1
|
Bargehr C, Knöfler R, Streif W. Treatment of Inherited Platelet Disorders: Current Status and Future Options. Hamostaseologie 2023; 43:261-270. [PMID: 37611608 DOI: 10.1055/a-2080-6602] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023] Open
Abstract
Inherited platelet disorders (IPDs) comprise a heterogeneous group of entities that manifest with variable bleeding tendencies. For successful treatment, the underlying platelet disorder, bleeding severity and location, age, and sex must be considered in the broader clinical context. Previous information from the AWMF S2K guideline #086-004 (www.awmf.org) is evaluated for validity and supplemented by information of new available and future treatment options and clinical scenarios that need specific measures. Special attention is given to the treatment of menorrhagia and risk management during pregnancy in women with IPDs. Established treatment options of IPDs include local hemostatic treatment, tranexamic acid, desmopressin, platelet concentrates, and recombinant activated factor VII. Hematopoietic stem cell therapy is a curative approach for selected patients. We also provide an outlook on promising new therapies. These include autologous hematopoietic stem cell gene therapy, artificial platelets and nanoparticles, and various other procoagulant treatments that are currently tested in clinical trials in the context of hemophilia.
Collapse
Affiliation(s)
- Caroline Bargehr
- Department of Paediatrics 1, Medical University of Innsbruck, Innsbruck, Austria
| | - Ralf Knöfler
- Department of Paediatric Haemostaseology, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Werner Streif
- Department of Paediatrics 1, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
2
|
Koukouritaki SB, Thinn AMM, Ashworth KJ, Fang J, Slater HS, Du LM, Nguyen HTT, Pillois X, Nurden AT, Ng CJ, Di Paola J, Zhu J, Wilcox DA. A single F153Sβ3 mutation causes constitutive integrin αIIbβ3 activation in a variant form of Glanzmann thrombasthenia. Blood Adv 2023; 7:3180-3191. [PMID: 36884296 PMCID: PMC10338211 DOI: 10.1182/bloodadvances.2022009495] [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: 12/06/2022] [Revised: 02/27/2023] [Accepted: 02/27/2023] [Indexed: 03/09/2023] Open
Abstract
This report identifies a novel variant form of the inherited bleeding disorder Glanzmann thrombasthenia, exhibiting only mild bleeding in a physically active individual. The platelets cannot aggregate ex vivo with physiologic agonists of activation, although microfluidic analysis with whole blood displays moderate ex vivo platelet adhesion and aggregation consistent with mild bleeding. Immunocytometry shows reduced expression of αIIbβ3 on quiescent platelets that spontaneously bind/store fibrinogen, and activation-dependent antibodies (ligand-induced binding site-319.4 and PAC-1) report β3 extension suggesting an intrinsic activation phenotype. Genetic analysis reveals a single F153Sβ3 substitution within the βI-domain from a heterozygous T556C nucleotide substitution of ITGB3 exon 4 in conjunction with a previously reported IVS5(+1)G>A splice site mutation with undetectable platelet messenger RNA accounting for hemizygous expression of S153β3. F153 is completely conserved among β3 of several species and all human β-integrin subunits suggesting that it may play a vital role in integrin structure/function. Mutagenesis of αIIb-F153Sβ3 also displays reduced levels of a constitutively activated αIIb-S153β3 on HEK293T cells. The overall structural analysis suggests that a bulky aromatic, nonpolar amino acid (F,W)153β3 is critical for maintaining the resting conformation of α2- and α1-helices of the βI-domain because small amino acid substitutions (S,A) facilitate an unhindered inward movement of the α2- and α1-helices of the βI-domain toward the constitutively active αIIbβ3 conformation, while a bulky aromatic, polar amino acid (Y) hinders such movements and restrains αIIbβ3 activation. The data collectively demonstrate that disruption of F153β3 can significantly alter normal integrin/platelet function, although reduced expression of αIIb-S153β3 may be compensated by a hyperactive conformation that promotes viable hemostasis.
Collapse
Affiliation(s)
- Sevasti B. Koukouritaki
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, WI
| | - Aye Myat M. Thinn
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| | - Katrina J. Ashworth
- Department of Pediatrics, Division of Hematology & Oncology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, WI
| | - Haley S. Slater
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, WI
| | - Lily M. Du
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, WI
| | | | - Xavier Pillois
- Xavier Arnozan Hôpital, Institut de Rythmologie et de Modélisation Cardiaque, Pessac, France
| | - Alan T. Nurden
- Xavier Arnozan Hôpital, Institut de Rythmologie et de Modélisation Cardiaque, Pessac, France
| | - Christopher J. Ng
- Department of Pediatrics, Section of Hematology/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jorge Di Paola
- Department of Pediatrics, Division of Hematology & Oncology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Jieqing Zhu
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| | - David A. Wilcox
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Milwaukee, WI
| |
Collapse
|
3
|
Fiore M, Giraudet JS, Alessi MC, Falaise C, Desprez D, d'Oiron R, Voisin S, Hurtaud MF, Boutroux H, Saultier P, Lavenu-Bombled C, Bagou G, Dubucs X, Chauvin A, Leroy C, Meckert F, Kerbaul F, Giraud N, Pühler A, Rath A. Emergency management of patients with Glanzmann thrombasthenia: consensus recommendations from the French reference center for inherited platelet disorders. Orphanet J Rare Dis 2023; 18:171. [PMID: 37386449 DOI: 10.1186/s13023-023-02787-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/18/2023] [Indexed: 07/01/2023] Open
Abstract
Glanzmann thrombasthenia (GT) is a genetic bleeding disorder characterised by severely reduced/absent platelet aggregation in response to multiple physiological agonists. The severity of bleeding in GT varies markedly, as does the emergency situations and complications encountered in patients. A number of emergency situations may occur in the context of GT, including spontaneous or provoked bleeding, such as surgery or childbirth. While general management principles apply in each of these settings, specific considerations are essential for the management of GT to avoid escalating minor bleeding events. These recommendations have been developed from a literature review and consensus from experts of the French Network for Inherited Platelet Disorders, the French Society of Emergency Medicine, representatives of patients' associations, and Orphanet to aid decision making and optimise clinical care by non-GT expert health professionals who encounter emergency situations in patients with GT.
Collapse
Affiliation(s)
- Mathieu Fiore
- Laboratoire d'hématologie, Centre de Référence des Pathologies Plaquettaires, CHU de Bordeaux, Hôpital Cardiologique, Inserm U1034 - Biologie des Maladies Cardio-Vasculaires, Pessac, France.
- Centre de Référence des Pathologies Plaquettaires, Pessac, France.
| | | | - Marie-Christine Alessi
- Laboratory of Hematology, Aix Marseille Univ, APHM, INSERM, INRAe, C2VN, La Timone Hospital, Marseille, France
- Reference Center of Platelet Disorders, APHM, Marseille, France
| | - Céline Falaise
- Department of Pediatric Hematology, Immunology and Oncology, La Timone Children's Hospital, Marseille, France
- Reference Center of Platelet Disorders, APHM, Marseille, France
| | - Dominique Desprez
- Centre de Ressources et de Compétences des Maladies Hémorragiques Constitutionnelles, CHU de Strasbourg, Strasbourg, France
| | - Roseline d'Oiron
- Centre de Ressources et de Compétences des Maladies Hémorragiques Constitutionnelles, CHU du Kremlin-Bicêtre, Le-Kremlin-Bicêtre, France
| | - Sophie Voisin
- Laboratoire d'Hématologie, Centre de Référence des Pathologies Plaquettaires, CHU de Toulouse, Toulouse, France
| | | | - Hélène Boutroux
- Laboratoire d'Hématologie, Centre de Référence des pathologies Plaquettaires, CHU Armand Trousseau, Paris, France
| | - Paul Saultier
- Department of pediatric hematology, immunology and oncology, Aix Marseille Univ, APHM, INSERM, INRAe, C2VN, La Timone Children's Hospital, Marseille, France
- Reference Center of Platelet Disorders, APHM, Marseille, France
| | - Cécile Lavenu-Bombled
- Service Hématologie Biologique, Centre de ressources et compétences MHEMO, CHU Bicêtre, Assistance Publique-Hôpitaux de Paris, Faculté de médecine Paris Saclay, Le Kremlin-Bicetre, France
| | - Gilles Bagou
- Anesthésiste-Réanimateur Urgentiste - SAMU-SMUR de Lyon - Hôpital Edouard-Herriot, 69437, Lyon Cedex 03, France
| | - Xavier Dubucs
- Pôle Médecine-Urgences, CHU de Toulouse, Toulouse, France
| | - Anthony Chauvin
- Président de la Commission des Référentiels de la SFMU (CREF), Chef de Service Adjoint - Service d'Accueil des Urgences/SMUR, CHU Lariboisière, Université de Paris, Paris, France
| | - Christophe Leroy
- Médecin Urgentiste - Service de Gestion des Crises Sanitaires - Département Qualité Gestion des Risques, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Francine Meckert
- Direction Opérationnelle du Prélèvement et de la Greffe de l'Agence de la Biomédecine (ABM), Saint Denis, France
| | - François Kerbaul
- Direction Opérationnelle du Prélèvement et de la Greffe de l'Agence de la Biomédecine (ABM), Saint Denis, France
| | | | - Ambra Pühler
- ORPHANET, INSERM US14, Plateforme Maladies Rares, 96 Rue Didot, 75014, Paris, France
| | - Ana Rath
- ORPHANET, INSERM US14, Plateforme Maladies Rares, 96 Rue Didot, 75014, Paris, France
| |
Collapse
|
4
|
Pokrovskaya ID, Rhee SW, Ball KK, Kamykowski JA, Zhao OS, Cruz DRD, Cohen J, Aronova MA, Leapman RD, Storrie B. Tethered platelet capture provides a mechanism for restricting circulating platelet activation to the wound site. Res Pract Thromb Haemost 2023; 7:100058. [PMID: 36865905 PMCID: PMC9971284 DOI: 10.1016/j.rpth.2023.100058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/20/2022] [Accepted: 12/30/2022] [Indexed: 01/24/2023] Open
Abstract
Background Puncture wounding is a longstanding challenge to human health for which understanding is limited, in part, by a lack of detailed morphological data on how the circulating platelet capture to the vessel matrix leads to sustained, self-limiting platelet accumulation. Objectives The objective of this study was to produce a paradigm for self-limiting thrombus growth in a mouse jugular vein model. Methods Data mining of advanced electron microscopy images was performed from authors' laboratories. Results Wide-area transmission electron mcrographs revealed initial platelet capture to the exposed adventitia resulted in localized patches of degranulated, procoagulant-like platelets. Platelet activation to a procoagulant state was sensitive to dabigatran, a direct-acting PAR receptor inhibitor, but not to cangrelor, a P2Y12 receptor inhibitor. Subsequent thrombus growth was sensitive to both cangrelor and dabigatran and sustained by the capture of discoid platelet strings first to collagen-anchored platelets and later to loosely adherent peripheral platelets. Spatial examination indicated that staged platelet activation resulted in a discoid platelet tethering zone that was pushed progressively outward as platelets converted from one activation state to another. As thrombus growth slowed, discoid platelet recruitment became rare and loosely adherent intravascular platelets failed to convert to tightly adherent platelets. Conclusions In summary, the data support a model that we term Capture and Activate, in which the initial high platelet activation is directly linked to the exposed adventitia, all subsequent tethering of discoid platelets is to loosely adherent platelets that convert to tightly adherent platelets, and self-limiting, intravascular platelet activation over time is the result of decreased signaling intensity.
