Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 179
Filter
1.
Blood ; 143(6): 548-560, 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-37944157

ABSTRACT

ABSTRACT: Nonmuscle cell contractility is an essential feature underlying diverse cellular processes such as motility, morphogenesis, division and genome replication, intracellular transport, and secretion. Blood clot contraction is a well-studied process driven by contracting platelets. Megakaryocytes (MKs), which are the precursors to platelets, can be found in bone marrow and lungs. Although they express many of the same proteins and structures found in platelets, little is known about their ability to engage with extracellular proteins such as fibrin and contract. Here, we have measured the ability of MKs to compress plasma clots. Megakaryocytes derived from human induced pluripotent stem cells (iPSCs) were suspended in human platelet-free blood plasma and stimulated with thrombin. Using real-time macroscale optical tracking, confocal microscopy, and biomechanical measurements, we found that activated iPSC-derived MKs (iMKs) caused macroscopic volumetric clot shrinkage, as well as densification and stiffening of the fibrin network via fibrin-attached plasma membrane protrusions undergoing extension-retraction cycles that cause shortening and bending of fibrin fibers. Contraction induced by iMKs involved 2 kinetic phases with distinct rates and durations. It was suppressed by inhibitors of nonmuscle myosin IIA, actin polymerization, and integrin αIIbß3-fibrin interactions, indicating that the molecular mechanisms of iMK contractility were similar or identical to those in activated platelets. Our findings provide new insights into MK biomechanics and suggest that iMKs can be used as a model system to study platelet contractility. Physiologically, the ability of MKs to contract plasma clots may play a role in the mechanical remodeling of intravascular blood clots and thrombi.


Subject(s)
Induced Pluripotent Stem Cells , Thrombosis , Humans , Megakaryocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , Blood Platelets/metabolism , Thrombosis/metabolism , Fibrin/metabolism , Plasma
2.
Mol Genet Metab ; 141(2): 108116, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38161139

ABSTRACT

Multiple sulfatase deficiency (MSD) is an ultra-rare, inherited lysosomal storage disease caused by mutations in the gene sulfatase modifying factor 1 (SUMF1). MSD is characterized by the functional deficiency of all sulfatase enzymes, leading to the storage of sulfated substrates including glycosaminoglycans (GAGs), sulfolipids, and steroid sulfates. Patients with MSD experience severe neurological impairment, hearing loss, organomegaly, corneal clouding, cardiac valve disease, dysostosis multiplex, contractures, and ichthyosis. Here, we generated a novel human model of MSD by reprogramming patient peripheral blood mononuclear cells to establish an MSD induced pluripotent stem cell (iPSC) line (SUMF1 p.A279V). We also generated an isogenic control iPSC line by correcting the pathogenic variant with CRISPR/Cas9 gene editing. We successfully differentiated these iPSC lines into neural progenitor cells (NPCs) and NGN2-induced neurons (NGN2-iN) to model the neuropathology of MSD. Mature neuronal cells exhibited decreased SUMF1 gene expression, increased lysosomal stress, impaired neurite outgrowth and maturation, reduced sulfatase activities, and GAG accumulation. Interestingly, MSD iPSCs and NPCs did not exhibit as severe of phenotypes, suggesting that as neurons differentiate and mature, they become more vulnerable to loss of SUMF1. In summary, we demonstrate that this human iPSC-derived neuronal model recapitulates the cellular and biochemical features of MSD. These cell models can be used as tools to further elucidate the mechanisms of MSD pathology and for the development of therapeutics.


Subject(s)
Induced Pluripotent Stem Cells , Multiple Sulfatase Deficiency Disease , Humans , Leukocytes, Mononuclear/metabolism , Neurons/pathology , Sulfatases , Oxidoreductases Acting on Sulfur Group Donors
3.
Blood ; 140(15): 1723-1734, 2022 10 13.
Article in English | MEDLINE | ID: mdl-35977098

ABSTRACT

Red blood cell (RBC) transfusion is one of the most common medical treatments, with more than 10 million units transfused per year in the United States alone. Alloimmunization to foreign Rh proteins (RhD and RhCE) on donor RBCs remains a challenge for transfusion effectiveness and safety. Alloantibody production disproportionately affects patients with sickle cell disease who frequently receive blood transfusions and exhibit high genetic diversity in the Rh blood group system. With hundreds of RH variants now known, precise identification of Rh antibody targets is hampered by the lack of appropriate reagent RBCs with uncommon Rh antigen phenotypes. Using a combination of human-induced pluripotent stem cell (iPSC) reprogramming and gene editing, we designed a renewable source of cells with unique Rh profiles to facilitate the identification of complex Rh antibodies. We engineered a very rare Rh null iPSC line lacking both RHD and RHCE. By targeting the AAVS1 safe harbor locus in this Rh null background, any combination of RHD or RHCE complementary DNAs could be reintroduced to generate RBCs that express specific Rh antigens such as RhD alone (designated D--), Goa+, or DAK+. The RBCs derived from these iPSCs (iRBCs) are compatible with standard laboratory assays used worldwide and can determine the precise specificity of Rh antibodies in patient plasma. Rh-engineered iRBCs can provide a readily accessible diagnostic tool and guide future efforts to produce an alternative source of rare RBCs for alloimmunized patients.


