since the identification of the first causative mutation for congenital afibrinogenemia, studies have elucidated the underlying molecular pathophysiology of numerous causative mutations leading to fibrinogen deficiency, developed cell-based and animal models to study human fibrinogen disorders, and further explored the clinical consequences of absent, low, or dysfunctional fibrinogen. Since qualitative disorders are addressed by another review in this special issue, this review will focus on quantitative disorders and will discuss their diagnosis, clinical features, molecular bases, and introduce new models to study the phenotypic consequences of fibrinogen deficiency.
Collapse
Affiliation(s)
- Marguerite Neerman-Arbez
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland.
| | - Alessandro Casini
- Division of Angiology and Hemostasis, Faculty of Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland.
| |
Collapse
|
30
|
Inhibitory serpins. New insights into their folding, polymerization, regulation and clearance. Biochem J 2017; 473:2273-93. [PMID: 27470592 DOI: 10.1042/bcj20160014] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/31/2016] [Indexed: 12/20/2022]
Abstract
Serpins are a widely distributed family of high molecular mass protein proteinase inhibitors that can inhibit both serine and cysteine proteinases by a remarkable mechanism-based kinetic trapping of an acyl or thioacyl enzyme intermediate that involves massive conformational transformation. The trapping is based on distortion of the proteinase in the complex, with energy derived from the unique metastability of the active serpin. Serpins are the favoured inhibitors for regulation of proteinases in complex proteolytic cascades, such as are involved in blood coagulation, fibrinolysis and complement activation, by virtue of the ability to modulate their specificity and reactivity. Given their prominence as inhibitors, much work has been carried out to understand not only the mechanism of inhibition, but how it is fine-tuned, both spatially and temporally. The metastability of the active state raises the question of how serpins fold, whereas the misfolding of some serpin variants that leads to polymerization and pathologies of liver disease, emphysema and dementia makes it clinically important to understand how such polymerization might occur. Finally, since binding of serpins and their proteinase complexes, particularly plasminogen activator inhibitor-1 (PAI-1), to the clearance and signalling receptor LRP1 (low density lipoprotein receptor-related protein 1), may affect pathways linked to cell migration, angiogenesis, and tumour progression, it is important to understand the nature and specificity of binding. The current state of understanding of these areas is addressed here.
Collapse
|
31
|
Genome editing of factor X in zebrafish reveals unexpected tolerance of severe defects in the common pathway. Blood 2017; 130:666-676. [PMID: 28576875 DOI: 10.1182/blood-2017-02-765206] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/23/2017] [Indexed: 12/24/2022] Open
Abstract
Deficiency of factor X (F10) in humans is a rare bleeding disorder with a heterogeneous phenotype and limited therapeutic options. Targeted disruption of F10 and other common pathway factors in mice results in embryonic/neonatal lethality with rapid resorption of homozygous mutants, hampering additional studies. Several of these mutants also display yolk sac vascular defects, suggesting a role for thrombin signaling in vessel development. The zebrafish is a vertebrate model that demonstrates conservation of the mammalian hemostatic and vascular systems. We have leveraged these advantages for in-depth study of the role of the coagulation cascade in the developmental regulation of hemostasis and vasculogenesis. In this article, we show that ablation of zebrafish f10 by using genome editing with transcription activator-like effector nucleases results in a major embryonic hemostatic defect. However, widespread hemorrhage and subsequent lethality does not occur until later stages, with absence of any detectable defect in vascular development. We also use f10-/- zebrafish to confirm 5 novel human F10 variants as causative mutations in affected patients, providing a rapid and reliable in vivo model for testing the severity of F10 variants. These findings as well as the prolonged survival of f10-/- mutants will enable us to expand our understanding of the molecular mechanisms of hemostasis, including a platform for screening variants of uncertain significance in patients with F10 deficiency and other coagulation disorders. Further study as to how fish tolerate what is an early lethal mutation in mammals could facilitate improvement of diagnostics and therapeutics for affected patients with bleeding disorders.