Collapse
Affiliation(s)
- Irina D Pokrovskaya
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Sung W Rhee
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kelly K Ball
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jeffrey A Kamykowski
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Oliver S Zhao
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Denzel R D Cruz
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Joshua Cohen
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Maria A Aronova
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Richard D Leapman
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Brian Storrie
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
5
|
Cai Y, Schroeder JA, Jing W, Gurski C, Williams CB, Wang S, Dittel BN, Shi Q. Targeting transmembrane-domain-less MOG expression to platelets prevents disease development in experimental autoimmune encephalomyelitis. Front Immunol 2022; 13:1029356. [PMID: 36389708 PMCID: PMC9647046 DOI: 10.3389/fimmu.2022.1029356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory autoimmune disease of the central nervous system with no cure yet. Here, we report genetic engineering of hematopoietic stem cells (HSCs) to express myelin oligodendrocyte glycoprotein (MOG), specifically in platelets, as a means of intervention to induce immune tolerance in experimental autoimmune encephalomyelitis (EAE), the mouse model of MS. The platelet-specific αIIb promoter was used to drive either a full-length or truncated MOG expression cassette. Platelet-MOG expression was introduced by lentivirus transduction of HSCs followed by transplantation. MOG protein was detected on the cell surface of platelets only in full-length MOG-transduced recipients, but MOG was detected in transmembrane-domain-less MOG1-157-transduced platelets intracellularly. We found that targeting MOG expression to platelets could prevent EAE development and attenuate disease severity, including the loss of bladder control in transduced recipients. Elimination of the transmembrane domains of MOG significantly enhanced the clinical efficacy in preventing the onset and development of the disease and induced CD4+Foxp3+ Treg cells in the EAE model. Together, our data demonstrated that targeting transmembrane domain-deleted MOG expression to platelets is an effective strategy to induce immune tolerance in EAE, which could be a promising approach for the treatment of patients with MS autoimmune disease.
Collapse
Affiliation(s)
- Yuanhua Cai
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Blood Research Institute, Versiti, Milwaukee, WI, United States
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jocelyn A. Schroeder
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Blood Research Institute, Versiti, Milwaukee, WI, United States
| | - Weiqing Jing
- Blood Research Institute, Versiti, Milwaukee, WI, United States
| | - Cody Gurski
- Blood Research Institute, Versiti, Milwaukee, WI, United States
| | - Calvin B. Williams
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Shaoyuan Wang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Bonnie N. Dittel
- Blood Research Institute, Versiti, Milwaukee, WI, United States
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Qizhen Shi
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Blood Research Institute, Versiti, Milwaukee, WI, United States
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, WI, United States
- Midwest Athletes Against Childhood Cancer (MACC) Fund Research Center, Milwaukee, WI, United States
| |
Collapse
|
6
|
Schroeder JA, Kuether EA, Fang J, Jing W, Weiler H, Wilcox DA, Montgomery RR, Shi Q. Thromboelastometry assessment of hemostatic properties in various murine models with coagulopathy and the effect of factor VIII therapeutics. J Thromb Haemost 2021; 19:2417-2427. [PMID: 34245090 PMCID: PMC8865566 DOI: 10.1111/jth.15456] [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: 05/09/2021] [Accepted: 07/07/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Rotational thromboelastometry (ROTEM) has been commonly used to assess the viscoelastic properties of the blood clotting process in the clinic for patients with a hemostatic or prothrombotic disorder. OBJECTIVE To evaluate the capability of ROTEM in assessing hemostatic properties in whole blood from various mouse models with genetic bleeding or clotting disease and the effect of factor VIII (FVIII) therapeutics in FVIIInull mice. METHODS Mice with a genetic deficiency in either a coagulation factor or a platelet glycoprotein were used in this study. The properties of platelet- or plasma-FVIII were also assessed. Citrated blood from mice was recalcified and used for ROTEM analysis. RESULTS We found that blood collected from the vena cava could generate reliable results from ROTEM analysis, but not blood collected from the tail vein, retro-orbital plexus, or submandibular vein. Age and sex did not significantly affect the hemostatic properties determined by ROTEM analysis. Clotting time (CT) and clot formation time (CFT) were significantly prolonged in FVIIInull (5- and 9-fold, respectively) and FIXnull (4- and 5.7-fold, respectively) mice compared to wild-type (WT)-C57BL/6J mice. Platelet glycoprotein (GP)IIIanull mice had significantly prolonged CFT (8.4-fold) compared to WT-C57BL/6J mice. CT and CFT in factor V (FV) Leiden mice were significantly shortened with an increased α-angle compared to WT-C57BL/6J mice. Using ROTEM analysis, we showed that FVIII expressed in platelets or infused into whole blood restored hemostasis of FVIIInull mice in a dose-dependent manner. CONCLUSION ROTEM is a reliable and sensitive assay for assessing therapeutics on hemostatic properties in mouse models with a bleeding or clotting disorder.
Collapse
Affiliation(s)
- Jocelyn A. Schroeder
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, USA
- MACC Fund Research Center, Milwaukee, Wisconsin, USA
| | - Erin A. Kuether
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, USA
- MACC Fund Research Center, Milwaukee, Wisconsin, USA
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- MACC Fund Research Center, Milwaukee, Wisconsin, USA
| | - Weiqing Jing
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA
| | - Hartmut Weiler
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA
| | - David A. Wilcox
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, USA
- MACC Fund Research Center, Milwaukee, Wisconsin, USA
| | - Robert R. Montgomery
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, USA
- MACC Fund Research Center, Milwaukee, Wisconsin, USA
| | - Qizhen Shi
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, USA
- MACC Fund Research Center, Milwaukee, Wisconsin, USA
| |
Collapse
|
7
|
Nurden A. Profiling the Genetic and Molecular Characteristics of Glanzmann Thrombasthenia: Can It Guide Current and Future Therapies? J Blood Med 2021; 12:581-599. [PMID: 34267570 PMCID: PMC8275161 DOI: 10.2147/jbm.s273053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/16/2021] [Indexed: 12/15/2022] Open
Abstract
Glanzmann thrombasthenia (GT) is the most widely studied inherited disease of platelet function. Platelets fail to aggregate due to a defect in platelet-to-platelet attachment. The hemostatic plug fails to form and a moderate to severe bleeding diathesis results. Classically of autosomal recessive inheritance, GT is caused by defects within the ITGA2B and ITGB3 genes that encode the αIIbβ3 integrin expressed at high density on the platelet surface and also in intracellular pools. Activated αIIbβ3 acts as a receptor for fibrinogen and other adhesive proteins that hold platelets together in a thrombus. Over 50 years of careful clinical and biological investigation have provided important advances that have improved not only the quality of life of the patients but which have also contributed to an understanding of how αIIbβ3 functions. Despite major improvements in our knowledge of GT and its genetic causes, extensive biological and clinical variability with respect to the severity and intensity of bleeding remains poorly understood. I now scan the repertoire of ITGA2B and ITGB3 gene defects and highlight the wide genetic and biological heterogeneity within the type II and variant subgroups especially with regard to bleeding, clot retraction, the internal platelet Fg storage pool and the nature of the mutations causing the disease. I underline the continued importance of gene profiling and biological studies and emphasize the multifactorial etiology of the clinical expression of the disease. This is done in a manner to provide guidelines for future studies and future treatments of a disease that has not only aided research on rare diseases but also contributed to advances in antithrombotic therapy.
Collapse
Affiliation(s)
- Alan Nurden
- Institut Hospitalo-Universitaire LIRYC, Pessac, France
| |
Collapse
|
8
|
Inherited Platelet Disorders: An Updated Overview. Int J Mol Sci 2021; 22:ijms22094521. [PMID: 33926054 PMCID: PMC8123627 DOI: 10.3390/ijms22094521] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/17/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Platelets play a major role in hemostasis as ppwell as in many other physiological and pathological processes. Accordingly, production of about 1011 platelet per day as well as appropriate survival and functions are life essential events. Inherited platelet disorders (IPDs), affecting either platelet count or platelet functions, comprise a heterogenous group of about sixty rare diseases caused by molecular anomalies in many culprit genes. Their clinical relevance is highly variable according to the specific disease and even within the same type, ranging from almost negligible to life-threatening. Mucocutaneous bleeding diathesis (epistaxis, gum bleeding, purpura, menorrhagia), but also multisystemic disorders and/or malignancy comprise the clinical spectrum of IPDs. The early and accurate diagnosis of IPDs and a close patient medical follow-up is of great importance. A genotype-phenotype relationship in many IPDs makes a molecular diagnosis especially relevant to proper clinical management. Genetic diagnosis of IPDs has been greatly facilitated by the introduction of high throughput sequencing (HTS) techniques into mainstream investigation practice in these diseases. However, there are still unsolved ethical concerns on general genetic investigations. Patients should be informed and comprehend the potential implications of their genetic analysis. Unlike the progress in diagnosis, there have been no major advances in the clinical management of IPDs. Educational and preventive measures, few hemostatic drugs, platelet transfusions, thrombopoietin receptor agonists, and in life-threatening IPDs, allogeneic hematopoietic stem cell transplantation are therapeutic possibilities. Gene therapy may be a future option. Regular follow-up by a specialized hematology service with multidisciplinary support especially for syndromic IPDs is mandatory.
