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.

Blood platelets play part in a myriad of processes, such as inflammation, tumor growth and metastasis, immunology and, of course, thrombosis and blood clotting where they provide a first and crucial line of defense against vascular injury, thus maintaining normal hemostasis.1,2  Primary hemostasis starts when platelets recognize a site of vascular injury where the subendothelial matrix is exposed, bind to collagen, and become activated.3  The subsequent rise in intracellular calcium triggers conformational changes in integrin receptors, degranulation, exposition of a procoagulant surface, and generation and release of secondary agonists resulting in a thrombus that will cover the site of injury and prevent further blood loss.4  Platelets are furthermore an important factor in thrombotic events, such as stroke and myocardial infarction.5  To identify more proteins regulating platelet function that may serve as new targets for the development of anti-thrombotics or in the prevention of bleeding, the platelet research community has seen the completion of several large-scale screening programs and the spectacular rise in the “platelet-omics” field. Several genome-wide association studies and subsequent meta-analysis in patients with coronary artery disease and healthy volunteers identified numerous genetic loci that are possibly involved in regulating platelet formation, count, volume, and function and might confer a risk for coronary artery disease.6-11  On the other hand, gene expression profiling of healthy volunteer platelets, in combination with comparative microarray analysis between in vitro differentiated megakaryocytes (MKs) and closely related cell types, established a comprehensive platelet transcriptome.6,12-19  Finally, advanced proteomics studies identified proteins of the platelet sheddome, secretome, interactome, kinome, and phosphoproteome potentially involved in platelet function.20  The overall result is a large number of newly identified gene products for which we are only beginning to understand their role in platelet formation and physiology.17,21-23  It seems reasonable to assume that our current knowledge about platelets is only the tip of the iceberg and that functional characterization of all these gene products will revolutionize our view on platelet function.

The study of these genes by classical molecular biologic means is hampered by the anucleate nature of the platelet and the physiologic alterations, possibly resulting from direct permeabilization, thus causing an experimental bias.24-26  This creates a growing demand for new techniques and model systems, which allow the study of genes in platelets in vivo. Recent advances in platelet functional genomics techniques are now galvanizing this field. This review discusses the latest animal models for in vivo generation of genetically modified platelets along with an extensive overview of current and future developments.

The zebrafish Danio rerio has only been introduced in 1999 in the platelet functional genomics field, when zebrafish thrombocytes were first described as the primary actors in teleost hemostasis sharing features with mammalian platelets and MKs in structure, formation, and function.27  On the one hand, zebrafish thrombocytes resemble mammalian MKs because they both are nucleated. Furthermore, transcription factors, such as GATA-1, Ets, and Fli1, are active in teleost thrombocytes during specific timeframes, hereby mimicking mammalian megakaryopoiesis. Remarkably, thrombocytes can also increase in size, another hallmark of mammalian MK maturation.28  On the other hand, thrombocytes share structural features with platelets, such as the open canalicular system and the presence of α and dense granules. Like mammalian platelets, D rerio thrombocytes also form pseudopodia-like extensions upon activation.27,29  Finally, along with the presence of most of the coagulation factors in D rerio,30,31  thrombocytes contain homologues of important mammalian platelet proteins, such as VWF, glycoprotein (GP) Ibα, GPIbβ, integrin αIIb, protease-associated receptors, cyclooxygenase, and putative ADP receptors.27,31-35  Taken together, these findings support the concept of the zebrafish thrombocyte as a suitable model for mammalian megakaryopoiesis and hemostasis.

Zebrafish have also gained interest as a model for mammalian hematopoiesis and hemostasis from a practical viewpoint: zebrafish are not only easy to house and raise in a laboratory environment, their high fecundicity along with the external fertilization and development of transparent embryos allows easy visual inspection of the major developmental processes, including organogenesis, hematopoiesis, and analysis of early disease-related phenotypes without the need for invasive imaging techniques. Furthermore, zebrafish survive up to 7 days without blood circulation via passive diffusion of oxygen, permitting the study of genes whose absence might result in severe vascular defects and early embryonic lethality in mammals.36  The latest assembly of the zebrafish genome combined with fast methods of genetic modification will furthermore push the identification of zebrafish orthologs of human (platelet) genes forward (Genome Reference Consortium; www.ncbi.nlm.nih.gov/projects/genome/assembly/grc/).37 

Studying thrombocyte function is now facilitated by the existence of certain transgenic zebrafish lines having fluorescently labeled thrombocytes or vasculature, allowing easy visualization.28,35  The observation and transplantation of 2 distinct populations of CD41-GFP–positive cells has recently led to identification of zebrafish hematopoietic stem cells (HSCs), thus permitting to screen for genes or proteins involved in thrombocyte formation, such as the zebrafish orthologue of the thrombopoietin receptor c-Mpl, MASTL kinase, and cleavage and polyadenylation specificity factor 1.35,38-40  Genetically modified zebrafish can be generated either randomly by exposure to ethylnitrosourea or by using morpholino technology (Figure 1).41-43  Exposure to ethylnitrosourea is a forward genetics approach that induces random mutations in the DNA of premeiotic germ cells at a rate of approximately 100 to 200 per fish, which after mating with unexposed zebrafish results in mutant heterozygotes and further inbreeding results in the isolation of the carriers of the mutations and eventually the mutations themselves (Table 1). Although this approach has led to several relevant disease models, such as the fade out mutant resembling the Hermansky-Pudlak syndrome phenotype, it is time-consuming because of the random nature of the mutations, which each have to be characterized and identified.44  Morpholino oligonucleotides (MOs), on the other hand, are short anti–sense RNA oligomers designed to bind to a specific mRNA at the translation initiation site or at an intron-exon boundary resulting in either translational inhibition or incorrect splicing yielding a nonfunctional protein (Figure 1). Classic MO technology involves microinjection of the oligomer in zebrafish embryos at the one-cell stage, leading to spreading throughout the body of the developing embryo and a whole-organism knockdown (Table 1). Although the life span of a typical MO is only 4 to 5 days, it allows direct assessment of platelet adhesion and visualization of the thrombus size in, for example, FeCl3- or laser-induced thrombosis models in the arterial and venous circulation (Figure 1).29,36  As such, well-known human disorders, such as von Willebrand disease and c-Mpl deficiency, could be successfully reproduced in D rerio.32,35 

Figure 1

Overview of in vivo systems for generating transgenic platelets. Experimental design to generate transgenic zebrafish embryos using MO technology (left), platelet-specific KO mice with the Cre-LoxP method (middle), or genetically modified human or murine platelets by transduction of hematopoietic progenitor cells followed by transplantation in myelo-conditioned recipient mice (right). (Figure was produced using Servier Medical Art; http://www.servier.com/servier-medical-art.) MK/Plt indicates megakaryocyte or platelet-specific promoter.

Figure 1

Overview of in vivo systems for generating transgenic platelets. Experimental design to generate transgenic zebrafish embryos using MO technology (left), platelet-specific KO mice with the Cre-LoxP method (middle), or genetically modified human or murine platelets by transduction of hematopoietic progenitor cells followed by transplantation in myelo-conditioned recipient mice (right). (Figure was produced using Servier Medical Art; http://www.servier.com/servier-medical-art.) MK/Plt indicates megakaryocyte or platelet-specific promoter.

Close modal
Table 1

In vivo systems for production of transgenic platelets

In vivo systems for production of transgenic platelets
In vivo systems for production of transgenic platelets

The real benefit of the zebrafish model lies mainly in its use as a rapid screening method for platelet genes with a hitherto unknown function. This is illustrated by a recent paper in which genetic screenings and association studies in healthy volunteers as well as patients with myocardial infarction suggested a putative role for LRRFIP1, which was next shown to be a positive regulator of thrombus formation in a zebrafish MO model. Further proteomic analysis confirmed a possible role for LRRFIP1 as a component of the platelet cytoskeleton where it can interact with 2 actin-remodeling proteins Flightless-1 and Drebrin.6  In addition, zebrafish are also used to identify genes responsible for certain pathologic phenotypes. Recently, an exome sequencing study published back to back with proteomic analysis and genomic DNA sequencing in a patient suggested that NBEAL2 is the causative gene for Gray platelet syndrome, which was then confirmed in zebrafish.23,45,46  In another study, genetic linkage analysis of 2 case reports suggested mutations in a 5 million-bp locus on chromosome 10p11-12, where a.o., the microtubule-associated Ser/Thr-like (MASTL) kinase gene, is located, as responsible for an autosomal dominant inherited thrombocytopenia.47,48  Transient MO knockdown of MASTL kinase in zebrafish indeed results in thrombocytopenia and correlates with decreased expression of c-mpl and itga2B, thus identifying the cause of the disease.39  Perhaps the power of D rerio in discovering and characterizing the function of a gene can be best illustrated by a study in which comparative whole-genome expression analysis of the major blood cells and MKs and erythroblasts resulted in identification of 279 MK-specific transcripts, which contained approximately 35 putative transmembrane proteins, 4 of which were knocked down in zebrafish resulting in modified kinetics of laser-induced thrombus formation that revealed a role of BAMBI and LRRC32 as positive regulators and DCBLD2 and ESAM as inhibitors of thrombus formation.16,17  The latter was confirmed in ESAM knockout (KO) mice, which also develop larger thrombi compared with controls, thus again providing strong evidence for the relevance of the zebrafish MO model as a reverse genetics screen for novel proteins involved in thrombus formation.49 