Collapse
|
32
|
Jagadeeswaran P, Cooley BC, Gross PL, Mackman N. Animal Models of Thrombosis From Zebrafish to Nonhuman Primates: Use in the Elucidation of New Pathologic Pathways and the Development of Antithrombotic Drugs. Circ Res 2017; 118:1363-79. [PMID: 27126647 DOI: 10.1161/circresaha.115.306823] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/30/2015] [Indexed: 12/23/2022]
Abstract
Thrombosis is a leading cause of morbidity and mortality worldwide. Animal models are used to understand the pathological pathways involved in thrombosis and to test the efficacy and safety of new antithrombotic drugs. In this review, we will first describe the central role a variety of animal models of thrombosis and hemostasis has played in the development of new antiplatelet and anticoagulant drugs. These include the widely used P2Y12 antagonists and the recently developed orally available anticoagulants that directly target factor Xa or thrombin. Next, we will describe the new players, such as polyphosphate, neutrophil extracellular traps, and microparticles, which have been shown to contribute to thrombosis in mouse models, particularly venous thrombosis models. Other mouse studies have demonstrated roles for the factor XIIa and factor XIa in thrombosis. This has spurred the development of strategies to reduce their levels or activities as a new approach for preventing thrombosis. Finally, we will discuss the emergence of zebrafish as a model to study thrombosis and its potential use in the discovery of novel factors involved in thrombosis and hemostasis. Animal models of thrombosis from zebrafish to nonhuman primates are vital in identifying pathological pathways of thrombosis that can be safely targeted with a minimal effect on hemostasis. Future studies should focus on understanding the different triggers of thrombosis and the best drugs to prevent each type of thrombotic event.
Collapse
Affiliation(s)
- Pudur Jagadeeswaran
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.).
| | - Brian C Cooley
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.)
| | - Peter L Gross
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.)
| | - Nigel Mackman
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.)
| |
Collapse
|
33
|
Ismail HF, Hashim Z, Soon WT, Rahman NSA, Zainudin AN, Majid FAA. Comparative study of herbal plants on the phenolic and flavonoid content, antioxidant activities and toxicity on cells and zebrafish embryo. J Tradit Complement Med 2017; 7:452-465. [PMID: 29034193 PMCID: PMC5634737 DOI: 10.1016/j.jtcme.2016.12.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/26/2016] [Accepted: 12/27/2016] [Indexed: 02/08/2023] Open
Abstract
Natural antioxidants derived from plants have shown a tremendous inhibitory effect on free radicals in actively metabolizing cells. Overproduction of free radicals increases the risk factor of chronic diseases associated with diabetes, cancer, arthritis and cardiovascular disease. Andrographis paniculata, Cinnamon zeylanicum, Curcuma xanthorrhiza, Eugenia polyantha and Orthosiphon stamineus are ethnomedicinal plants used in the Asian region to treat various illnesses from a common fever to metabolic disease. In this study, we have quantified the total phenolic (TPC) and flavonoid content (TFC) in these plants and its inhibitory effect on 1,1-diphenyl-2-picrylhydrazyl radical (DPPH) and 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) free radicals as well as the cytotoxicity effect on cell lines proliferation and zebrafish embryogenesis. Results showed that Cinnamon zeylanicum and E. polyantha have the highest phenolic and flavonoid content. Furthermore, both herbs significantly inhibited the formation of DPPH and ABTS free radicals. Meanwhile, O. stamineus exhibited minimum cytotoxicity and embryotoxicity on tested models. Good correlation between IC50 of 3T3-L1 cells and LC50 embyrotoxicity was also found. This study revealed the potent activity of antioxidant against free radical and the toxicology levels of the tested herbal plants.
Collapse
Affiliation(s)
- Hassan Fahmi Ismail
- Department of Bioprocess and Polymer Engineering, Faculty of Chemical & Energy Engineering, Universiti Teknologi Malaysia, 81310, Malaysia
| | - Zanariah Hashim
- Department of Bioprocess and Polymer Engineering, Faculty of Chemical & Energy Engineering, Universiti Teknologi Malaysia, 81310, Malaysia
| | - Wong Tet Soon
- Department of Bioprocess and Polymer Engineering, Faculty of Chemical & Energy Engineering, Universiti Teknologi Malaysia, 81310, Malaysia
| | | | - Ain Nabihah Zainudin
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, 21030, Malaysia
| | | |
Collapse
|
34
|
Establishment of a congenital amegakaryocytic thrombocytopenia model and a thrombocyte–specific reporter line in zebrafish. Leukemia 2016; 31:1206-1216. [DOI: 10.1038/leu.2016.320] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/06/2016] [Accepted: 10/10/2016] [Indexed: 11/08/2022]
|
35
|
Zhao K, Liu Q. The clinical application of mesenchymal stromal cells in hematopoietic stem cell transplantation. J Hematol Oncol 2016; 9:46. [PMID: 27193054 PMCID: PMC4870746 DOI: 10.1186/s13045-016-0276-z] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/11/2016] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent stem cells well known for repairing tissue, supporting hematopoiesis, and modulating immune and inflammation response. These outstanding properties make MSCs as an attractive candidate for cellular therapy in immune-based disorders, especially hematopoietic stem cell transplantation (HSCT). In this review, we outline the progress of MSCs in preventing and treating engraftment failure (EF), graft-versus-host disease (GVHD) following HSCT and critically discuss unsolved issues in clinical applications.