Collapse
|
9
|
Li D, Peng J, Li T, Liu Y, Chen M, Shi X. Itgb3-integrin-deficient mice may not be a sufficient model for patients with Glanzmann thrombasthenia. Mol Med Rep 2021; 23:449. [PMID: 33880575 PMCID: PMC8060805 DOI: 10.3892/mmr.2021.12088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/27/2021] [Indexed: 12/18/2022] Open
Abstract
Itgb3-integrin-deficient (Itgb3−/−) mice have been reported as a Glanzmann thrombasthenia (GT) model and have been used for platelet research. However, it remains unclear whether this mouse model can fully simulate patients with GT or whether it has different characteristics from these patients. The present study aimed to answer this question. Itgb3−/− mice were tested for platelet function, tail bleeding, whole-blood count, bone marrow hematopoiesis and organ enlargement. Itgb3−/− platelets showed impaired functions, including fibrinogen binding, aggregation, adhesion or spreading. Itgb3−/− mice demonstrated decreased platelet count and microcytic hypochromic anemia. Reduced iron staining of bone marrow and decreased plasma ferritin level confirmed the diagnosis of iron deficiency anemia. Evident splenomegaly was observed in Itgb3−/− mice. Immunohistochemical analysis of spleen biopsy revealed normal expression of CD3 and CD19, but elevated expression of CD71, which suggested that the splenomegaly in Itgb3−/− mice may be associated with extramedullary hematopoiesis. In conclusion, Itgb3−/− mice exhibited some unique characteristics that differed from those of human patients with GT and thus cannot completely simulate patients with GT.
Collapse
Affiliation(s)
- Dongya Li
- Department of Hematology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Jie Peng
- Department of Hematology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Tiantian Li
- Department of Hematology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Yichen Liu
- Department of Hematology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Min Chen
- Department of Hematology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Xiaofeng Shi
- Department of Hematology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| |
Collapse
|
10
|
Corneal proteome and differentially expressed corneal proteins in highly myopic chicks using a label-free SWATH-MS quantification approach. Sci Rep 2021; 11:5495. [PMID: 33750851 PMCID: PMC7943770 DOI: 10.1038/s41598-021-84904-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Myopia, or short-sightedness, is a highly prevalent refractive disorder in which the eye's focal length is too short for its axial dimension in its relaxed state. High myopia is associated with increased risks of blinding ocular complications and abnormal eye shape. In addition to consistent findings on posterior segment anomalies in high myopia (e.g., scleral remodeling), more recent biometric and biomechanical data in myopic humans and animal models also indicate anterior segment anomalies (e.g., corneal biomechanical properties). Because the cornea is the anterior-most ocular tissue, providing essential refractive power and physiological stability, it is important to understand the biochemical signaling pathway during myopia development. This study first aimed to establish the entire chicken corneal proteome. Then, using the classical form deprivation paradigm to induce high myopia in chicks, state-of-the-art bioinformatics technologies were applied to identify eight differentially expressed proteins in the highly myopic cornea. These results provide strong foundation for future corneal research, especially those using chicken as an animal model for myopia development.
Collapse
|
11
|
Abstract
Decades of preclinical and clinical studies developing gene therapy for hemophilia are poised to bear fruit with current promising pivotal studies likely to lead to regulatory approval. However, this recent success should not obscure the multiple challenges that were overcome to reach this destination. Gene therapy for hemophilia A and B benefited from advancements in the general gene therapy field, such as the development of adeno-associated viral vectors, as well as disease-specific breakthroughs, like the identification of B-domain deleted factor VIII and hyperactive factor IX Padua. The gene therapy field has also benefited from hemophilia B clinical studies, which revealed for the first time critical safety concerns related to immune responses to the vector capsid not anticipated in preclinical models. Preclinical studies have also investigated gene transfer approaches for other rare inherited bleeding disorders, including factor VII deficiency, von Willebrand disease, and Glanzmann thrombasthenia. Here we review the successful gene therapy journey for hemophilia and pose some unanswered questions. We then discuss the current state of gene therapy for these other rare inherited bleeding disorders and how the lessons of hemophilia gene therapy may guide clinical development.
Collapse
Affiliation(s)
- Valder R. Arruda
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Division of Hematology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Philadelphia, Pennsylvania
| | - Jesse Weber
- Department of Pediatrics, Division of Hematology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Benjamin J. Samelson-Jones
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Division of Hematology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Philadelphia, Pennsylvania
| |
Collapse
|
12
|
Multifaceted Functions of Platelets in Cancer: From Tumorigenesis to Liquid Biopsy Tool and Drug Delivery System. Int J Mol Sci 2020; 21:ijms21249585. [PMID: 33339204 PMCID: PMC7765591 DOI: 10.3390/ijms21249585] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
Platelets contribute to several types of cancer through plenty of mechanisms. Upon activation, platelets release many molecules, including growth and angiogenic factors, lipids, and extracellular vesicles, and activate numerous cell types, including vascular and immune cells, fibroblasts, and cancer cells. Hence, platelets are a crucial component of cell-cell communication. In particular, their interaction with cancer cells can enhance their malignancy and facilitate the invasion and colonization of distant organs. These findings suggest the use of antiplatelet agents to restrain cancer development and progression. Another peculiarity of platelets is their capability to uptake proteins and transcripts from the circulation. Thus, cancer-patient platelets show specific proteomic and transcriptomic expression patterns, a phenomenon called tumor-educated platelets (TEP). The transcriptomic/proteomic profile of platelets can provide information for the early detection of cancer and disease monitoring. Platelet ability to interact with tumor cells and transfer their molecular cargo has been exploited to design platelet-mediated drug delivery systems to enhance the efficacy and reduce toxicity often associated with traditional chemotherapy. Platelets are extraordinary cells with many functions whose exploitation will improve cancer diagnosis and treatment.
Collapse
|
13
|
Mesut Nezir Engin M. Bleeding Disorders Associated with Abnormal Platelets: Glanzmann Thrombasthenia and Bernard-Soulier Syndrome. Platelets 2020. [DOI: 10.5772/intechopen.93299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Platelets, the smallest cells in the blood, are associated with hemostasis, bowel formation, tissue remodeling, and wound healing. Although the prevalence of inherited platelet disorders is not fully known, it is a rare disease group and is encountered in approximately between 10000 and 1000000. Glanzmann thrombasthenia (GT) and Bernard-Soulier syndrome (BSS) are more frequently observed in inherited platelet disorders. In GT, the platelet aggregation stage due to deficiency or dysfunction of the platelet GPIIb/IIIa complex cannot take place. BSS is a platelet adhesion disorder due to the absence or abnormality of GPIb/IX complex on the platelet surface. If there is bleeding after easy bruising, mucous and oral cavities, menorrhagia, tooth extraction, tonsillectomy, or other surgical interventions, inherited platelet dysfunction should be considered if the platelet count is normal while the bleeding time is long. Firstly, other causes should be investigated by making differential diagnosis of GT and BSS. In this chapter, the definition, etiology, historical process, epidemiology, genetic basis, pathophysiology, clinical findings, diagnosis, differential diagnosis, and the follow-up and treatment approach of GT and BSS will be reviewed according to the current medical literature.
Collapse
|
14
|
Botero JP, Lee K, Branchford BR, Bray PF, Freson K, Lambert MP, Luo M, Mohan S, Ross JE, Bergmeier W, Di Paola J. Glanzmann thrombasthenia: genetic basis and clinical correlates. Haematologica 2020; 105:888-894. [PMID: 32139434 PMCID: PMC7109743 DOI: 10.3324/haematol.2018.214239] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/07/2020] [Indexed: 01/23/2023] Open
Abstract
Glanzmann thrombasthenia (GT) is an autosomal recessive disorder of platelet aggregation caused by quantitative or qualitative defects in integrins αIIb and β3. These integrins are encoded by the ITGA2B and ITGB3 genes and form platelet glycoprotein (GP)IIb/IIIa, which acts as the principal platelet receptor for fibrinogen. Although there is variability in the clinical phenotype, most patients present with severe mucocutaneous bleeding at an early age. A classic pattern of abnormal platelet aggregation, platelet glycoprotein expression and molecular studies confirm the diagnosis. Management of bleeding is based on a combination of hemostatic agents including recombinant activated factor VII with or without platelet transfusions and antifibrinolytic agents. Refractory bleeding and platelet alloimmunization are common complications. In addition, pregnant patients pose unique management challenges. This review highlights clinical and molecular aspects in the approach to patients with GT, with particular emphasis on the significance of multidisciplinary care.