Although all the aforementioned studies used whole embryo micro-injections to deliver MOs, a recent development in the chemical structure has resulted in cell permeability of MOs.42  These so-called Vivo-MOs have already successfully been used to inhibit the function of VWF and αIIb in adult zebrafish thrombocytes thereby further expanding the potential of MO technology.32,50 

One of the major pitfalls of using MO technology is that it inevitably results in a transient knockdown and more importantly in a knockdown in all cells, thus resulting in a global instead of a thrombocyte-specific phenotype. Addition of a thrombocyte-specific promoter to the recently developed Cre-LoxP system in zebrafish can help to solve this problem. Finally, there is inevitably a relatively large evolutionary distance between mammals and fish, which results in severaldifferences between mammalian platelets and MKs on the one hand and zebrafish thrombocytes on the other, as illustrated by the similar yet different coagulation pathways in zebrafish and other vertebrates resulting from a genome duplication after the 2 evolutionary lineages had diverged.31  Nevertheless, these recent data clearly illustrate that D rerio can be used successfully as a high-throughput early screening method to establish the relevance of newly identified platelet proteins in thrombosis and hemostasis.

A classic approach to generate large amounts of genetically modified platelets is breeding mutant mice. These mice can be either knock-in or KO animals. The use of knock-in mice, expressing a transgene or mutant form of a target protein, has led to a better understanding of, for example, regulators of G-protein–mediated signaling in modulating platelet responsiveness and of protein kinase D2 in dense granule secretion.51,52  The generation of KO mice lacking one or more proteins is nevertheless a more common approach. However, it takes several generations of mouse crossing and genotyping before a stable genotype and phenotype is achieved (Table 1). The time-consuming and laborious nature is therefore the major limiting factor in the application of KO mouse models in basic and translational research. The benefit of mutant mice mainly lies in the fact that they serve as an unlimited source of uniformly modified platelets or platelet progenitors, which can either be used for in vitro studies (eg, in aggregation tests or flow chamber models) or for in vivo studies (eg, in thrombosis models).53  The technique has been widely applied to investigate the role of certain proteins in platelet functioning (eg, arrestin-2, kindlin-3, CalDAG-GEFI, and Orai1) and also to successfully reproduce several monogeneic platelet disorders, such as Glanzmann thrombasthenia (GT) and the Bernard-Soulier syndrome (BSS).54-58  The latter is one of the most extensively studied platelet disorders in KO mice.59-63  Data in KO mice not only confirmed that absence or dysfunction of the GPIbα or GPIbβ subunit, but not GPV, is sufficient to cause prolonged bleeding time and macrothrombocytopenia, as observed in human BSS patients, but in addition provides evidence for a role of the GPIb/V/IX complex during megakaryopoiesis upon detection of an abnormal demarcation membrane system and enlarged peripheral zone.59,60,62-64  Furthermore, the GPIbα−/− bleeding phenotype could be rescued by expression of wild-type human GPIbα in GPIbα−/− mice, thus establishing BSS as a potential candidate disease for gene therapy.63  A recent paper extends these findings by showing that lentiviral-mediated platelet-specific GPIbα expression in GPIbα−/− HSCs followed by transplantation in GPIbα−/− mice results in a correction of the tail bleeding time and significant amelioration of the macrothrombocytopenia.65 

A major limiting factor of the classic approach of whole-organism KO is the difficulty to ascribe the observed phenotype to the intended cell type and the possible generation of a lethal phenotype as seen with, for example, talin, filamin A, or the novel platelet receptor CLEC-2, which nevertheless resulted in the assignment of a role for platelets in embryonic development.66-68 

These problems can nowadays be circumvented quite elegantly using inducible or cell type specific promoters in combination with the Cre-loxP KO technique, allowing spatiotemporal KO or targeted expression in a particular cell type, thereby avoiding developmental effects or effects of gene knockdown in nontarget cell types, respectively. A frequently used inducible system consists of MX1-Cre-LoxP mice in which the target allele can be effectively eliminated by injection of polyinosinic-polycytidylic acid.69  Such an approach has led to the appreciation of the role of AML-1 and SCL in megakaryopoiesis, which had previously gone unnoticed because of embryonic lethality.70,71  More recently, the use of cell type-specific or tissue-specific promoters to control Cre cDNA has gained a lot of interest (Table 1; Figure 1).72,73  The most frequently used promoters are the Vav promoter, which is specific for hematopoiesis, the GATA-1 promoter specific for the erythrocytic/megakaryocytic lineage, and finally the platelet factor 4 (PF4) and αIIb promoters, which are both specific for the megakaryocytic lineage, although the latter also shows basal expression in HSCs and is active throughout the entire differentiation process contrary to PF4, which reaches maximal activity only in the later stages.72-75  To date, there is a growing interest in the PF4-Cre mice, through which transgenic mice with MK and platelets devoid of proteins, such as talin, vinculin, focal adhesion kinase, survivin, and STIM1, have been bred.18,76-80  One of the most elegant demonstrations of the potential of the Cre-LoxP method was by Tiedt et al who applied a constitutive, a hematopoiesis-specific, and an inducible promoter to generate transgenic mice with different levels of mutant JAK2-V617F relative to wild-type JAK2, thus for the first time providing evidence that the ratio of mutant to wild-type JAK2 is a determining factor in the development of either essential thrombocythemia or polycythemia vera.75 

Despite its broad applicability, the Cre-LoxP technique is not without pitfalls. A general concern is the existence of so-called endogenous pseudo-LoxP sites, which can be targeted by Cre as well.81  In addition, the choice of the promoter controlling the Cre gene is of critical importance in achieving the desired KO. First, as stated above, some promoters are only active during the later stages of megakaryopoiesis, thereby possibly missing out on an early effect of the target gene.82,83  Second, promoter specificity is not always fully restricted to the intended cell type as demonstrated by the leakiness of the erythrocyte/MK-specific GATA-1 promoter resulting in ectopic expression.84  Third, promoters driving expression of Cre recombinase should be strong enough to obtain complete DNA recombination to generate a complete KO organism and avoid mosaicism. Fourth, transgene expression from a 5′-promoter element can differ from the endogenous expression pattern depending on, for example, the length of the cloned fragment, as illustrated by the differential activity of αIIb promoter 5′ deletion mutants.85 

Finally, a general concern lies in the species differences between humans and mice as not all counterparts of human platelet proteins are represented in mice as is the case for the protease-associated receptors.18,19,86  In addition, a significant difference in the induction of disease models has been reported; for example, attempts to establish a mouse thrombotic thrombocytopenic purpura model showed that ADAMTS13 KO mice require an additional trigger with shigatoxin to produce a thrombocytopenic purpura phenotype.87  However, when taking proper care of these possible pitfalls, the Cre-LoxP method is a great tool to generate MK or platelet-specific KO mice as proven by the number of reports published over the past few years.