Collapse
Affiliation(s)
- Ke Zhao
- Department of Hematology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Blvd North, Guangzhou, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Blvd North, Guangzhou, China.
| |
Collapse
|
36
|
Navarro-Fernández J, de la Morena-Barrio ME, Padilla J, Miñano A, Bohdan N, Águila S, Martínez-Martínez I, Sevivas TS, de Cos C, Fernández-Mosteirín N, Llamas P, Asenjo S, Medina P, Souto JC, Overvad K, Kristensen SR, Corral J, Vicente V. Antithrombin Dublin (p.Val30Glu): a relatively common variant with moderate thrombosis risk of causing transient antithrombin deficiency. Thromb Haemost 2016; 116:146-54. [PMID: 27098529 DOI: 10.1160/th15-11-0871] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/02/2016] [Indexed: 12/24/2022]
Abstract
The key haemostatic role of antithrombin and the risk of thrombosis associated with its deficiency support that the low incidence of antithrombin deficiency among patients with thrombosis might be explained by underestimation of this disorder. It was our aim to identify mutations in SERPINC1 causing transient antithrombin deficiency. SERPINC1 was sequenced in 214 cases with a positive test for antithrombin deficiency, including 67 with no deficiency in the sample delivered to our laboratory. The p.Val30Glu mutation (Antithrombin Dublin) was identified in five out of these 67 cases, as well as in three out of 127 cases with other SERPINC1 mutations. Genotyping in 1593 patients with venous thrombosis and 2592 controls from two populations, revealed a low prevalent polymorphism (0.3 %) that moderately increased the risk of venous thrombosis (OR: 2.9; 95 % CI: 1.07-8.09; p= 0.03) and identified one homozygous patient with an early thrombotic event. Carriers had normal anti-FXa activity, and plasma antithrombin was not sensitive to heat stress or proteolytic cleavage. Analysis of one sample with transient deficit revealed a type I deficiency, without aberrant or increased latent forms. The recombinant variant, which lacked the two amino-terminal residues, had reduced secretion from HEK-EBNA cells, formed hyperstable disulphide-linked polymers, and had negligible activity. In conclusion, p.Val30Glu by affecting the cleavage of antithrombin's signal peptide, results in a mature protein lacking the N-terminal dipeptide with no functional consequences in normal conditions, but that increases the sensitivity to be folded intracellularly into polymers, facilitating transient antithrombin deficiency and the subsequent risk of thrombosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Javier Corral
- Dr. Javier Corral, University of Murcia, Centro Regional de Hemodonación, Calle Ronda de Garay s/n, Murcia 30003, Spain, Tel.: +34968341990, Fax: +34968261914, E-mail:
| | | |
Collapse
|
37
|
Toderici M, de la Morena-Barrio ME, Padilla J, Miñano A, Antón AI, Iniesta JA, Herranz MT, Fernández N, Vicente V, Corral J. Identification of Regulatory Mutations in SERPINC1 Affecting Vitamin D Response Elements Associated with Antithrombin Deficiency. PLoS One 2016; 11:e0152159. [PMID: 27003919 PMCID: PMC4803246 DOI: 10.1371/journal.pone.0152159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/09/2016] [Indexed: 12/14/2022] Open
Abstract
Antithrombin is a crucial anticoagulant serpin whose even moderate deficiency significantly increases the risk of thrombosis. Most cases with antithrombin deficiency carried genetic defects affecting exons or flanking regions of SERPINC1.We aimed to identify regulatory mutations inSERPINC1 through sequencing the promoter, intron 1 and 2 of this gene in 23 patients with antithrombin deficiency but without known genetic defects. Three cases with moderate antithrombin deficiency (63–78%) carried potential regulatory mutations. One located 200 bp before the initiation ATG and two in intron 1. These mutations disrupted two out of five potential vitamin D receptor elements (VDRE) identified in SERPINC1 with different software. One genetic defect, c.42-1060_-1057dupTTGA, was a new low prevalent polymorphism (MAF: 0.01) with functional consequences on plasma antithrombin levels. The relevance of the vitamin D pathway on the regulation of SERPINC1 was confirmed in a cell model. Incubation of HepG2 with paricalcitol, a vitamin D analog, increased dose-dependently the levels of SERPINC1transcripts and antithrombin released to the conditioned medium. This study shows further evidence of the transcriptional regulation of SERPINC1 by vitamin D and first describes the functional and pathological relevance of mutations affecting VDRE of this gene. Our study opens new perspectives in the search of new genetic defects involved in antithrombin deficiency and the risk of thrombosis as well as in the design of new antithrombotic treatments.