Collapse
Affiliation(s)
- Juliana Perez Botero
- Versiti and Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kristy Lee
- Department of Genetics, University of North Carolina at Chapel Hill, NC, USA
| | - Brian R Branchford
- University of Colorado School of Medicine, Department of Pediatrics, Division of Hematology/Oncology/Bone Marrow Transplant, Aurora, CO, USA
| | - Paul F Bray
- Molecular Medicine Program, Division of Hematology and Hematologic Malignancies, Department of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Kathleen Freson
- Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| | - Michele P Lambert
- The Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Minjie Luo
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine, Philadelphia, PA, USA
| | - Shruthi Mohan
- Department of Genetics, University of North Carolina at Chapel Hill, NC, USA
| | - Justyne E Ross
- Department of Genetics, University of North Carolina at Chapel Hill, NC, USA
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics and UNC Blood Research Center, University of North Carolina at Chapel Hill, NC, USA
| | - Jorge Di Paola
- Division of Pediatric Hematology Oncology, Department of Pediatrics, Washington University School of Medicine in St. Louis, MO, USA
| | | |
Collapse
|
15
|
Abstract
Maternal alloimmunization to paternally inherited antigens on fetal/neonatal platelets can cause fetal/neonatal alloimmune thrombocytopenia (FNAIT) after antibody-mediated removal of platelets from the fetal circulation. The complications vary from mild bleeding symptoms to severe intracranial hemorrhage and subsequent neurological impairment or death. Studies on in vivo mechanisms are challenging to measure directly in pregnant women, rendering murine models as valuable and attractive alternatives, despite some critical differences between mice and men affecting the translational value. Here we present and discuss, the different murine models that substantially have increased our knowledge and understanding of FNAIT pathogenesis - as well as pre-clinical evaluation of therapeutic and preventive strategies.
Collapse
Affiliation(s)
- Trude Victoria Rasmussen
- Department of Laboratory Medicine, University Hospital of North Norway, Tromsø, Norway; Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Maria Therese Ahlen
- Department of Laboratory Medicine, University Hospital of North Norway, Tromsø, Norway.
| |
Collapse
|
16
|
Opneja A, Kapoor S, Stavrou EX. Contribution of platelets, the coagulation and fibrinolytic systems to cutaneous wound healing. Thromb Res 2019; 179:56-63. [PMID: 31078121 DOI: 10.1016/j.thromres.2019.05.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/14/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022]
Abstract
Wound healing is a complex process that consists of multiple phases, each of which are indispensable for adequate repair. Timely initiation and resolution of each of these phases namely, hemostasis, inflammation, proliferation and tissue remodeling, is critical for promoting healing and avoiding excess scar formation. While platelets have long been known to influence the healing process, other components of blood particularly coagulation factors and the fibrinolytic system also contribute to efficient wound repair. This review aims to summarize our current understanding of the role of platelets, the coagulation and fibrinolytic systems in cutaneous wound healing, with a focus on how these components communicate with immune and non-immune cells in the wound microenvironment. We also outline current and potential therapeutic strategies to improve the management of chronic, non-healing wounds.
Collapse
Affiliation(s)
- Aman Opneja
- Department of Medicine, Hematology and Oncology Division, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sargam Kapoor
- Department of Medicine, Hematology and Oncology Division, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Evi X Stavrou
- Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Medicine, Louis Stokes Veterans Administration Medical Center, VA Northeast Ohio Healthcare System, Cleveland, OH, USA.
| |
Collapse
|
17
|
Nurden AT. Acquired Glanzmann thrombasthenia: From antibodies to anti-platelet drugs. Blood Rev 2019; 36:10-22. [PMID: 31010659 DOI: 10.1016/j.blre.2019.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 02/06/2023]
Abstract
In contrast to the inherited platelet disorder given by mutations in the ITGA2B and ITGB3 genes, mucocutaneous bleeding from a spontaneous inhibition of normally expressed αIIbβ3 characterizes acquired Glanzmann thrombasthenia (GT). Classically, it is associated with autoantibodies or paraproteins that block platelet aggregation without causing a fall in platelet count. However, inhibitory antibodies to αIIbβ3 are widely associated with primary immune thrombocytopenia (ITP), occur in secondary ITP associated with leukemia and related disorders, solid cancers and myeloma, other autoimmune diseases, following organ transplantation while cytoplasmic dysregulation of αIIbβ3 function features in myeloproliferative and myelodysplastic syndromes. Antibodies to αIIbβ3 occur during viral and bacterial infections, while drug-dependent antibodies reacting with αIIbβ3 are a special case. Direct induction of acquired GT is a feature of therapies that block platelets in coronary artery disease. This review looks at these conditions, emphasizing molecular mechanisms, therapy, patient management and future directions for research.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut de Rhythmologie et de Modélisation Cardiaque, Plateforme Technologique d'Innovation Biomédicale, Hôpital Xavier Arnozan, Pessac, France.
| |
Collapse
|
18
|
Coller BS. Foreword: A Brief History of Ideas About Platelets in Health and Disease. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.09988-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
19
|
Wilcox DA. Gene Therapy for Platelet Disorders. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
20
|
Wang ZY, Ruan CG. [The progresses in research and treatment of inherited platelet disorders]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 39:877-880. [PMID: 30369214 PMCID: PMC7348291 DOI: 10.3760/cma.j.issn.0253-2727.2018.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Indexed: 11/05/2022]
|
21
|
Lee A, Poon MC. Inherited platelet functional disorders: General principles and practical aspects of management. Transfus Apher Sci 2018; 57:494-501. [PMID: 30031712 DOI: 10.1016/j.transci.2018.07.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Platelets are a critical component for effecting hemostasis and wound healing. Disorders affecting any platelet pathway mediating adhesion, activation, aggregation and procoagulant surface exposure can result in a bleeding diathesis. Specific diagnosis even with advanced techniques which are unavailable to most centers is often difficult. Inherited platelet function disorders therefore represent a heterogeneous and complex collection of disorders with a spectrum of bleeding severity, from relatively mild (and easily missed or misdiagnosed) to severe bleeding phenotype with salient diagnostic features. We advocate the use of bleeding assessment tools to help identification of patients and more importantly for assessment of individual patient bleeding phenotype to guide management decisions for treating and preventing bleeding. The complex management of these patients is best coordinated in a multidisciplinary comprehensive care clinic setting expert in managing bleeding disorders and associated complications, with particular attention to the physical and psychosocial health of patients and their families. Depending on the bleeding phenotype, the location and severity of bleeding, and the nature of an invasive procedure, available treatment modalities range from conservative measures using local pressure, topical thrombin, fibrin sealant, antifibrinolytics etc. to the use of systemic haemostatics such as desmopressin (DDAVP), platelets and recombinant human activated factor VII (rFVIIa). This review will provide opinions on the practical aspects and general management of inherited platelet function disorders, with discussion on the mechanism of action, and the pros and cons of various hemostatic agents. Finally, the prospect of curative treatment for patients with severe bleeding phenotype refractory to available treatments and with poor quality of life will be briefly discussed.
Collapse
Affiliation(s)
- Adrienne Lee
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Canada; Southern Alberta Rare Blood and Bleeding Disorders Comprehensive Care Program, Foothills Medical Centre, Alberta Health Services, Calgary, Canada.
| | - Man-Chiu Poon
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Canada; Department of Pediatric, Cumming School of Medicine, University of Calgary, Calgary, Canada; Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Canada; Southern Alberta Rare Blood and Bleeding Disorders Comprehensive Care Program, Foothills Medical Centre, Alberta Health Services, Calgary, Canada.
| |
Collapse
|
22
|
Nurden AT. Acquired Antibodies to αIIbβ3 in Glanzmann Thrombasthenia: From Transfusion and Pregnancy to Bone Marrow Transplants and Beyond. Transfus Med Rev 2018; 32:S0887-7963(18)30037-3. [PMID: 29884513 DOI: 10.1016/j.tmrv.2018.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/14/2018] [Accepted: 05/20/2018] [Indexed: 11/23/2022]
Abstract
Patients with the inherited bleeding disorder Glanzmann thrombasthenia (GT) possess platelets that lack αIIbβ3 integrin and fail to aggregate, and have moderate to severe mucocutaneous bleeding. Many become refractory to platelet transfusions due to the formation of isoantibodies to αIIbβ3 with the rapid elimination of donor platelets and/or a block of function. Epitope characterization has shown isoantibodies to be polyclonal and to recognize different epitopes on the integrin with β3 a major site and αvβ3 on endothelial and vascular cells a newly recognized target. Pregnancy in GT can also lead to isoantibody formation when fetal cells with β3 integrins pass into the circulation of a mother lacking them; a consequence is neonatal thrombocytopenia and a high risk of mortality. Antibody removal prior to donor transfusions can provide transient relief, but all evidence points to recombinant FVIIa as the first choice for GT patients either to stop bleeding or as prophylaxis. Promoting thrombin generation by rFVIIa favors GT platelet interaction with fibrin, and the risk of deep vein thrombosis also associated with prolonged immobilization and catheter use requires surveillance. Although having a high risk, allogeneic bone marrow transplantation associated with different stem cell sources and conditioning regimens has proved successful in many cases of severe GT with antibodies, and often, the associated conditioning and immunosuppressive therapy leads to loss of isoantibody production. Animal models of gene therapy for GT show promising results, but isoantibody production can be stimulated and CRISPR/Cas9 technology has yet to be applied. Up-to-date consensus protocols for dealing with isoantibodies in GT are urgently required, and networks providing patient care should be expanded.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut de Rhythmologie et de Modélisation Cardiaque, Plateforme Technologique d'Innovation Biomédicale, Hôpital Xavier Arnozan, Pessac, France.
| |
Collapse
|
23
|
Cao S, Anishkin A, Zinkevich NS, Nishijima Y, Korishettar A, Wang Z, Fang J, Wilcox DA, Zhang DX. Transient receptor potential vanilloid 4 (TRPV4) activation by arachidonic acid requires protein kinase A-mediated phosphorylation. J Biol Chem 2018; 293:5307-5322. [PMID: 29462784 DOI: 10.1074/jbc.m117.811075] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 01/24/2018] [Indexed: 11/06/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a Ca2+-permeable channel of the transient receptor potential (TRP) superfamily activated by diverse stimuli, including warm temperature, mechanical forces, and lipid mediators such as arachidonic acid (AA) and its metabolites. This activation is tightly regulated by protein phosphorylation carried out by various serine/threonine or tyrosine kinases. It remains poorly understood how phosphorylation differentially regulates TRPV4 activation in response to different stimuli. We investigated how TRPV4 activation by AA, an important signaling process in the dilation of coronary arterioles, is affected by protein kinase A (PKA)-mediated phosphorylation at Ser-824. Wildtype and mutant TRPV4 channels were expressed in human coronary artery endothelial cells (HCAECs). AA-induced TRPV4 activation was blunted in the S824A mutant but was enhanced in the phosphomimetic S824E mutant, whereas the channel activation by the synthetic agonist GSK1016790A was not affected. The low level of basal phosphorylation at Ser-824 was robustly increased by the redox signaling molecule hydrogen peroxide (H2O2). The H2O2-induced phosphorylation was accompanied by an enhanced channel activation by AA, and this enhanced response was largely abolished by PKA inhibition or S824A mutation. We further identified a potential structural context dependence of Ser-824 phosphorylation-mediated TRPV4 regulation involving an interplay between AA binding and the possible phosphorylation-induced rearrangements of the C-terminal helix bearing Ser-824. These results provide insight into how phosphorylation specifically regulates TRPV4 activation. Redox-mediated TRPV4 phosphorylation may contribute to pathologies associated with enhanced TRPV4 activity in endothelial and other systems.