Several groups have focused on (xeno)transplantation of ex vivo genetically modified hematopoietic progenitor cells in murine recipients to rapidly generate a transgenic HSC transplantation model or in a gene therapy setting. In this model, mouse or human HSCs are modified by, for example, the introduction of foreign DNA on transduction with a lentiviral vector (LV), allowing stable integration of the transgene in the host genome (Figure 1).88,89  After transplantation into myelo-conditioned recipient mice, they will repopulate the bone marrow and give rise to all differentiated blood lineages (Table 1; Figure 1). One of the major drawbacks concerning this technique is the relatively low permissiveness of progenitor cells to LV, which results in transduction efficiencies ranging from 19% of the murine MK progeny successfully expressing a therapeutic transgene to 71% of mouse platelets expressing eGFP and from 16% of human CD34+ HSCs containing a GFP sequence to 62% of human platelets expressing eGFP.88,90-98  Transduction efficiencies can be improved, for example, using higher multiplicities of infection, however, with concomitant increased risk of insertional mutagenesis and lentiviral-mediated cytotoxicity. Therefore, other ways of improving transduction efficiency are currently being explored (see “Perspectives”).99,100  As shown with a GFP reporter gene, the activity of a given promoter can vary greatly depending on the cell type and even the source of human progenitor cells (bone marrow, cord, or peripheral blood); therefore, the choice of promoter driving transgene expression is of particular importance.88,93,94,101  Like Cre-LoxP KO mice, transplantation approaches use either ubiquitous or lineage-restricted promoters. Ubiquitous promoters, such as cellular polypeptide chain elongation factor 1α, human cytomegalovirus, phosphoglycerate kinase, ubiquitin C, and simian virus 40, have the obvious benefit of driving transgene expression in all blood lineages, albeit not necessarily at comparable levels among the different cell types.101  However, ubiquitous expression is not always desired as it may cause adverse effects or difficulties in assigning an observed effect to a certain cell type.102  Therefore, MK or platelet-specific promoters to down-regulate or drive expression of a target gene have been added to the transduction and transplantation toolbox. One of the most successfully used promoters is the MK specific αIIb promoter, which displays basal expression in HSCs and gets up-regulated during megakaryopoiesis reaching up to 100% expression in platelets, hereby allowing the effect of the genetic modification to be studied during all stages of platelet development.103,104  Alternatively, 2 late MK specific promoters (ie, the PF4 promoter, used to express FVIII variants in platelets in a therapeutic approach, and the GPIbα promoter) have been reported to restrict transgene expression to platelets.92-94,105,106  A comparative study by Lavenu-Bombled et al nevertheless demonstrated significantly lower expression levels throughout MK differentiation for a transgene under control of the GPIbα promoter compared with the αIIb promoter.93  Finally, the murine and human-Mpl promoter permits MK-restricted expression in differentiating murine BM cells. Surprisingly, the human c-Mpl promoter allowed higher expression levels compared with its murine counterpart but was still outweighed by the GPIbα promoter.92  Interestingly, in contrast to all previously described promoters, transgene expression under control of a c-Mpl promoter gradually declined during MK differentiation. Therefore, not only cell types but also the time frame of transgene expression is an important factor to take into consideration, especially when studying megakaryopoiesis and thrombopoiesis.

The main benefit of the transplantation approach lies in the fact that it is a less laborious and time-consuming process compared with the generation of mutant mice (Table 1). Therefore, this technique not only allows in-depth functional studies but also permits rapid screening of numerous genes using for instance libraries of short hairpin RNA (shRNA) in RNA interference (RNAi).107  It can furthermore provide information regarding the optimal conditions and transgenic protein levels required for correction or establishment of a certain phenotype, thereby paving the way for (human) gene therapy. One of the earliest disease candidates for gene therapy has been hemophilia A, which is caused by a deficiency in factor VIII (FVIII). A lot of work in this field has been performed by the groups of Robert Montgomery and David Wilcox in Milwaukee, who, using the αIIb promoter, were able to specifically target production and storage of FVIII to platelet α granules along with its carrier protein VWF, thereby promoting local release at the site of injury and establishing the platelet as a possible carrier vehicle for the delivery of therapeutical proteins.108  Moreover, in FVIII−/− mice, correction of the bleeding phenotype was shown even in the presence of inhibitory antibodies and in FVIII−/− mice, which had been previously immunized against FVIII.109,110  Similar studies have been performed on BM cells from integrin β3−/− mice presenting with a GT bleeding phenotype, where transgenic expressed human β3 efficiently paired with murine αIIb and restored platelet function in transplanted β3−/− mice.90  Using the same β3 construct, the platelet functionality of 2 GT patients could be restored after in vitro transduction of autologous CD34+ cells.111  Recently, hemostatic functions in αIIbβ3-deficient dogs presenting with a nearly identical phenotype to human GT patients could be restored, resulting in markedly decreased bleeding times and reduced blood loss for up to 5 years after transduction and transplantation of canine CD34+ HSCs with a LV containing the full-length αIIb subunit under control of the αIIb promoter.112  The successful long-term correction of the bleeding phenotype in such a large mammalian model paves the way toward gene therapy and will undoubtedly provide a wealth of information for future studies. This breakthrough illustrates the potential of transplantation-based studies in the gene therapy context. Other examples of this approach are reported in a variety of settings ranging from correction of aplastic anemia caused by deficiency in the TPO receptor c-Mpl to functional studies of the receptor VPAC1 and from rescue of the aforementioned BSS mouse to the establishment of platelet-mediated FIX gene therapy as a possible treatment for hemophilia B, thus spanning the research field from thrombopoiesis to primary hemostasis and coagulation.63,92,113,114 

Although the previous sections illustrated the potential of several model systems, they all remain limited by either interspecies differences, transient effects (eg, limited MO life span), safety issues (eg, the use of viral vectors), and the incompleteness of nucleic acid delivery or knockdown (all RNAi-based approaches). Therefore, the development of novel techniques and approaches is still ongoing and should ultimately result in model systems that allow safe, complete, and stable manipulation of organisms, platelets, and platelet progenitor cells in a short time, thus providing the researcher with a (transgenic) model that mimics human platelet behavior under normal and pathologic conditions as close as possible. Significant progress is currently being made in terms of (1) improved efficiency in the generation of transgenic animals and reducing off-target effects, (2) an improved safety profile of gene delivery techniques, and (3) more precisely targeted gene delivery.

A new powerful tool in genetic engineering comes from the field of nuclease technology in which 2 related yet distinct techniques are drawing a lot of attention these days. The first strategy involves zinc finger nuclease (ZFN) technology to achieve highly efficient and specific genetic engineering. These endonucleases contain a zinc finger domain, designed to recognize a specific DNA triplet sequence, and the nuclease domain of nonspecific FokI nuclease. Specificity comes from a combination of several zinc fingers to recognize a specific DNA sequence and has improved recently by the strict requirement of heterodimerization of the FokI domains, thus relying on 2 independent DNA-binding events. The ZFN is delivered to a target cell by standard lipofection methods, lentiviral transduction, or micro-injection of either DNA or mRNA depending on the target cell type. Its action results in a double-stranded break, which can then be repaired via nonhomologous end joining in the absence of donor DNA or via homology directed repair in the presence of donor DNA. The former process efficiently ligates double-stranded breaks in all eukaryotes but is prone to errors, thus leading to genetic disruption. Addition of a custom-designed homologous donor DNA can serve as a template to repair the double-stranded break and be exploited to mutate either specific nucleotides (gene correction) or even introduce a whole gene at a specific site (gene addition). This technique is currently successfully used to disrupt or replace genes in human and mouse embryonic stem cells, zebrafish, and rats to generate transgenic cells or whole organisms.115  A related technique uses transcription activator-like effector nucleases (TALEN) to silence a gene of interest. Again, the nuclease activity is provided by the FokI domain, but in this case specificity comes from transcription factor-like effectors, a family of virulence factors originally found in a genus of plant pathogens. Although the possible applications of TALENs are much the same as for ZFN, the TAL effectors recognize single nucleotides and not triplets, which significantly simplifies their design and broadens the target range to virtually any sequence. TALENS are the latest addition to the genetic engineering toolbox and have already been used successfully in gene disruption in human cells, somatic zebrafish cells, and in the generation of knockout rats.116-119  Nuclease technology in general can provide a complete transformation of the target cell, but in practice the applicability can be restricted by the local DNA structure with the presence of chromatin and local genetic complexity. Because of the novelty of ZFN and TALEN, development of algorithms for the design of DNA binding motifs and improvements in terms of specificity is still ongoing.

A more established technique to knock a target gene down is RNAi, which is achieved by introduction of either small interfering RNA (siRNA), short hairpin RNA (shRNA), or artificial micro-RNA in a target cell.94,120-126  The low transfection efficiency in combination with the altered platelet physiology on permeabilization hinders direct application of basic siRNA on platelets. The limited life span of siRNAs furthermore hampers the generation of transgenic platelets by genetic modification of progenitor cells.24-26,122  Alternatively, lentiviral delivery of shRNA has been used to stably modify HSCs and generate transgenic platelets.94  However, shRNA transcription from polymerase III promoters precludes platelet specific expression, thus possibly inducing off-target effects in other HSC-derived cell types. Moreover, several reports showed cytotoxic effects induced by shRNA.127-130  Therefore, artificial microRNA seem to be the preferred tool to knock down a target gene because they permit inducible or tissue specific expression from polymerase II promoters and have not been associated with any cytotoxic effects to date.129,131,132  Such a platelet specific approach, however, has currently not been reported on.