Collapse
Affiliation(s)
- Mara Toderici
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, Murcia, Spain
| | - María Eugenia de la Morena-Barrio
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, Murcia, Spain
- * E-mail:
| | - José Padilla
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, Murcia, Spain
| | - Antonia Miñano
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, Murcia, Spain
| | - Ana Isabel Antón
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, Murcia, Spain
| | | | - María Teresa Herranz
- Servicio de Medicina Interna, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Nuria Fernández
- Servicio de Hematología y Hemoterapia, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Vicente Vicente
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, Murcia, Spain
| | - Javier Corral
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, Murcia, Spain
| |
Collapse
|
38
|
Abstract
Hemostasis is a coordinated system through which blood is prevented from exiting a closed circulatory system. We have taken advantage of the zebrafish, an emerging model for the study of blood coagulation, and describe three techniques for quantitative analysis of primary and secondary hemostasis. Collectively, these three techniques comprise a toolset to aid in our understanding of hemostasis and pathological clotting.
Collapse
Affiliation(s)
- M S Rost
- University of Michigan, Ann Arbor, MI, United States
| | | | - J A Shavit
- University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
39
|
Elenbaas JS, Maitra D, Liu Y, Lentz SI, Nelson B, Hoenerhoff MJ, Shavit JA, Omary MB. A precursor-inducible zebrafish model of acute protoporphyria with hepatic protein aggregation and multiorganelle stress. FASEB J 2016; 30:1798-810. [PMID: 26839379 DOI: 10.1096/fj.201500111r] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 01/06/2016] [Indexed: 01/24/2023]
Abstract
Protoporphyria is a metabolic disease that causes excess production of protoporphyrin IX (PP-IX), the final biosynthetic precursor to heme. Hepatic PP-IX accumulation may lead to end-stage liver disease. We tested the hypothesis that systemic administration of porphyrin precursors to zebrafish larvae results in protoporphyrin accumulation and a reproducible nongenetic porphyria model. Retro-orbital infusion of PP-IX or the iron chelator deferoxamine mesylate (DFO), with the first committed heme precursor α-aminolevulinic acid (ALA), generates high levels of PP-IX in zebrafish larvae. Exogenously infused or endogenously produced PP-IX accumulates preferentially in the liver of zebrafish larvae and peaks 1 to 3 d after infusion. Similar to patients with protoporphyria, PP-IX is excreted through the biliary system. Porphyrin accumulation in zebrafish liver causes multiorganelle protein aggregation as determined by mass spectrometry and immunoblotting. Endoplasmic reticulum stress and induction of autophagy were noted in zebrafish larvae and corroborated in 2 mouse models of protoporphyria. Furthermore, electron microscopy of zebrafish livers from larvae administered ALA + DFO showed hepatocyte autophagosomes, nuclear membrane ruffling, and porphyrin-containing vacuoles with endoplasmic reticulum distortion. In conclusion, systemic administration of the heme precursors PP-IX or ALA + DFO into zebrafish larvae provides a new model of acute protoporphyria with consequent hepatocyte protein aggregation and proteotoxic multiorganelle alterations and stress.-Elenbaas, J. S., Maitra, D., Liu, Y., Lentz, S. I., Nelson, B., Hoenerhoff, M. J., Shavit, J. A., Omary, M. B. A precursor-inducible zebrafish model of acute protoporphyria with hepatic protein aggregation and multiorganelle stress.