Collapse
Affiliation(s)
- Sheng Cao
- From the Department of Medicine, Cardiovascular Center
| | - Andriy Anishkin
- Department of Biology, University of Maryland, College Park, Maryland 20742
| | - Natalya S Zinkevich
- From the Department of Medicine, Cardiovascular Center.,Department of Health and Medicine, Carroll University, Waukesha, Wisconsin 53186, and
| | | | | | - Zhihao Wang
- From the Department of Medicine, Cardiovascular Center
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226.,Children's Research Institute, The Children's Hospital of Wisconsin, Milwaukee, Wisconsin 53226
| | - David A Wilcox
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226.,Children's Research Institute, The Children's Hospital of Wisconsin, Milwaukee, Wisconsin 53226
| | - David X Zhang
- From the Department of Medicine, Cardiovascular Center,
| |
Collapse
|
24
|
Sauer K, Winkler B, Eyrich M, Schlegel PG, Wiegering V. Indication for allogeneic stem cell transplantation in Glanzmann’s thrombasthenia. Hamostaseologie 2017; 33:305-12. [DOI: 10.5482/hamo-12-08-0014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 06/28/2013] [Indexed: 11/05/2022] Open
Abstract
SummaryGlanzmann’s thrombasthenia (GT) is an autosomal recessive disorder characterized by a lack of thrombocyte aggregation due to the absence of thrombocyte glycoproteins IIb and αIIbβ3. The role of haematopoietic stem cell transplantation (HSCT) in GT remains controversial. However, HSCT offers the only curative approach for patients with a severe clinical phenotype.In this review, we will discuss the limitation of current status evidence and the specific risk of GT, in particular the alloimmunization and refractoriness to thrombocyte infusions. 19 successful HSCT in 18 GT type I patients have been reported. Mean age at transplantation was 5 years. All patients are still alive. The majority received sibling bone marrow transplant with busulfan and cyclophosphamid conditioning. GvHD incidence was within the normal range, but 10 patients showed alloimmunization of thrombocytes. Median follow up is 25 months.
Collapse
|
25
|
Poole LG, Massey VL, Siow DL, Torres-Gonzáles E, Warner NL, Luyendyk JP, Ritzenthaler JD, Roman J, Arteel GE. Plasminogen Activator Inhibitor-1 Is Critical in Alcohol-Enhanced Acute Lung Injury in Mice. Am J Respir Cell Mol Biol 2017; 57:315-323. [PMID: 28445073 PMCID: PMC5625219 DOI: 10.1165/rcmb.2016-0184oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 04/19/2017] [Indexed: 01/07/2023] Open
Abstract
Chronic alcohol exposure is a clinically important risk factor for the development of acute respiratory distress syndrome, the most severe form of acute lung injury (ALI). However, the mechanisms by which alcohol sensitizes the lung to development of this disease are poorly understood. We determined the role of the antifibrinolytic protein plasminogen activator inhibitor-1 (PAI-1) in alcohol enhancement of experimental endotoxin-induced ALI. Wild-type, PAI-1-/-, and integrin β3-/- mice were fed ethanol-containing Lieber-DeCarli liquid or a control diet for 6 weeks, followed by systemic LPS challenge. LPS administration triggered coagulation cascade activation as evidenced by increased plasma thrombin-antithrombin levels and pulmonary fibrin deposition. Ethanol-exposed animals showed enhanced PAI-1 expression and pulmonary fibrin deposition with coincident exaggeration of pulmonary inflammatory edematous injury. PAI-1 deficiency markedly reduced pulmonary fibrin deposition and greatly reduced inflammation and injury without impacting upstream coagulation. Interestingly, pulmonary platelet accumulation was effectively abolished by PAI-1 deficiency in ethanol/LPS-challenged mice. Moreover, mice lacking integrin αIIBβ3, the primary platelet receptor for fibrinogen, displayed a dramatic reduction in early inflammatory changes after ethanol/LPS challenge. These results indicate that the mechanism whereby alcohol exaggerates LPS-induced lung injury requires PAI-1-mediated pulmonary fibrin accumulation, and suggest a novel mechanism whereby alcohol contributes to inflammatory ALI by enhancing fibrinogen-platelet engagement.
Collapse
Affiliation(s)
- Lauren G. Poole
- Department of Pharmacology and Toxicology
- Alcohol Research Center, University of Louisville, Louisville, Kentucky; and
| | - Veronica L. Massey
- Department of Pharmacology and Toxicology
- Alcohol Research Center, University of Louisville, Louisville, Kentucky; and
| | - Deanna L. Siow
- Department of Pharmacology and Toxicology
- Alcohol Research Center, University of Louisville, Louisville, Kentucky; and
| | - Edilson Torres-Gonzáles
- Division of Pulmonary, Critical Care and Sleep Disorders Medicine, Department of Medicine, and
| | - Nikole L. Warner
- Department of Microbiology and Immunology, University of Louisville Health Sciences Center, Louisville, Kentucky
| | - James P. Luyendyk
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Jeffrey D. Ritzenthaler
- Division of Pulmonary, Critical Care and Sleep Disorders Medicine, Department of Medicine, and
| | - Jesse Roman
- Division of Pulmonary, Critical Care and Sleep Disorders Medicine, Department of Medicine, and
| | - Gavin E. Arteel
- Department of Pharmacology and Toxicology
- Alcohol Research Center, University of Louisville, Louisville, Kentucky; and
| |
Collapse
|
26
|
Poon MC, Di Minno G, Zotz R, d’Oiron R. Glanzmann’s thrombasthenia: strategies for identification and management. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1341306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Man-Chiu Poon
- Cumming School of Medicine, University of Calgary, Calgary, Canada
- Southern Alberta Rare Blood and Bleeding Disorders Comprehensive Care Program, Foothills Medical Centre, Alberta Health Services, Calgary, Canada
| | - Giovanni Di Minno
- Department of Clinical Medicine and Surgery, Regional Reference Center for Coagulation Disorders, Federico II University, Naples, Italy
| | - Rainer Zotz
- Institute for Laboratory Medicine, Blood Coagulation and Transfusion Medicine (LBT), Düsseldorf, Germany
- Department of Haemostasis, Haemotherapy and Transfusion Medicine, Heinrich Heine University Medical Centre, D-40225 Düsseldorf, Germany
| | - Roseline d’Oiron
- Centre for Haemophilia and Rare Congenital Bleeding Disorders, University Hospitals Paris-Sud, Le Kremlin-Bicêtre, France
| |
Collapse
|
27
|
Gresele P, Falcinelli E, Bury L. Inherited platelet function disorders. Diagnostic approach and management. Hamostaseologie 2016; 36:265-278. [PMID: 27484722 DOI: 10.5482/hamo-16-02-0002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 07/16/2016] [Indexed: 12/25/2022] Open
Abstract
Inherited platelet function disorders (IPFDs) make up a significant proportion of congenital bleeding diatheses, but they remain poorly understood and often difficult to diagnose. Therefore, a rational diagnostic approach, based on a standardized sequence of laboratory tests, with consecutive steps of increasing level of complexity, plays a crucial role in the diagnosis of most IPFDs. In this review we discuss a diagnostic approach through platelet phenotyping and genotyping and we give an overview of the options for the management of bleeding in these disorders and an account of the few systematic studies on the bleeding risk associated with invasive procedures and its treatment.
Collapse
Affiliation(s)
- Paolo Gresele
- Paolo Gresele, MD, PhD, Division of Internal and Cardiovascular Medicine Department of Medicine, University of Perugia, Via E. dal Pozzo, 06126 Perugia, Italy, Tel. +39/07 55 78 39 89, Fax +39/07 55 71 60 83, E-Mail:
| | | | | |
Collapse
|
28
|
Advances of blood cell-based drug delivery systems. Eur J Pharm Sci 2016; 96:115-128. [PMID: 27496050 DOI: 10.1016/j.ejps.2016.07.021] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/28/2016] [Accepted: 07/31/2016] [Indexed: 11/22/2022]
Abstract
Blood cells, including erythrocytes, leukocytes and platelets are used as drug carriers in a wide range of applications. They have many unique advantages such as long life-span in circulation (especially erythrocytes), target release capacities (especially platelets), and natural adhesive properties (leukocytes and platelets). These properties make blood cell based delivery systems, as well as their membrane-derived carriers, far superior to other drug delivery systems. Despite the advantages, the further development of blood cell-based delivery systems was hindered by limitations in the source, storage, and mass production. To overcome these problems, synthetic biomaterials that mimic blood cell and nanocrystallization of blood cells have been developed and may represent the future direction for blood cell membrane-based delivery systems. In this paper, we review recent progress of the rising blood cell-based drug delivery systems, and also discuss their challenges and future tendency of development.