As mentioned previously, efficient delivery of DNA or RNA to a target cell is often a limiting factor in genetic engineering, functional genomics, and gene therapy studies. Therefore, efforts to improve the design of (lenti)viral vectors are still ongoing. An interesting development is the pseudotyping of LV with more specific envelope proteins or even cell-type specific envelopes by incorporating specific ligands or antibodies in the viral envelope, whereas another approach focuses on using various biomaterials as a means for more efficient gene delivery by, for example, tethering of viral particle and/or target cell.131,133,134  Taken together, this shows that there are several exciting new developments that can provide more efficient delivery or effectiveness of a transgene to and in a target cell.

A completely different delivery approach involves the use of (hyperactive) transposons as a means to stably introduce DNA in target (hematopoietic progenitor) cells.135-137  Although hyperactive sleeping beauty transposons have not yet achieved significantly higher transfection efficiencies than lentiviral vectors, they do have an improved safety profile avoiding viral particle-mediated cellular toxicity or immune responses, a reduced risk of insertional mutagenesis or oncogenesis. Furthermore, the application of different promoters, the discovery of potential enhancer elements, and the exploration of different transfection methods will possibly improve transgene integration and expression levels in the near future.

Finally, to further diminish the differences between small animal models and humans, xenotransplantation models and the use of humanized mice have attracted attention in the past decade (Table 1).138  The nonobese diabetic/severe combined immunodeficiency mice (NOD.CB17-Prkdcscid/J or NOD/SCID) already allowed stable engraftment of human CD34+ HSCs originating from either mobilized peripheral blood or umbilical cord blood.95,139,140  The presence of all blood cell types in the bone marrow was confirmed; and although MK represented only a small fraction, successful platelet production was achieved. In addition, these platelets not only get activated in response to known agonists but also incorporated in a (predominantly) mouse thrombus on perfusion of transplanted mouse peripheral blood over a collagen-coated surface.139  The current generation of immunodeficient mice (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ or NSG) with virtual absence of natural killer cell activity already allows higher engraftment; and with numerous other immunodeficient mouse strains still under development, it is likely to see a novel strain with even better production of human platelets emerging in the ensuing years.141  Combining this transplantation model with safer and improved delivery and genetic engineering systems can hopefully provide the research community with a small animal model in which transgenic human MKs and platelets can be successfully generated and used in either functional genomics studies, thus aiding in expanding our basic knowledge of platelets, their formation and behavior or as superior models of human pathologies that allow faster and better screening and testing of novel compounds acting to improve platelet production or survival, or to prevent bleeding or thrombosis.

T.T. is a PhD fellow of the Agency for Innovation by Science and Technology in Flanders. This work was further supported by KU Leuven (concerted action grant GOA 2009/829) and the Fund for Scientific Research, Flanders (grant FWO G.0564.08).

Contribution: T.T. designed and wrote the manuscript and created Figure 1; K.B. conceived and edited the manuscript and Figure 1; and H.D. critically reviewed the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Hans Deckmyn, Laboratory for Thrombosis Research, KU Leuven campus Kortrijk, E Sabbelaan 53, B-8500 Kortrijk, Belgium; e-mail: hans.deckmyn@kuleuven-kortrijk.be.

1
Broos
 
K
Feys
 
HB
De Meyer
 
SF
Vanhoorelbeke
 
K
Deckmyn
 
H
Platelets at work in primary hemostasis.
Blood Rev
2011
, vol. 
25
 
4
(pg. 
155
-
167
)
2
Leslie
 
M
Cell biology: beyond clotting: the powers of platelets.
Science
2010
, vol. 
328
 
5978
(pg. 
562
-
564
)
3
Nuyttens
 
BP
Thijs
 
T
Deckmyn
 
H
Broos
 
K
Platelet adhesion to collagen.
Thromb Res
2011
, vol. 
127
 
Suppl 2
(pg. 
S26
-
S29
)
4
Thijs
 
T
Nuyttens
 
BP
Deckmyn
 
H
Broos
 
K
Platelet physiology and antiplatelet agents.
Clin Chem Lab Med
2010
, vol. 
48
 
Suppl 1
(pg. 
S3
-
S13
)
5
Nieswandt
 
B
Pleines
 
I
Bender
 
M
Platelet adhesion and activation mechanisms in arterial thrombosis and ischaemic stroke.
J Thromb Haemost
2011
, vol. 
9
 
Suppl 1
(pg. 
92
-
104
)
6
Goodall
 
AH
Burns
 
P
Salles
 
I
, et al. 
Transcription profiling in human platelets reveals LRRFIP1 as a novel protein regulating platelet function.
Blood
2010
, vol. 
116
 
22
(pg. 
4646
-
4656
)
7
Meisinger
 
C
Prokisch
 
H
Gieger
 
C
, et al. 
A genome-wide association study identifies three loci associated with mean platelet volume.
Am J Hum Genet
2009
, vol. 
84
 
1
(pg. 
66
-
71
)
8
Samani
 
NJ
Erdmann
 
J
Hall
 
AS
, et al. 
Genomewide association analysis of coronary artery disease.
N Engl J Med
2007
, vol. 
357
 
5
(pg. 
443
-
453
)
9
Schunkert
 
H
Konig
 
IR
Kathiresan
 
S
, et al. 
Large-scale association analysis identifies 13 new susceptibility loci for coronary artery disease.
Nat Genet
2011
, vol. 
43
 
4
(pg. 
333
-
338
)
10
Soranzo
 
N
Spector
 
TD
Mangino
 
M
, et al. 
A genome-wide meta-analysis identifies 22 loci associated with eight hematological parameters in the HaemGen consortium.
Nat Genet
2009
, vol. 
41
 
11
(pg. 
1182
-
1190
)
11
Soranzo
 
N
Rendon
 
A
Gieger
 
C
, et al. 
A novel variant on chromosome 7q22.3 associated with mean platelet volume, counts, and function.
Blood
2009
, vol. 
113
 
16
(pg. 
3831
-
3837
)
12
Gnatenko
 
DV
Dunn
 
JJ
McCorkle
 
SR
, et al. 
Transcript profiling of human platelets using microarray and serial analysis of gene expression.
Blood
2003
, vol. 
101
 
6
(pg. 
2285
-
2293
)
13
Jones
 
CI
Bray
 
S
Garner
 
SF
, et al. 
A functional genomics approach reveals novel quantitative trait loci associated with platelet signaling pathways.
Blood
2009
, vol. 
114
 
7
(pg. 
1405
-
1416
)
14
McRedmond
 
JP
Park
 
SD
Reilly
 
DF
, et al. 
Integration of proteomics and genomics in platelets: a profile of platelet proteins and platelet-specific genes.
Mol Cell Proteomics
2004
, vol. 
3
 
2
(pg. 
133
-
144
)
15
Senis
 
YA
Tomlinson
 
MG
Garcia
 
A
, et al. 
A comprehensive proteomics and genomics analysis reveals novel transmembrane proteins in human platelets and mouse megakaryocytes including G6b-B, a novel immunoreceptor tyrosine-based inhibitory motif protein.
Mol Cell Proteomics
2007
, vol. 
6
 
3
(pg. 
548
-
564
)
16
Macaulay
 
IC
Tijssen
 
MR
Thijssen-Timmer
 
DC
, et al. 
Comparative gene expression profiling of in vitro differentiated megakaryocytes and erythroblasts identifies novel activatory and inhibitory platelet membrane proteins.
Blood
2007
, vol. 
109
 
8
(pg. 
3260
-
3269
)
17
O'Connor
 
MN
Salles
 
II
Cvejic
 
A
, et al. 
Functional genomics in zebrafish permits rapid characterization of novel platelet membrane proteins.
Blood
2009
, vol. 
113
 
19
(pg. 
4754
-
4762
)
18
Jurak Begonja
 
A
Hoffmeister
 
KM
Hartwig
 
JH
Falet
 
H
FlnA-null megakaryocytes prematurely release large and fragile platelets that circulate poorly.
Blood
2011
, vol. 
118
 
8
(pg. 
2285
-
2295
)
19
Rowley
 
JW
Oler
 
AJ
Tolley
 
ND
, et al. 
Genome-wide RNA-seq analysis of human and mouse platelet transcriptomes.
Blood
2011
, vol. 
118
 
14
(pg. 
e101
-
e111
)
20
Zufferey
 
A
Fontana
 
P
Reny
 
JL
Nolli
 
S
Sanchez
 
JC
Platelet proteomics [published online ahead of print October 18, 2011].
Mass Spectrom Rev
 