Collapse
Affiliation(s)
| | | | - Yang Liu
- Department of Pediatrics and Communicable Diseases, Division of Pediatric Hematology and Oncology
| | | | | | - Mark J Hoenerhoff
- In-Vivo Animal Core, Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA; and
| | - Jordan A Shavit
- Department of Pediatrics and Communicable Diseases, Division of Pediatric Hematology and Oncology
| | - M Bishr Omary
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, and Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|
40
|
Sertori R, Trengove M, Basheer F, Ward AC, Liongue C. Genome editing in zebrafish: a practical overview. Brief Funct Genomics 2015; 15:322-30. [DOI: 10.1093/bfgp/elv051] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
41
|
Roth R, Swanson R, Izaguirre G, Bock SC, Gettins PGW, Olson ST. Saturation Mutagenesis of the Antithrombin Reactive Center Loop P14 Residue Supports a Three-step Mechanism of Heparin Allosteric Activation Involving Intermediate and Fully Activated States. J Biol Chem 2015; 290:28020-28036. [PMID: 26359493 DOI: 10.1074/jbc.m115.678839] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Indexed: 11/06/2022] Open
Abstract
Past studies have suggested that a key feature of the mechanism of heparin allosteric activation of the anticoagulant serpin, antithrombin, is the release of the reactive center loop P14 residue from a native state stabilizing interaction with the hydrophobic core. However, more recent studies have indicated that this structural change plays a secondary role in the activation mechanism. To clarify this role, we expressed and characterized 15 antithrombin P14 variants. The variants exhibited basal reactivities with factors Xa and IXa, heparin affinities and thermal stabilities that were dramatically altered from wild type, consistent with the P14 mutations perturbing native state stability and shifting an allosteric equilibrium between native and activated states. Rapid kinetic studies confirmed that limiting rate constants for heparin allosteric activation of the mutants were altered in conjunction with the observed shifts of the allosteric equilibrium. However, correlations of the P14 mutations' effects on parameters reflecting the allosteric activation state of the serpin were inconsistent with a two-state model of allosteric activation and suggested multiple activated states. Together, these findings support a minimal three-state model of allosteric activation in which the P14 mutations perturb equilibria involving distinct native, intermediate, and fully activated states wherein the P14 residue retains an interaction with the hydrophobic core in the intermediate state but is released from the core in the fully activated state, and the bulk of allosteric activation has occurred in the intermediate.
Collapse
Affiliation(s)
- Ryan Roth
- Center for Molecular Biology of Oral Diseases and Departments of Periodontics
| | - Richard Swanson
- Center for Molecular Biology of Oral Diseases and Departments of Periodontics
| | - Gonzalo Izaguirre
- Center for Molecular Biology of Oral Diseases and Departments of Periodontics
| | - Susan C Bock
- Departments of Medicine and Bioengineering, University of Utah, Salt Lake City, Utah 84132
| | - Peter G W Gettins
- Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Steven T Olson
- Center for Molecular Biology of Oral Diseases and Departments of Periodontics.
| |
Collapse
|
42
|
Abstract
Hemostasis, the process of blood clot formation and resolution in response to vascular injury, and thrombosis, the dysregulation of hemostasis leading to pathological clot formation, are widely studied. However, the genetic variability in hemostatic and thrombotic disorders is incompletely understood, suggesting that novel mediators have yet to be uncovered. The zebrafish is developing into a powerful in vivo model to study hemostasis, and its features as a model organism are well suited to (a) develop high-throughput screens to identify novel mediators of hemostasis and thrombosis, (b) validate candidate genes identified in human populations, and (c) characterize the structure/function relationship of gene products. In this review, we discuss conservation of the zebrafish hemostatic system, highlight areas for future study, and outline the utility of this model to study blood coagulation and its dysregulation.
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Although the zebrafish has been established as a research tool over the past two to three decades, in hematology it has primarily been used to investigate areas distinct from coagulation. The advantages of this vertebrate model include high fecundity, rapid and external development, and conservation of virtually all clotting factors in the zebrafish genomic sequence. Here, we summarize the growing application of this technology to the study of hemostasis and thrombosis. RECENT FINDINGS Loss of function studies have demonstrated conservation of function for a number of zebrafish coagulation factors. These include positive and negative regulators of coagulation, as well as key components of the thrombus itself, such as von Willebrand factor, fibrinogen, and thrombocytes. Such analyses have also been leveraged to aid in the understanding of human variation and disease, as well as to perform in-vivo structure/function experiments. SUMMARY The zebrafish is an organism that lends itself to a number of unique and powerful approaches not possible in mammals. This review demonstrates that there is a high degree of genetic and functional conservation of coagulation, portending future insights into hemostasis and thrombosis through the use of this model.