Collapse
|
29
|
Dai B, Wu P, Xue F, Yang R, Yu Z, Dai K, Ruan C, Liu G, Newman PJ, Gao C. Integrin-αIIbβ3-mediated outside-in signalling activates a negative feedback pathway to suppress platelet activation. Thromb Haemost 2016; 116:918-930. [PMID: 27465472 DOI: 10.1160/th16-02-0096] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 07/04/2016] [Indexed: 12/22/2022]
Abstract
Integrin-αIIbβ3-mediated outside-in signalling is widely accepted as an amplifier of platelet activation; accumulating evidence suggests that outside-in signalling can, under certain conditions, also function as an inhibitor of platelet activation. The role of integrin-αIIbβ3-mediated outside-in signalling in platelet activation is disputable. We employed flow cytometry, aggregometry, immunoprecipitation, and immunoblotting to investigate the role of integrin-αIIbβ3-mediated outside-in signalling in platelet activation. Integrin αIIbβ3 inhibition enhances agonist-induced platelet ATP secretion. Human platelets lacking expression of αIIbβ3 exhibited more platelet ATP secretion than their wild-type counterparts. Moreover, integrin-αIIbβ3-mediated outside-in signals activate SHIP-1, which in turn mediates p-Akt dephosphorylation, leading to inactivation of PI3K/Akt signalling. Furthermore, 3AC (SHIP-1 inhibitor) inhibits platelet disaggregation, and promotes platelet ATP secretion. Upon ADP stimulation, Talin is recruited to αIIbβ3, and it is dissociated from αIIbβ3 when platelets disaggregate. In addition, treatment with RUC2, an inhibitor of αIIbβ3, which blocks αIIbβ3-mediated outside-in signalling, can markedly prevent the dissociation of talin from integrin. SHIP1 Inhibitor 3AC inhibits the dissociation of talin from integrin-β3. These results suggest that integrin-αIIbβ3-mediated outside-in signalling can serve as a brake to restrict unnecessary platelet activation by activated SHIP-1, which mediated the disassociation of talin from β3, leading to integrin inactivation and blocking of PI3K/Akt signalling to restrict platelet ATP secretion.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Peter J Newman
- Peter J. Newman, PhD, Blood Research Institute, Blood Center of Wisconsin, Milwaukee, 53711 WI, USA, E-mail:
| | - Cunji Gao
- Cunji Gao, PhD, Chronic Disease Research Institute, Department of Nutrition and Food Hygiene, Zhejiang University School of Public Health, 310058 Hangzhou, China, Tel: +86 571 882 066 00, Fax: +86 571 882 066 00, E-mail:
| |
Collapse
|
30
|
Glycogen synthase kinase 3β inhibition promotes human iTreg differentiation and suppressive function. Immunol Res 2016; 62:60-70. [PMID: 25752456 DOI: 10.1007/s12026-015-8635-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Induced regulatory T cells (iTregs) are essential to maintain immunological tolerance, immune homeostasis and prevention of autoimmunity. Some studies suggest that glycogen synthase kinase 3β (GSK3β) is involved in the mouse iTreg differentiation; however, whether GSK3β inhibits or enhances iTreg differentiation is still a matter of controversy. To address this issue, we have utilized human naïve CD4(+) T cells and investigated whether GSK3 activity changes during iTreg differentiation and whether altering GSK3 activity influences the development of iTregs and its suppressive function. As a constitutively activated kinase, during iTreg differentiation GSK3β became quickly deactivated (phosphorylated at serine 9), which is dependent on MAPK pathway rather than PI3-kinase/Akt pathway. Our results indicated that inhibition of GSK3β by specific inhibitors, SB216763 or TDZD-8, promoted the differentiation of iTreg and increased their suppressive activity. In contrast, overexpression of GSK3β significantly inhibited iTreg differentiation. Furthermore, GSK3β inhibition enhanced iTreg differentiation through the TGF-β/Smad3 pathway. Taken together, this study demonstrates that inhibition of GSK3β enhances human iTreg differentiation and its suppressive activity, and provides a rationale to target GSK3β as a novel immunotherapeutic strategy.
Collapse
|
31
|
New Insights Into the Treatment of Glanzmann Thrombasthenia. Transfus Med Rev 2016; 30:92-9. [PMID: 26968829 DOI: 10.1016/j.tmrv.2016.01.001] [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: 07/22/2015] [Revised: 01/12/2016] [Accepted: 01/12/2016] [Indexed: 11/21/2022]
Abstract
Glanzmann thrombasthenia (GT) is a rare inherited autosomal recessive bleeding disorder of platelet function caused by a quantitative or qualitative defect of platelet membrane glycoprotein IIb/IIIa (integrin αIIbβ3), a fibrinogen receptor required for platelet aggregation. Bleeds in GT are variable and may be severe and unpredictable. Bleeding not responsive to local and adjunctive measures, as well as surgical procedures, is treated with platelets, recombinant activated factor VII (rFVIIa), or antifibrinolytics, alone or in combination. Although platelets are the standard treatment for GT, their use is associated with the risk of blood-borne infection transmission and may also cause the development of platelet antibodies (to human leukocyte antigens and/or αIIbβ3), potentially resulting in platelet refractoriness. Currently, where rFVIIa is approved for use in GT, this is mostly for patients with platelet antibodies and/or a history of platelet refractoriness. However, data from the prospective Glanzmann's Thrombasthenia Registry (829 bleeds and 206 procedures in 218 GT patients) show that rFVIIa was frequently used in nonsurgical and surgical bleeds, with high efficacy rates, irrespective of platelet antibodies/refractoriness status. The mechanisms underpinning rFVIIa effectiveness in GT have been studied. At therapeutic concentrations, rFVIIa binds to activated platelets and directly activates FX to FXa, resulting in a burst of thrombin generation. Thrombin converts fibrinogen to fibrin and also enhances GT platelet adhesion and aggregation mediated by the newly converted (polymeric) fibrin, leading to primary hemostasis at the wound site. In addition, thrombin improves the final clot structure and activates thrombin-activatable fibrinolysis inhibitor to decrease clot lysis.
Collapse
|
32
|
Megakaryocyte- and megakaryocyte precursor-related gene therapies. Blood 2016; 127:1260-8. [PMID: 26787735 DOI: 10.1182/blood-2015-07-607937] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/30/2015] [Indexed: 01/27/2023] Open
Abstract
Hematopoietic stem cells (HSCs) can be safely collected from the body, genetically modified, and re-infused into a patient with the goal to express the transgene product for an individual's lifetime. Hematologic defects that can be corrected with an allogeneic bone marrow transplant can theoretically also be treated with gene replacement therapy. Because some genetic disorders affect distinct cell lineages, researchers are utilizing HSC gene transfer techniques using lineage-specific endogenous gene promoters to confine transgene expression to individual cell types (eg, ITGA2B for inherited platelet defects). HSCs appear to be an ideal target for platelet gene therapy because they can differentiate into megakaryocytes which are capable of forming several thousand anucleate platelets that circulate within blood vessels to establish hemostasis by repairing vascular injury. Platelets play an essential role in other biological processes (immune response, angiogenesis) as well as diseased states (atherosclerosis, cancer, thrombosis). Thus, recent advances in genetic manipulation of megakaryocytes could lead to new and improved therapies for treating a variety of disorders. In summary, genetic manipulation of megakaryocytes has progressed to the point where clinically relevant strategies are being developed for human trials for genetic disorders affecting platelets. Nevertheless, challenges still need to be overcome to perfect this field; therefore, strategies to increase the safety and benefit of megakaryocyte gene therapy will be discussed.
Collapse
|
33
|
Platelet-targeted gene therapy with human factor VIII establishes haemostasis in dogs with haemophilia A. Nat Commun 2014; 4:2773. [PMID: 24253479 PMCID: PMC3868233 DOI: 10.1038/ncomms3773] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 10/15/2013] [Indexed: 12/29/2022] Open
Abstract
It is essential to improve therapies for controlling excessive bleeding in patients with haemorrhagic disorders. As activated blood platelets mediate the primary response to vascular injury, we hypothesize that storage of coagulation Factor VIII within platelets may provide a locally inducible treatment to maintain haemostasis for haemophilia A. Here we show that haematopoietic stem cell gene therapy can prevent the occurrence of severe bleeding episodes in dogs with haemophilia A for at least 2.5 years after transplantation. We employ a clinically relevant strategy based on a lentiviral vector encoding the ITGA2B gene promoter, which drives platelet-specific expression of human FVIII permitting storage and release of FVIII from activated platelets. One animal receives a hybrid molecule of FVIII fused to the von Willebrand Factor propeptide-D2 domain that traffics FVIII more effectively into α-granules. The absence of inhibitory antibodies to platelet-derived FVIII indicates that this approach may have benefit in patients who reject FVIII replacement therapies. Thus, platelet FVIII may provide effective long-term control of bleeding in patients with haemophilia A.
Collapse
|
34
|
|
35
|
Nurden AT, Pillois X, Wilcox DA. Glanzmann thrombasthenia: state of the art and future directions. Semin Thromb Hemost 2013; 39:642-55. [PMID: 23929305 DOI: 10.1055/s-0033-1353393] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Glanzmann thrombasthenia (GT) is the principal inherited disease of platelets and the most commonly encountered disorder of an integrin. GT is characterized by spontaneous mucocutaneous bleeding and an exaggerated response to trauma caused by platelets that fail to aggregate when stimulated by physiologic agonists. GT is caused by quantitative or qualitative deficiencies of αIIbβ3, an integrin coded by the ITGA2B and ITGB3 genes and which by binding fibrinogen and other adhesive proteins joins platelets together in the aggregate. Widespread genotyping has revealed that mutations spread across both genes, yet the reason for the extensive variation in both the severity and intensity of bleeding between affected individuals remains poorly understood. Furthermore, although genetic defects of ITGB3 affect other tissues with β3 present as αvβ3 (the vitronectin receptor), the bleeding phenotype continues to dominate. Here, we look in detail at mutations that affect (i) the β-propeller region of the αIIb head domain and (ii) the membrane proximal disulfide-rich epidermal growth factor (EGF) domains of β3 and which often result in spontaneous integrin activation. We also examine deep vein thrombosis as an unexpected complication of GT and look at curative procedures for the diseases, including allogeneic stem cell transfer and the potential for gene therapy.