21
Dittrich
 
M
Birschmann
 
I
Mietner
 
S
, et al. 
Platelet protein interactions: map, signaling components, and phosphorylation groundstate.
Arterioscler Thromb Vasc Biol
2008
, vol. 
28
 
7
(pg. 
1326
-
1331
)
22
Watkins
 
NA
Gusnanto
 
A
de Bono
 
B
, et al. 
A HaemAtlas: characterizing gene expression in differentiated human blood cells.
Blood
2009
, vol. 
113
 
19
(pg. 
e1
-
e9
)
23
Albers
 
CA
Cvejic
 
A
Favier
 
R
, et al. 
Exome sequencing identifies NBEAL2 as the causative gene for gray platelet syndrome.
Nat Genet
2011
, vol. 
43
 
8
(pg. 
735
-
737
)
24
Feng
 
D
Crane
 
K
Rozenvayn
 
N
Dvorak
 
AM
Flaumenhaft
 
R
Subcellular distribution of 3 functional platelet SNARE proteins: human cellubrevin, SNAP-23, and syntaxin 2.
Blood
2002
, vol. 
99
 
11
(pg. 
4006
-
4014
)
25
Polgár
 
J
Chung
 
SH
Reed
 
GL
Vesicle-associated membrane protein 3 (VAMP-3) and VAMP-8 are present in human platelets and are required for granule secretion.
Blood
2002
, vol. 
100
 
3
(pg. 
1081
-
1083
)
26
Schraw
 
TD
Rutledge
 
TW
Crawford
 
GL
, et al. 
Granule stores from cellubrevin/VAMP-3 null mouse platelets exhibit normal stimulus-induced release.
Blood
2003
, vol. 
102
 
5
(pg. 
1716
-
1722
)
27
Jagadeeswaran
 
P
Sheehan
 
JP
Craig
 
FE
Troyer
 
D
Identification and characterization of zebrafish thrombocytes.
Br J Haematol
1999
, vol. 
107
 
4
(pg. 
731
-
738
)
28
Jagadeeswaran
 
P
Lin
 
S
Weinstein
 
B
Hutson
 
A
Kim
 
S
Loss of GATA1 and gain of FLI1 expression during thrombocyte maturation.
Blood Cells Mol Dis
2010
, vol. 
44
 
3
(pg. 
175
-
180
)
29
Gregory
 
M
Hanumanthaiah
 
R
Jagadeeswaran
 
P
Genetic analysis of hemostasis and thrombosis using vascular occlusion.
Blood Cells Mol Dis
2002
, vol. 
29
 
3
(pg. 
286
-
295
)
30
Jagadeeswaran
 
P
Zebrafish: a tool to study hemostasis and thrombosis.
Curr Opin Hematol
2005
, vol. 
12
 
2
(pg. 
149
-
152
)
31
Lang
 
MR
Gihr
 
G
Gawaz
 
MP
Muller
 
II
Hemostasis in Danio rerio: is the zebrafish a useful model for platelet research?
J Thromb Haemost
2010
, vol. 
8
 
6
(pg. 
1159
-
1169
)
32
Carrillo
 
M
Kim
 
S
Rajpurohit
 
SK
Kulkarni
 
V
Jagadeeswaran
 
P
Zebrafish von Willebrand factor.
Blood Cells Mol Dis
2010
, vol. 
45
 
4
(pg. 
326
-
333
)
33
Grosser
 
T
Yusuff
 
S
Cheskis
 
E
Pack
 
MA
FitzGerald
 
GA
Developmental expression of functional cyclooxygenases in zebrafish.
Proc Natl Acad Sci U S A
2002
, vol. 
99
 
12
(pg. 
8418
-
8423
)
34
Kim
 
S
Carrillo
 
M
Kulkarni
 
V
Jagadeeswaran
 
P
Evolution of primary hemostasis in early vertebrates.
PLoS One
2009
, vol. 
4
 
12
pg. 
e8403
 
35
Lin
 
HF
Traver
 
D
Zhu
 
H
, et al. 
Analysis of thrombocyte development in CD41-GFP transgenic zebrafish.
Blood
2005
, vol. 
106
 
12
(pg. 
3803
-
3810
)
36
Salles
 
II
O'Connor
 
MN
Thijssen-Timmer
 
DC
Broos
 
K
Deckmyn
 
H
Garcia
 
A
Senis
 
YA
Platelet functional genomics.
Platelet Proteomics: Principles, Analysis and Applications
2011
New York, NY
Wiley & Sons
37
Flicek
 
P
Amode
 
MR
Barrell
 
D
, et al. 
Ensembl 2011.
Nucleic Acids Res
2011
, vol. 
39
 (pg. 
D800
-
D806
Database issue
38
Bolli
 
N
Payne
 
EM
Rhodes
 
J
, et al. 
cpsf1 is required for definitive HSC survival in zebrafish.
Blood
2011
, vol. 
117
 
15
(pg. 
3996
-
4007
)
39
Johnson
 
HJ
Gandhi
 
MJ
Shafizadeh
 
E
, et al. 
In vivo inactivation of MASTL kinase results in thrombocytopenia.
Exp Hematol
2009
, vol. 
37
 
8
(pg. 
901
-
908
)
40
Ma
 
D
Zhang
 
J
Lin
 
HF
Italiano
 
J
Handin
 
RI
The identification and characterization of zebrafish hematopoietic stem cells.
Blood
2011
, vol. 
118
 
2
(pg. 
289
-
297
)
41
Driever
 
W
Fishman
 
MC
The zebrafish: heritable disorders in transparent embryos.
J Clin Invest
1996
, vol. 
97
 
8
(pg. 
1788
-
1794
)
42
Morcos
 
PA
Li
 
Y
Jiang
 
S
Vivo-Morpholinos: a non-peptide transporter delivers Morpholinos into a wide array of mouse tissues.
Biotechniques
2008
, vol. 
45
 
6
(pg. 
613
-
614
)
43
Nasevicius
 
A
Ekker
 
SC
Effective targeted gene ‘knockdown’ in zebrafish.
Nat Genet
2000
, vol. 
26
 
2
(pg. 
216
-
220
)
44
Bahadori
 
R
Rinner
 
O
Schonthaler
 
HB
, et al. 
The Zebrafish fade out mutant: a novel genetic model for Hermansky-Pudlak syndrome.
Invest Ophthalmol Vis Sci
2006
, vol. 
47
 
10
(pg. 
4523
-
4531
)
45
Gunay-Aygun
 
M
Falik-Zaccai
 
TC
Vilboux
 
T
, et al. 
NBEAL2 is mutated in gray platelet syndrome and is required for biogenesis of platelet alpha-granules.
Nat Genet
2011
, vol. 
43
 
8
(pg. 
732
-
734
)
46
Kahr
 
WH
Hinckley
 
J
Li
 
L
, et al. 
Mutations in NBEAL2, encoding a BEACH protein, cause gray platelet syndrome.
Nat Genet
2011
, vol. 
43
 
8
(pg. 
738
-
740
)
47
Drachman
 
JG
Jarvik
 
GP
Mehaffey
 
MG
Autosomal dominant thrombocytopenia: incomplete megakaryocyte differentiation and linkage to human chromosome 10.
Blood
2000
, vol. 
96
 
1
(pg. 
118
-
125
)
48
Savoia
 
A
Del
 
VM
Totaro
 
A
, et al. 
An autosomal dominant thrombocytopenia gene maps to chromosomal region 10p.
Am J Hum Genet
1999
, vol. 
65
 
5
(pg. 
1401
-
1405
)
49
Stalker
 
TJ
Wu
 
J
Morgans
 
A
, et al. 
Endothelial cell specific adhesion molecule (ESAM) localizes to platelet-platelet contacts and regulates thrombus formation in vivo.
J Thromb Haemost
2009
, vol. 
7
 
11
(pg. 
1886
-
1896
)
50
Kim
 
S
Radhakrishnan
 
UP
Rajpurohit
 
SK
Kulkarni
 
V
Jagadeeswaran
 
P
Vivo-Morpholino knockdown of alphaIIb: a novel approach to inhibit thrombocyte function in adult zebrafish.
Blood Cells Mol Dis
2010
, vol. 
44
 
3
(pg. 
169
-
174
)
51
Konopatskaya
 
O
Matthews
 
SA
Harper
 
MT
, et al. 
Protein kinase C mediates platelet secretion and thrombus formation through protein kinase D2.
Blood
2011
, vol. 
118
 