Collapse
|
44
|
Abstract
Zebrafish (Danio rerio) embryos have proven to be a powerful model for studying a variety of developmental and disease processes. External development and optical transparency make these embryos especially amenable to microscopy, and numerous transgenic lines that label specific cell types with fluorescent proteins are available, making the zebrafish embryo an ideal system for visualizing the interaction of vascular, hematopoietic, and other cell types during injury and repair in vivo. Forward and reverse genetics in zebrafish are well developed, and pharmacological manipulation is possible. We describe a mechanical vascular injury model using micromanipulation techniques that exploits several of these features to study responses to vascular injury including hemostasis and blood vessel repair. Using a combination of video and timelapse microscopy, we demonstrate that this method of vascular injury results in measurable and reproducible responses during hemostasis and wound repair. This method provides a system for studying vascular injury and repair in detail in a whole animal model.
Collapse
Affiliation(s)
- Hilary Clay
- Cardiovascular Research Institute, University of California
| | | |
Collapse
|
45
|
|
46
|
Izaguirre G, Aguila S, Qi L, Swanson R, Roth R, Rezaie AR, Gettins PGW, Olson ST. Conformational activation of antithrombin by heparin involves an altered exosite interaction with protease. J Biol Chem 2014; 289:34049-64. [PMID: 25331949 DOI: 10.1074/jbc.m114.611707] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heparin allosterically activates antithrombin as an inhibitor of factors Xa and IXa by enhancing the initial Michaelis complex interaction of inhibitor with protease through exosites. Here, we investigate the mechanism of this enhancement by analyzing the effects of alanine mutations of six putative antithrombin exosite residues and three complementary protease exosite residues on antithrombin reactivity with these proteases in unactivated and heparin-activated states. Mutations of antithrombin Tyr(253) and His(319) exosite residues produced massive 10-200-fold losses in reactivity with factors Xa and IXa in both unactivated and heparin-activated states, indicating that these residues made critical attractive interactions with protease independent of heparin activation. By contrast, mutations of Asn(233), Arg(235), Glu(237), and Glu(255) exosite residues showed that these residues made both repulsive and attractive interactions with protease that depended on the activation state and whether the critical Tyr(253)/His(319) residues were mutated. Mutation of factor Xa Arg(143), Lys(148), and Arg(150) residues that interact with the exosite in the x-ray structure of the Michaelis complex confirmed the importance of all residues for heparin-activated antithrombin reactivity and Arg(150) for native serpin reactivity. These results demonstrate that the exosite is a key determinant of antithrombin reactivity with factors Xa and IXa in the native as well as the heparin-activated state and support a new model of allosteric activation we recently proposed in which a balance between attractive and repulsive exosite interactions in the native state is shifted to favor the attractive interactions in the activated state through core conformational changes induced by heparin binding.
Collapse
Affiliation(s)
- Gonzalo Izaguirre
- From the Department of Periodontics, Center for Molecular Biology of Oral Diseases and
| | - Sonia Aguila
- the Centro Regional de Hemodonación, University of Murcia, Murcia 30003, Spain, and
| | - Lixin Qi
- From the Department of Periodontics, Center for Molecular Biology of Oral Diseases and
| | - Richard Swanson
- From the Department of Periodontics, Center for Molecular Biology of Oral Diseases and
| | - Ryan Roth
- From the Department of Periodontics, Center for Molecular Biology of Oral Diseases and
| | - Alireza R Rezaie
- the Department of Biochemistry and Molecular Biology, St. Louis University, St. Louis, Missouri 63104
| | - Peter G W Gettins
- the Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Steven T Olson
- From the Department of Periodontics, Center for Molecular Biology of Oral Diseases and
| |
Collapse
|
47
|
Abstract
In this issue of Blood, Liu et al describe the creation of a null mutation for the antithrombin III gene (at3) in zebrafish by using zinc finger nuclease technology.
Collapse
|