Collapse
Affiliation(s)
- Alan T Nurden
- Plateforme Technologique et d'Innovation Biomédicale, Hôpital Xavier Arnozan, Pessac, France.
| | | | | |
Collapse
|
36
|
Fang J, Nurden P, North P, Nurden AT, Du LM, Valentin N, Wilcox DA. C560Rβ3 caused platelet integrin αII b β3 to bind fibrinogen continuously, but resulted in a severe bleeding syndrome and increased murine mortality. J Thromb Haemost 2013; 11:1163-71. [PMID: 23551977 PMCID: PMC3702628 DOI: 10.1111/jth.12209] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 03/13/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVES β(3)-Deficient megakaryocytes were modified by human β(3)-lentivirus transduction and transplantation to express sufficient levels of a C560Rβ(3) amino acid substitution, for investigation of how an activated αII b β(3) conformation affects platelets in vivo in mice. PATIENT/METHODS As in our previous report of an R560β(3) mutation in a patient with Glanzmann thrombasthenia, R560β(3) murine platelets spontaneously bound antibody that only recognizes activated αII b β3 bound to its ligand, fibrinogen. RESULTS With this murine model, we showed that αII b -R560β3 mutation-mediated continuous binding of fibrinogen occurred in the absence of P-selectin surface expression, indicating that the integrin was in an active conformation, although the platelets circulated in a quiescent manner. Remarkably, only 35% of R560β(3) 'mutant' mice survived for 6 months after transplantation, whereas 87% of C560β(3) 'wild-type' mice remained alive. Pathologic examination revealed that R560β(3) mice had enlarged spleens with extramedullary hematopoiesis and increased hemosiderin, indicating hemorrhage. R560β(3) megakaryocytes and platelets showed abnormal morphology and irregular granule distribution. Interestingly, R560β(3) washed platelets could aggregate upon simultaneous addition of fibrinogen and physiologic agonists, but aggregation failed when platelets were exposed to fibrinogen before activation in vitro and in vivo. CONCLUSIONS The results demonstrate that continuous occupancy of αIIb β3 with fibrinogen disrupts platelet structure and function, leading to hemorrhagic death consistent with Glanzmann thrombasthenia rather than a thrombotic state.
Collapse
Affiliation(s)
- J Fang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Zheng X, Zinkevich NS, Gebremedhin D, Gauthier KM, Nishijima Y, Fang J, Wilcox DA, Campbell WB, Gutterman DD, Zhang DX. Arachidonic acid-induced dilation in human coronary arterioles: convergence of signaling mechanisms on endothelial TRPV4-mediated Ca2+ entry. J Am Heart Assoc 2013; 2:e000080. [PMID: 23619744 PMCID: PMC3698766 DOI: 10.1161/jaha.113.000080] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Arachidonic acid (AA) and/or its enzymatic metabolites are important lipid mediators contributing to endothelium-derived hyperpolarizing factor (EDHF)-mediated dilation in multiple vascular beds, including human coronary arterioles (HCAs). However, the mechanisms of action of these lipid mediators in endothelial cells (ECs) remain incompletely defined. In this study, we investigated the role of the transient receptor potential vanilloid 4 (TRPV4) channel in AA-induced endothelial Ca(2+) response and dilation of HCAs. METHODS AND RESULTS AA induced concentration-dependent dilation in isolated HCAs. The dilation was largely abolished by the TRPV4 antagonist RN-1734 and by inhibition of endothelial Ca(2+)-activated K(+) channels. In native and TRPV4-overexpressing human coronary artery ECs (HCAECs), AA increased intracellular Ca(2+) concentration ([Ca(2+)]i), which was mediated by TRPV4-dependent Ca(2+) entry. The AA-induced [Ca(2+)]i increase was inhibited by cytochrome P450 (CYP) inhibitors. Surprisingly, the CYP metabolites of AA, epoxyeicosatrienoic acids (EETs), were much less potent activators of TRPV4, and CYP inhibitors did not affect EET production in HCAECs. Apart from its effect on [Ca(2+)]i, AA induced endothelial hyperpolarization, and this effect was required for Ca(2+) entry through TRPV4. AA-induced and TRPV4-mediated Ca(2+) entry was also inhibited by the protein kinase A inhibitor PKI. TRPV4 exhibited a basal level of phosphorylation, which was inhibited by PKI. Patch-clamp studies indicated that AA activated TRPV4 single-channel currents in cell-attached and inside-out patches of HCAECs. CONCLUSIONS AA dilates HCAs through a novel mechanism involving endothelial TRPV4 channel-dependent Ca(2+) entry that requires endothelial hyperpolarization, PKA-mediated basal phosphorylation of TRPV4, and direct activation of TRPV4 channels by AA.
Collapse
Affiliation(s)
- Xiaodong Zheng
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wilcox DA. Gene Therapy for Platelet Disorders. Platelets 2013. [DOI: 10.1016/b978-0-12-387837-3.00064-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
39
|
Bugelski PJ, Martin PL. Concordance of preclinical and clinical pharmacology and toxicology of therapeutic monoclonal antibodies and fusion proteins: cell surface targets. Br J Pharmacol 2012; 166:823-46. [PMID: 22168282 PMCID: PMC3417412 DOI: 10.1111/j.1476-5381.2011.01811.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 10/14/2011] [Accepted: 11/28/2011] [Indexed: 12/20/2022] Open
Abstract
Monoclonal antibodies (mAbs) and fusion proteins directed towards cell surface targets make an important contribution to the treatment of disease. The purpose of this review was to correlate the clinical and preclinical data on the 15 currently approved mAbs and fusion proteins targeted to the cell surface. The principal sources used to gather data were: the peer reviewed Literature; European Medicines Agency 'Scientific Discussions'; and the US Food and Drug Administration 'Pharmacology/Toxicology Reviews' and package inserts (United States Prescribing Information). Data on the 15 approved biopharmaceuticals were included: abatacept; abciximab; alefacept; alemtuzumab; basiliximab; cetuximab; daclizumab; efalizumab; ipilimumab; muromonab; natalizumab; panitumumab; rituximab; tocilizumab; and trastuzumab. For statistical analysis of concordance, data from these 15 were combined with data on the approved mAbs and fusion proteins directed towards soluble targets. Good concordance with human pharmacodynamics was found for mice receiving surrogates or non-human primates (NHPs) receiving the human pharmaceutical. In contrast, there was poor concordance for human pharmacodynamics in genetically deficient mice and for human adverse effects in all three test systems. No evidence that NHPs have superior predictive value was found.
Collapse
Affiliation(s)
- Peter J Bugelski
- Biologics Toxicology, Janssen Research & Development, division of Johnson & Johnson Pharmaceutical Research & Development, LLC, Radnor, PA 19087, USA
| | | |
Collapse
|
40
|
Kanaji S, Kuether EL, Fahs SA, Schroeder JA, Ware J, Montgomery RR, Shi Q. Correction of murine Bernard-Soulier syndrome by lentivirus-mediated gene therapy. Mol Ther 2012; 20:625-32. [PMID: 22044935 PMCID: PMC3293608 DOI: 10.1038/mt.2011.231] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 09/28/2011] [Indexed: 12/29/2022] Open
Abstract
Bernard-Soulier syndrome (BSS) is an inherited bleeding disorder caused by a defect in the platelet glycoprotein (GP) Ib-IX-V complex. The main treatment for BSS is platelet transfusion but it is often limited to severe bleeding episodes or surgical interventions due to the risk of alloimmunization. We have previously reported successful expression of human GPIbα (hGPIbα) in human megakaryocytes using a lentiviral vector (LV) encoding human GP1BA under control of the platelet-specific integrin αIIb promoter (2bIbα). In this study, we examined the efficacy of this strategy for the gene therapy of BSS using GPIbα(null) as a murine model of BSS. GPIbα(null) hematopoietic stem cells (HSC) transduced with 2bIbα LV were transplanted into lethally irradiated GPIbα(null) littermates. Therapeutic levels of hGPIbα expression were achieved that corrected the tail bleeding time and improved the macrothrombocytopenia. Sequential bone marrow (BM) transplants showed sustained expression of hGPIbα with similar phenotypic correction. Antibody response to hGPIbα was documented in 1 of 17 total recipient mice but was tolerated without any further treatment. These results demonstrate that lentivirus-mediated gene transfer can provide sustained phenotypic correction of murine BSS, indicating that this approach may be a promising strategy for gene therapy of BSS patients.
Collapse
Affiliation(s)
- Sachiko Kanaji
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin, USA
| | - Erin L Kuether
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children's Research Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, USA
| | - Scot A Fahs
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jocelyn A Schroeder
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children's Research Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jerry Ware
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Robert R Montgomery
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children's Research Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, USA
| | - Qizhen Shi
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children's Research Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
41
|
Abstract
Although platelets are the smallest cells in the blood, they are implied in various processes ranging from immunology and oncology to thrombosis and hemostasis. Many large-scale screening programs, genome-wide association, and "omics" studies have generated lists of genes and loci that are probably involved in the formation or physiology of platelets under normal and pathologic conditions. This creates an increasing demand for new and improved model systems that allow functional assessment of the corresponding gene products in vivo. Such animal models not only render invaluable insight in the platelet biology, but in addition, provide improved test systems for the validation of newly developed anti-thrombotics. This review summarizes the most important models to generate transgenic platelets and to study their influence on platelet physiology in vivo. Here we focus on the zebrafish morpholino oligonucleotide technology, the (platelet-specific) knockout mouse, and the transplantation of genetically modified human or murine platelet progenitor cells in myelo-conditioned mice. The various strengths and pitfalls of these animal models are illustrated by recent examples from the platelet field. Finally, we highlight the latest developments in genetic engineering techniques and their possible application in platelet research.