2
(pg. 
416
-
424
)
52
Signarvic
 
RS
Cierniewska
 
A
Stalker
 
TJ
, et al. 
RGS/Gi2alpha interactions modulate platelet accumulation and thrombus formation at sites of vascular injury.
Blood
2010
, vol. 
116
 
26
(pg. 
6092
-
6100
)
53
Whinna
 
HC
Overview of murine thrombosis models.
Thromb Res
2008
, vol. 
122
 
Suppl 1
(pg. 
S64
-
S69
)
54
Braun
 
A
Varga-Szabo
 
D
Kleinschnitz
 
C
, et al. 
Orai1 (CRACM1) is the platelet SOC channel and essential for pathological thrombus formation.
Blood
2009
, vol. 
113
 
9
(pg. 
2056
-
2063
)
55
Crittenden
 
JR
Bergmeier
 
W
Zhang
 
Y
, et al. 
CalDAG-GEFI integrates signaling for platelet aggregation and thrombus formation.
Nat Med
2004
, vol. 
10
 
9
(pg. 
982
-
986
)
56
Hodivala-Dilke
 
KM
McHugh
 
KP
Tsakiris
 
DA
, et al. 
Beta3-integrin-deficient mice are a model for Glanzmann thrombasthenia showing placental defects and reduced survival.
J Clin Invest
1999
, vol. 
103
 
2
(pg. 
229
-
238
)
57
Li
 
D
D'Angelo
 
L
Chavez
 
M
Woulfe
 
DS
Arrestin-2 differentially regulates PAR4 and ADP receptor signaling in platelets.
J Biol Chem
2011
, vol. 
286
 
5
(pg. 
3805
-
3814
)
58
Moser
 
M
Nieswandt
 
B
Ussar
 
S
Pozgajova
 
M
Fassler
 
R
Kindlin-3 is essential for integrin activation and platelet aggregation.
Nat Med
2008
, vol. 
14
 
3
(pg. 
325
-
330
)
59
Kanaji
 
T
Russell
 
S
Cunningham
 
J
, et al. 
Megakaryocyte proliferation and ploidy regulated by the cytoplasmic tail of glycoprotein Ibalpha.
Blood
2004
, vol. 
104
 
10
(pg. 
3161
-
3168
)
60
Poujol
 
C
Ware
 
J
Nieswandt
 
B
Nurden
 
AT
Nurden
 
P
Absence of GPIbalpha is responsible for aberrant membrane development during megakaryocyte maturation: ultrastructural study using a transgenic model.
Exp Hematol
2002
, vol. 
30
 
4
(pg. 
352
-
360
)
61
Strassel
 
C
Nonne
 
C
Eckly
 
A
, et al. 
Decreased thrombotic tendency in mouse models of the Bernard-Soulier syndrome.
Arterioscler Thromb Vasc Biol
2007
, vol. 
27
 
1
(pg. 
241
-
247
)
62
Strassel
 
C
Eckly
 
A
Leon
 
C
, et al. 
Intrinsic impaired proplatelet formation and microtubule coil assembly of megakaryocytes in a mouse model of Bernard-Soulier syndrome.
Haematologica
2009
, vol. 
94
 
6
(pg. 
800
-
810
)
63
Ware
 
J
Russell
 
S
Ruggeri
 
ZM
Generation and rescue of a murine model of platelet dysfunction: the Bernard-Soulier syndrome.
Proc Natl Acad Sci U S A
2000
, vol. 
97
 
6
(pg. 
2803
-
2808
)
64
Poujol
 
C
Ramakrishnan
 
V
DeGuzman
 
F
, et al. 
Ultrastructural analysis of megakaryocytes in GPV knockout mice.
Thromb Haemost
2000
, vol. 
84
 
2
(pg. 
312
-
318
)
65
Kanaji
 
S
Kuether
 
EL
Fahs
 
SA
, et al. 
Correction of murine Bernard-Soulier syndrome by lentivirus-mediated gene therapy [published online ahead of print, November 1, 2011].
Mol Ther
 
66
Hart
 
AW
Morgan
 
JE
Schneider
 
J
, et al. 
Cardiac malformations and midline skeletal defects in mice lacking filamin A.
Hum Mol Genet
2006
, vol. 
15
 
16
(pg. 
2457
-
2467
)
67
Monkley
 
SJ
Zhou
 
XH
Kinston
 
SJ
, et al. 
Disruption of the talin gene arrests mouse development at the gastrulation stage.
Dev Dyn
2000
, vol. 
219
 
4
(pg. 
560
-
574
)
68
Suzuki-Inoue
 
K
Inoue
 
O
Ding
 
G
, et al. 
Essential in vivo roles of the C-type lectin receptor CLEC-2: embryonic/neonatal lethality of CLEC-2-deficient mice by blood/lymphatic misconnections and impaired thrombus formation of CLEC-2-deficient platelets.
J Biol Chem
2010
, vol. 
285
 
32
(pg. 
24494
-
24507
)
69
Kühn
 
R
Schwenk
 
F
Aguet
 
M
Rajewsky
 
K
Inducible gene targeting in mice.
Science
1995
, vol. 
269
 
5229
(pg. 
1427
-
1429
)
70
Hall
 
MA
Curtis
 
DJ
Metcalf
 
D
, et al. 
The critical regulator of embryonic hematopoiesis, SCL, is vital in the adult for megakaryopoiesis, erythropoiesis, and lineage choice in CFU-S12.
Proc Natl Acad Sci U S A
2003
, vol. 
100
 
3
(pg. 
992
-
997
)
71
Ichikawa
 
M
Asai
 
T
Saito
 
T
, et al. 
AML-1 is required for megakaryocytic maturation and lymphocytic differentiation, but not for maintenance of hematopoietic stem cells in adult hematopoiesis.
Nat Med
2004
, vol. 
10
 
3
(pg. 
299
-
304
)
72
Nowakowski
 
A
Alonso-Martin
 
S
Arias-Salgado
 
EG
, et al. 
Megakaryocyte gene targeting mediated by restricted expression of recombinase Cre.
Thromb Haemost
2011
, vol. 
105
 
1
(pg. 
138
-
144
)
73
Tiedt
 
R
Schomber
 
T
Hao-Shen
 
H
Skoda
 
RC
Pf4-Cre transgenic mice allow the generation of lineage-restricted gene knockouts for studying megakaryocyte and platelet function in vivo.
Blood
2007
, vol. 
109
 
4
(pg. 
1503
-
1506
)
74
Jasinski
 
M
Keller
 
P
Fujiwara
 
Y
Orkin
 
SH
Bessler
 
M
GATA1-Cre mediates Piga gene inactivation in the erythroid/megakaryocytic lineage and leads to circulating red cells with a partial deficiency in glycosyl phosphatidylinositol-linked proteins (paroxysmal nocturnal hemoglobinuria type II cells).
Blood
2001
, vol. 
98
 
7
(pg. 
2248
-
2255
)
75
Tiedt
 
R
Hao-Shen
 
H
Sobas
 
MA
, et al. 
Ratio of mutant JAK2-V617F to wild-type Jak2 determines the MPD phenotypes in transgenic mice.
Blood
2008
, vol. 
111
 
8
(pg. 
3931
-
3940
)
76
Bhavaraju
 
K
Lakhani
 
PR
Dorsam
 
RT
, et al. 
G(12/13) signaling pathways substitute for integrin alphaIIbbeta3-signaling for thromboxane generation in platelets.
PLoS One
2011
, vol. 
6
 
2
pg. 
e16586
 
77
Mitsios
 
JV
Prevost
 
N
Kasirer-Friede
 
A
, et al. 
What is vinculin needed for in platelets?
J Thromb Haemost
2010
, vol. 
8
 
10
(pg. 
2294
-
2304
)
78
Petrich
 
BG
Marchese
 
P
Ruggeri
 
ZM
, et al. 
Talin is required for integrin-mediated platelet function in hemostasis and thrombosis.
J Exp Med
2007
, vol. 
204
 
13
(pg. 
3103
-
3111
)
79
Wen
 
Q
Leung
 
C
Huang
 
Z
, et al. 
Survivin is not required for the endomitotic cell cycle of megakaryocytes.
Blood
2009
, vol. 
114
 
1
(pg. 
153
-
156
)
80
Ahmad
 
F
Boulaftali
 
Y
Greene
 
TK
, et al. 
Relative contributions of stromal interaction molecule 1 and CalDAG-GEFI to calcium-dependent platelet activation and thrombosis.
J Thromb Haemost
2011
, vol. 
9
 
10
(pg. 
2077
-
2086
)
81
Thyagarajan
 
B
Guimaraes
 
MJ
Groth
 
AC
Calos
 
MP
Mammalian genomes contain active recombinase recognition sites.
Gene
2000
, vol. 
244
 