Collapse
|
42
|
Abstract
Abstract
Inherited platelet disorders (IPDs) comprise a heterogenous group of diseases that include defects in platelet function and disordered megakaryopoiesis. Some IPDs overlap as both defects in function and thrombopoiesis, resulting in both altered aggregation and/or secretion and thrombocytopenia. This review examines the key features of the presentation of IPDs in children and adults and presents a diagnostic algorithm for the evaluation of these patients. In addition, recent advances in our understanding of the pathophysiology of platelet disorders are addressed, with attention given to some of the novel genetic associations. Finally, treatment options and future therapies are briefly discussed.
Collapse
|
43
|
Platelet gene therapy improves hemostatic function for integrin alphaIIbbeta3-deficient dogs. Proc Natl Acad Sci U S A 2011; 108:9583-8. [PMID: 21606353 DOI: 10.1073/pnas.1016394108] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Activated blood platelets mediate the primary response to vascular injury. Although molecular abnormalities of platelet proteins occur infrequently, taken collectively, an inherited platelet defect accounts for a bleeding diathesis in ≈1:20,000 individuals. One rare example of a platelet disorder, Glanzmann thrombasthenia (GT), is characterized by life-long morbidity and mortality due to molecular abnormalities in a major platelet adhesion receptor, integrin αIIbβ3. Transfusion therapy is frequently inadequate because patients often generate antibodies to αIIbβ3, leading to immune-mediated destruction of healthy platelets. In the most severe cases allogeneic bone marrow transplantation has been used, yet because of the risk of the procedure it has been limited to few patients. Thus, hematopoietic stem cell gene transfer was explored as a strategy to improve platelet function within a canine model for GT. Bleeding complications necessitated the use of a mild pretransplant conditioning regimen; therefore, in vivo drug selection was used to improve engraftment of autologously transplanted cells. Approximately 5,000 αIIbβ3 receptors formed on 10% of platelets. These modest levels allowed platelets to adhere to αIIbβ3's major ligand (fibrinogen), form aggregates, and mediate retraction of a fibrin clot. Remarkably, improved hemostatic function was evident, with ≤135-fold reduced blood loss, and improved buccal bleeding times decreased to 4 min for up to 5 y after transplant. One of four transplanted dogs developed a significant antibody response to αIIbβ3 that was attenuated effectively with transient immune suppression. These results indicate that gene therapy could become a practical approach for treating inherited platelet defects.
Collapse
|
44
|
Abstract
Extensive experimental evidence shows that platelets support tumour metastasis. The activation of platelets and the coagulation system have a crucial role in the progression of cancer. Within the circulatory system, platelets guard tumour cells from immune elimination and promote their arrest at the endothelium, supporting the establishment of secondary lesions. These contributions of platelets to tumour cell survival and spread suggest platelets as a new avenue for therapy.
Collapse
Affiliation(s)
- Laurie J Gay
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
45
|
Shi Q, Montgomery RR. Platelets as delivery systems for disease treatments. Adv Drug Deliv Rev 2010; 62:1196-203. [PMID: 20619307 DOI: 10.1016/j.addr.2010.06.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 06/29/2010] [Indexed: 12/23/2022]
Abstract
Platelets are small, anucleate, discoid shaped blood cells that play a fundamental role in hemostasis. Platelets contain a large number of biologically active molecules within cytoplasmic granules that are critical to normal platelet function. Because platelets circulate in blood through out the body, release biological molecules and mediators on demand and participate in hemostasis as well as many other pathophysiologic processes, targeting expression of proteins of interest to platelets and utilizing platelets as delivery systems for disease treatment would be a logical approach. This paper reviews the genetic therapy for inherited bleeding disorders utilizing platelets as delivery system, with a particular focus on platelet-derived FVIII for hemophilia A treatment.
Collapse
|
46
|
Gene therapy of canine leukocyte adhesion deficiency using lentiviral vectors with human CD11b and CD18 promoters driving canine CD18 expression. Mol Ther 2010; 19:113-21. [PMID: 20859258 DOI: 10.1038/mt.2010.203] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
To identify cellular promoters in a self-inactivating (SIN) lentiviral vector that might be beneficial in treating children with leukocyte adhesion deficiency type 1 (LAD-1), we tested lentiviral vectors with human CD11 and CD18 leukocyte integrin proximal promoter elements directing expression of canine CD18 in animals with canine LAD (CLAD). Lentiviral vectors with either the human CD11b (637 bp) proximal promoter or the human CD18 (1,060 bp) proximal promoter resulted in the highest percentages of CD18(+) CLAD CD34(+) cells in vitro. Subsequently, two CLAD dogs were infused with autologous CD34(+) cells transduced with the hCD11b (637 bp)-cCD18 vector, and two CLAD dogs were infused with autologous CD34(+) cells transduced with the hCD18 (1,060 bp)-cCD18 vector. Each dog received a nonmyeloablative dose of 200 cGy total body irradiation (TBI) before the infusion of transduced cells. The two CLAD dogs treated with the hCD18 (1,060 bp)-cCD18 vector, and one of the two dogs treated with the hCD11b (637 bp)-cCD18 vector, had reversal of the CLAD phenotype. These studies using endogenous leukocyte integrin proximal promoters represent an important step in the development of gene therapy for children with LAD-1.
Collapse
|
47
|
|
48
|
Jiang R, Xia Y, Li J, Deng L, Zhao L, Shi J, Wang X, Sun B. High expression levels of IKKalpha and IKKbeta are necessary for the malignant properties of liver cancer. Int J Cancer 2010; 126:1263-74. [PMID: 19728335 DOI: 10.1002/ijc.24854] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
IKK-NF-kappaB signaling is regarded as an important factor in hepatocarcinogenesis and a potential target for liver cancer therapy. Therefore, in this study, we analyzed the expression of mRNAs encoding components and targets of NF-kappaB signaling including IKKalpha, IKKbeta, RANK, RANKL, OPG, CyclinD3, mammary serine protease inhibitor (Maspin), CyclinD1, c-FLIP, Bcl-xl, Stat3, Cip1 and Cip2 by real-time PCR in 40 patients with liver cancer. After statistical analysis, 7 indices including IKKalpha, IKKbeta, RANK, Maspin, c-FLIP, Cip2 and cyclinD1 were found to show significant differences between tumor tissue and its corresponding adjacent tissue. When IKKalpha and IKKbeta were downregulated in the hepatocellular carcinoma (HCC) cell lines of MHCC-97L and MHCC-97H in vitro, the numbers of BrdU positive cells were decreased in both IKKalpha and IKKbeta knockdown cells. Levels of apoptosis were also investigated in IKKalpha and IKKbeta knockdown cells. The growth of HCC was inhibited in the subcutaneous implantation model, and lung metastatogenesis was also significantly inhibited in the kidney capsule transplantation model. Downregulation of IKKalpha and IKKbeta in HCC cultured in vitro revealed that increased Maspin, OPG and RANKL expression was associated with metastasis of HCC. These findings were associated with downregulation of Bcl-XL and c-FLIP, which may be the reason for increased apoptosis. The therapeutic effect of IKKalpha and IKKbeta downregulation depends on extent of NF-kappaB inhibition and the malignant nature of the HCC. We anticipate that IKK-targeted gene therapy can be used in the treatment of HCC, a cancer that is notoriously resistant to radiation and chemotherapy.
Collapse
Affiliation(s)
- Runqiu Jiang
- Liver Transplantation Center of the First Affiliated Hospital and Cancer Center, Nanjing Medical University, Nanjing, Jiangsu Province, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Affiliation(s)
- A T Nurden
- French National Reference Centre for Platelet Disorders, Hôpital Xavier Arnozan, Pessac, France.
| |
Collapse
|
50
|
Deng L, Li G, Xi L, Yin A, Gao Y, You W, Wang X, Sun B. Hepatitis B virus inhibition in mice by lentiviral vector mediated short hairpin RNA. BMC Gastroenterol 2009; 9:73. [PMID: 19804649 PMCID: PMC2765442 DOI: 10.1186/1471-230x-9-73] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Accepted: 10/06/2009] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Chronic hepatitis B virus (HBV) infection is an important cause of cirrhosis and hepatocellular carcinoma. The major challenges for current therapies are the low efficacy of current drugs and the occurrence of drug resistant HBV mutations. RNA interference (RNAi) of virus-specific genes offers the possibility of developing a new anti-HBV therapy. Recent reports have shown that lentiviral vectors based on HIV-1 are promising gene delivery vehicles due to their ability to integrate transgenes into non-dividing cells. Herein, a lentivirus-based RNAi system was developed to drive expression and delivery of HBV-specific short hairpin RNA (shRNA) in a mouse model for HBV replication. METHODS Hepatitis B surface antigen (HBsAg) and hepatitis B e antigen (HBeAg) in the sera of the mice were analyzed by quantitative sandwich enzyme linked immunosorbent assay (ELISA) technique, hepatitis B core antigen (HBcAg) and HBsAg in the livers of the mice were detected by immunohistochemical assay, HBV DNA and HBV mRNA were measured by fluorogenic quantitative polymerase chain reaction (FQ-PCR) and quantitative real-time PCR respectively. RESULTS Co-injection of HBV plasmids together with the lentivirus targeting HBV shRNA induced an RNAi response. Secreted HBsAg was reduced by 89% in mouse serum, and HBeAg was also significantly inhibited, immunohistochemical detection of HBcAg or HBsAg in the liver tissues also revealed substantial reduction. Lentiviral mediated shRNA caused a significant suppression in the levels of viral mRNA and DNA synthesis compared to the control group. CONCLUSION Lentivirus-based RNAi can be used to suppress HBV replication in vivo, it might become a potential therapeutic strategy for treating HBV and other viral infections.
Collapse
Affiliation(s)
- Lei Deng
- Liver Transplantation Center of the First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, PR China.
| | | | | | | | | | | | | | | |
Collapse
|