1-2
(pg. 
47
-
54
)
82
Lepage
 
A
Leboeuf
 
M
Cazenave
 
JP
, et al. 
The alpha(IIb)beta(3) integrin and GPIb-V-IX complex identify distinct stages in the maturation of CD34(+) cord blood cells to megakaryocytes.
Blood
2000
, vol. 
96
 
13
(pg. 
4169
-
4177
)
83
Szalai
 
G
LaRue
 
AC
Watson
 
DK
Molecular mechanisms of megakaryopoiesis.
Cell Mol Life Sci
2006
, vol. 
63
 
21
(pg. 
2460
-
2476
)
84
Falet
 
H
Pollitt
 
AY
Begonja
 
AJ
, et al. 
A novel interaction between FlnA and Syk regulates platelet ITAM-mediated receptor signaling and function.
J Exp Med
2010
, vol. 
207
 
9
(pg. 
1967
-
1979
)
85
Prandini
 
MH
Uzan
 
G
Martin
 
F
Thevenon
 
D
Marguerie
 
G
Characterization of a specific erythromegakaryocytic enhancer within the glycoprotein IIb promoter.
J Biol Chem
1992
, vol. 
267
 
15
(pg. 
10370
-
10374
)
86
Denis
 
CV
Wagner
 
DD
Platelet adhesion receptors and their ligands in mouse models of thrombosis.
Arterioscler Thromb Vasc Biol
2007
, vol. 
27
 
4
(pg. 
728
-
739
)
87
Motto
 
DG
Chauhan
 
AK
Zhu
 
G
, et al. 
Shigatoxin triggers thrombotic thrombocytopenic purpura in genetically susceptible ADAMTS13-deficient mice.
J Clin Invest
2005
, vol. 
115
 
10
(pg. 
2752
-
2761
)
88
Salmon
 
P
Kindler
 
V
Ducrey
 
O
, et al. 
High-level transgene expression in human hematopoietic progenitors and differentiated blood lineages after transduction with improved lentiviral vectors.
Blood
2000
, vol. 
96
 
10
(pg. 
3392
-
3398
)
89
Wu
 
MH
Smith
 
SL
Dolan
 
ME
High efficiency electroporation of human umbilical cord blood CD34+ hematopoietic precursor cells.
Stem Cells
2001
, vol. 
19
 
6
(pg. 
492
-
499
)
90
Fang
 
J
Hodivala-Dilke
 
K
Johnson
 
BD
, et al. 
Therapeutic expression of the platelet-specific integrin, alphaIIbbeta3, in a murine model for Glanzmann thrombasthenia.
Blood
2005
, vol. 
106
 
8
(pg. 
2671
-
2679
)
91
Gao
 
Z
Golob
 
J
Tanavde
 
VM
, et al. 
High levels of transgene expression following transduction of long-term NOD/SCID-repopulating human cells with a modified lentiviral vector.
Stem Cells
2001
, vol. 
19
 
3
(pg. 
247
-
259
)
92
Heckl
 
D
Wicke
 
DC
Brugman
 
MH
, et al. 
Lentiviral gene transfer regenerates hematopoietic stem cells in a mouse model for Mpl-deficient aplastic anemia.
Blood
2011
, vol. 
117
 
14
(pg. 
3737
-
3747
)
93
Lavenu-Bombled
 
C
Izac
 
B
Legrand
 
F
, et al. 
Glycoprotein Ibalpha promoter drives megakaryocytic lineage-restricted expression after hematopoietic stem cell transduction using a self-inactivating lentiviral vector.
Stem Cells
2007
, vol. 
25
 
6
(pg. 
1571
-
1577
)
94
Ohmori
 
T
Kashiwakura
 
Y
Ishiwata
 
A
, et al. 
Silencing of a targeted protein in in vivo platelets using a lentiviral vector delivering short hairpin RNA sequence.
Arterioscler Thromb Vasc Biol
2007
, vol. 
27
 
10
(pg. 
2266
-
2272
)
95
Piacibello
 
W
Bruno
 
S
Sanavio
 
F
, et al. 
Lentiviral gene transfer and ex vivo expansion of human primitive stem cells capable of primary, secondary, and tertiary multilineage repopulation in NOD/SCID mice: nonobese diabetic/severe combined immunodeficient.
Blood
2002
, vol. 
100
 
13
(pg. 
4391
-
4400
)
96
Shi
 
Q
Wilcox
 
DA
Morateck
 
PA
, et al. 
Targeting platelet GPIbalpha transgene expression to human megakaryocytes and forming a complete complex with endogenous GPIbbeta and GPIX.
J Thromb Haemost
2004
, vol. 
2
 
11
(pg. 
1989
-
1997
)
97
Tesio
 
M
Gammaitoni
 
L
Gunetti
 
M
, et al. 
Sustained long-term engraftment and transgene expression of peripheral blood CD34+ cells transduced with third-generation lentiviral vectors.
Stem Cells
2008
, vol. 
26
 
6
(pg. 
1620
-
1627
)
98
Woods
 
NB
Fahlman
 
C
Mikkola
 
H
, et al. 
Lentiviral gene transfer into primary and secondary NOD/SCID repopulating cells.
Blood
2000
, vol. 
96
 
12
(pg. 
3725
-
3733
)
99
Sinn
 
PL
Sauter
 
SL
McCray
 
PB
Gene therapy progress and prospects: development of improved lentiviral and retroviral vectors—design, biosafety, and production.
Gene Ther
2005
, vol. 
12
 
14
(pg. 
1089
-
1098
)
100
Woods
 
NB
Muessig
 
A
Schmidt
 
M
, et al. 
Lentiviral vector transduction of NOD/SCID repopulating cells results in multiple vector integrations per transduced cell: risk of insertional mutagenesis.
Blood
2003
, vol. 
101
 
4
(pg. 
1284
-
1289
)
101
Varma
 
NR
Janic
 
B
Ali
 
MM
Iskander
 
A
Arbab
 
AS
Lentiviral based gene transduction and promoter studies in human hematopoietic stem cells (hHSCs).
J Stem Cells Regen Med
2011
, vol. 
7
 
1
(pg. 
41
-
53
)
102
Cocault
 
L
Bouscary
 
D
Le Bousse
 
KC
, et al. 
Ectopic expression of murine TPO receptor (c-mpl) in mice is pathogenic and induces erythroblastic proliferation.
Blood
1996
, vol. 
88
 
5
(pg. 
1656
-
1665
)
103
Heidenreich
 
R
Eisman
 
R
Surrey
 
S
, et al. 
Organization of the gene for platelet glycoprotein IIb.
Biochemistry
1990
, vol. 
29
 
5
(pg. 
1232
-
1244
)
104
Wilcox
 
DA
Olsen
 
JC
Ishizawa
 
L
Griffith
 
M
White
 
GC
Integrin alphaIIb promoter-targeted expression of gene products in megakaryocytes derived from retrovirus-transduced human hematopoietic cells.
Proc Natl Acad Sci U S A
1999
, vol. 
96
 
17
(pg. 
9654
-
9659
)
105
Greene
 
TK
Wang
 
C
Hirsch
 
JD
, et al. 
In vivo efficacy of platelet-delivered, high specific activity factor VIII variants.
Blood
2010
, vol. 
116
 
26
(pg. 
6114
-
6122
)
106
Ohmori
 
T
Mimuro
 
J
Takano
 
K
, et al. 
Efficient expression of a transgene in platelets using simian immunodeficiency virus-based vector harboring glycoprotein Ibalpha promoter: in vivo model for platelet-targeting gene therapy.
FASEB J
2006
, vol. 
20
 
9
(pg. 
1522
-
1524
)
107
Moffat
 
J
Grueneberg
 
DA
Yang
 
X
, et al. 
A lentiviral RNAi library for human and mouse genes applied to an arrayed viral high-content screen.
Cell
2006
, vol. 
124
 
6
(pg. 
1283
-
1298
)
108
Shi
 
Q
Wilcox
 
DA
Fahs
 
SA
, et al. 
Lentivirus-mediated platelet-derived factor VIII gene therapy in murine haemophilia A.
J Thromb Haemost
2007
, vol. 
5
 
2
(pg. 
352
-
361
)
109
Shi
 
Q
Wilcox
 
DA
Fahs
 
SA
, et al. 
Factor VIII ectopically targeted to platelets is therapeutic in hemophilia A with high-titer inhibitory antibodies.
J Clin Invest
2006
, vol. 
116
 
7
(pg. 
1974
-
1982
)
110
Shi
 
Q
Fahs
 
SA
Wilcox
 
DA
, et al. 
Syngeneic transplantation of hematopoietic stem cells that are genetically modified to express factor VIII in platelets restores hemostasis to hemophilia A mice with preexisting FVIII immunity.
Blood
2008
, vol. 
112
 
7
(pg. 
2713
-
2721
)
111
Wilcox
 
DA
Olsen
 
JC
Ishizawa
 
L
, et al. 
Megakaryocyte-targeted synthesis of the integrin beta(3)-subunit results in the phenotypic correction of Glanzmann thrombasthenia.
Blood
2000
, vol. 
95
 
12
(pg. 
3645
-
3651
)
112
Fang
 
J
Jensen
 
ES
Boudreaux
 
MK
, et al. 
Platelet gene therapy improves hemostatic function for integrin alphaIIbbeta3-deficient dogs.
Proc Natl Acad Sci U S A
2011
, vol. 
108
 
23
(pg. 
9583
-
9588
)
113
Peeters
 
K
Loyen
 
S
Van
 
KS
, et al. 
Thrombopoietic effect of VPAC1 inhibition during megakaryopoiesis.
Br J Haematol
2010
, vol. 
151
 
1
(pg. 
54
-
61
)
114
Zhang
 
G
Shi
 
Q
Fahs
 
SA
, et al. 
Factor IX ectopically expressed in platelets can be stored in alpha-granules and corrects the phenotype of hemophilia B mice.
Blood
2010
, vol. 
116
 
8
(pg. 
1235
-
1243
)
115
Urnov
 
FD
Rebar
 
EJ
Holmes
 
MC
Zhang
 
HS
Gregory
 
PD
Genome editing with engineered zinc finger nucleases.
Nat Rev Genet
2010
, vol. 
11
 
9
(pg. 
636
-
646
)
116
Hockemeyer
 
D
Wang
 
H
Kiani
 
S
, et al. 
Genetic engineering of human pluripotent cells using TALE nucleases.
Nat Biotechnol
2011
, vol. 
29
 
8
(pg. 
731
-
734
)
117
Miller
 
JC
Tan
 
S
Qiao
 
G
, et al. 
A TALE nuclease architecture for efficient genome editing.
Nat Biotechnol
2011
, vol. 
29
 
2
(pg. 
143
-
148
)
118
Sander
 
JD
Cade
 
L
Khayter
 
C
, et al. 
Targeted gene disruption in somatic zebrafish cells using engineered TALENs.
Nat Biotechnol
2011
, vol. 
29
 
8
(pg. 
697
-
698
)
119
Tesson
 
L
Usal
 
C
Menoret
 
S
, et al. 
Knockout rats generated by embryo microinjection of TALENs.
Nat Biotechnol
2011
, vol. 
29
 
8
(pg. 
695
-
696
)
120
Amendola
 
M
Passerini
 
L
Pucci
 
F
, et al. 
Regulated and multiple miRNA and siRNA delivery into primary cells by a lentiviral platform.
Mol Ther
2009
, vol. 
17
 
6
(pg. 
1039
-
1052
)
121
Figueiredo
 
C
Goudeva
 
L
Horn
 
PA
, et al. 
Generation of HLA-deficient platelets from hematopoietic progenitor cells.
Transfusion
2010
, vol. 
50
 
8
(pg. 
1690
-
1701
)
122
Hong
 
W
Kondkar
 
AA
Nagalla
 
S
, et al. 
Transfection of human platelets with short interfering RNA.
Clin Transl Sci
2011
, vol. 
4
 
3
(pg. 
180
-
182
)
123
Stegmeier
 
F
Hu
 
G
Rickles
 
RJ
Hannon
 
GJ
Elledge
 
SJ
A lentiviral microRNA-based system for single-copy polymerase II-regulated RNA interference in mammalian cells.
Proc Natl Acad Sci U S A
2005
, vol. 
102
 
37
(pg. 
13212
-
13217
)
124
Scherr
 
M
Eder
 
M
Gene silencing by small regulatory RNAs in mammalian cells.
Cell Cycle
2007
, vol. 
6
 
4
(pg. 
444
-
449
)
125
Mittal
 
V
Improving the efficiency of RNA interference in mammals.
Nat Rev Genet
2004
, vol. 
5
 
5
(pg. 
355
-
365
)
126
Pei
 
Y
Tuschl
 
T
On the art of identifying effective and specific siRNAs.
Nat Methods
2006
, vol. 
3
 
9
(pg. 
670
-
676
)
127
Bauer
 
M
Kinkl
 
N
Meixner
 
A
, et al. 
Prevention of interferon-stimulated gene expression using microRNA-designed hairpins.
Gene Ther
2009
, vol. 
16
 
1
(pg. 
142
-
147
)
128
Beer
 
S
Bellovin
 
DI
Lee
 
JS
, et al. 
Low-level shRNA cytotoxicity can contribute to MYC-induced hepatocellular carcinoma in adult mice.
Mol Ther
2010
, vol. 
18
 
1
(pg. 
161
-
170
)
129
Boudreau
 
RL
Martins
 
I
Davidson
 
BL
Artificial microRNAs as siRNA shuttles: improved safety as compared to shRNAs in vitro and in vivo.
Mol Ther
2009
, vol. 
17
 
1
(pg. 
169
-
175
)
130
Grimm
 
D
Streetz
 
KL
Jopling
 
CL
, et al. 
Fatality in mice due to oversaturation of cellular microRNA/short hairpin RNA pathways.
Nature
2006
, vol. 
441
 
7092
(pg. 
537
-
541
)
131
Nielsen
 
TT
Marion
 
I
Hasholt
 
L
Lundberg
 
C
Neuron-specific RNA interference using lentiviral vectors.
J Gene Med
2009
, vol. 
11
 
7
(pg. 
559
-
569
)
132
Yang
 
W
Paschen
 
W
Conditional gene silencing in mammalian cells mediated by a stress-inducible promoter.
Biochem Biophys Res Commun
2008
, vol. 
365
 
3
(pg. 
521
-
527
)
133
Jang
 
JH
Schaffer
 
DV
Shea
 
LD
Engineering biomaterial systems to enhance viral vector gene delivery.
Mol Ther
2011
, vol. 
19
 
8
(pg. 
1407
-
1415
)
134
Mátrai
 
J
Chuah
 
MK
VandenDriessche
 
T
Recent advances in lentiviral vector development and applications.
Mol Ther
2010
, vol. 
18
 
3
(pg. 
477
-
490
)
135
Mátés
 
L
Chuah
 
MK
Belay
 
E
, et al. 
Molecular evolution of a novel hyperactive Sleeping Beauty transposase enables robust stable gene transfer in vertebrates.
Nat Genet
2009
, vol. 
41
 
6
(pg. 
753
-
761
)
136
Sumiyoshi
 
T
Holt
 
NG
Hollis
 
RP
, et al. 
Stable transgene expression in primitive human CD34+ hematopoietic stem/progenitor cells, using the Sleeping Beauty transposon system.
Hum Gene Ther
2009
, vol. 
20
 
12
(pg. 
1607
-
1626
)
137
Xue
 
X
Huang
 
X
Nodland
 
SE
, et al. 
Stable gene transfer and expression in cord blood-derived CD34+ hematopoietic stem and progenitor cells by a hyperactive Sleeping Beauty transposon system.
Blood
2009
, vol. 
114
 
7
(pg. 
1319
-
1330
)
138
Shultz
 
LD
Ishikawa
 
F
Greiner
 
DL
Humanized mice in translational biomedical research.
Nat Rev Immunol
2007
, vol. 
7
 
2
(pg. 
118
-
130
)
139
Salles
 
II
Thijs
 
T
Brunaud
 
C
, et al. 
Human platelets produced in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice upon transplantation of human cord blood CD34(+) cells are functionally active in an ex vivo flow model of thrombosis.
Blood
2009
, vol. 
114
 
24
(pg. 
5044
-
5051
)
140
Tijssen
 
MR
van Hennik
 
PB
di Summa
 
F
, et al. 
Transplantation of human peripheral blood CD34-positive cells in combination with ex vivo generated megakaryocytes results in fast platelet formation in NOD/SCID mice.
Leukemia
2008
, vol. 
22
 
1
(pg. 
203
-
208
)
141
McDermott
 
SP
Eppert
 
K
Lechman
 
ER
Doedens
 
M
Dick
 
JE
Comparison of human cord blood engraftment between immunocompromised mouse strains.
Blood
2010
, vol. 
116
 
2
(pg. 
193
-
200
)
Sign in via your Institution