Subject(s)
Blood Group Antigens , Pluripotent Stem Cells , Transfusion Medicine , Alleles , Blood Group Antigens/genetics , Humans , Rh-Hr Blood-Group System/genetics
4.
Haematologica ; 109(8): 2533-2541, 2024 08 01.
Article in English | MEDLINE | ID: mdl-38152053

ABSTRACT

Mutations in five canonical Ras pathway genes (NF1, NRAS, KRAS, PTPN11 and CBL) are detected in nearly 90% of patients with juvenile myelomonocytic leukemia (JMML), a frequently fatal malignant neoplasm of early childhood. In this report, we describe seven patients diagnosed with SH2B3-mutated JMML, including five patients who were found to have initiating, loss-of-function mutations in the gene. SH2B3 encodes the adaptor protein LNK, a negative regulator of normal hematopoiesis upstream of the Ras pathway. These mutations were identified to be germline, somatic or a combination of both. Loss of function of LNK, which has been observed in other myeloid malignancies, results in abnormal proliferation of hematopoietic cells due to cytokine hypersensitivity and activation of the JAK/STAT signaling pathway. In vitro studies of induced pluripotent stem cell-derived JMML-like hematopoietic progenitor cells also demonstrated sensitivity of SH2B3-mutated hematopoietic progenitor cells to JAK inhibition. Lastly, we describe two patients with JMML and SH2B3 mutations who were treated with the JAK1/2 inhibitor ruxolitinib. This report expands the spectrum of initiating mutations in JMML and raises the possibility of targeting the JAK/STAT pathway in patients with SH2B3 mutations.


Subject(s)
Adaptor Proteins, Signal Transducing , Leukemia, Myelomonocytic, Juvenile , Mutation , Humans , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/metabolism , Leukemia, Myelomonocytic, Juvenile/pathology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Male , Female , Infant , Child, Preschool , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Child , Signal Transduction , Pyrazoles/therapeutic use , Pyrazoles/pharmacology , Nitriles , Pyrimidines
5.
J Sex Med ; 21(10): 872-877, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39165032

ABSTRACT

BACKGROUND: To date, there is a lack of a structured national plan of sexual education in Italy, with large interregional differences and poor uniformity in the promotion and learning of sexual health. AIM: The aim of this study was to evaluate the level of knowledge on sexuality and reproductive function among adolescents before and after a sexual education program, considering gender differences in the results. METHODS: A longitudinal and interventional study was conducted on high school students. Six sessions for each class were carried out by a multidisciplinary team. Anonymous questionnaires were administered to investigate basic knowledge of human sexuality and reproduction before and after intervention. OUTCOMES: Participants were high school students who completed the questionnaires. Outcomes included pre and posttest answers on the knowledge questionnaire in study population. RESULTS: We obtained data from 842 high school students (mean ± SD age, 16.11 ± 1.74 years). The main informative sources on sexuality were social media in 37.9% of participants; family, school, and scientific material were the main source in 15.1%, 5.2%, and 4.4%, respectively. A total score of 12.6 ± 3.4 (range, 1-20) was obtained on the knowledge questionnaire; specifically, the score was higher in females than males (12.8 ± 3.4 vs 11.9 ± 3.4, P = .0001). In males and females, a statistically significant increase in level of knowledge was found after the multidisciplinary intervention (14.5 ± 3.6, P = .0001). CLINICAL IMPLICATIONS: The lack of sexual education programs in schools in Italy compels adolescents to refer to frequently unreliable sources, such as social media and peers, contributing to misinformation and adoption of risky sexual behaviors. STRENGTHS AND LIMITATIONS: This study is based on a large sample size and highlights the importance of sexual education programs. The main limitations are a greater representation of the female population and the inclusive nature of the school (ie, an artistic school), attracting students who identify with sexual minorities. CONCLUSIONS: Adolescents have limited knowledge about sexuality and reproduction, with a notable gender disparity; sexual education programs in schools, providing information from sexuality experts, led to a substantial increase in knowledge and a reduction in the gender gap.


Subject(s)
Health Knowledge, Attitudes, Practice , Reproductive Health , Sex Education , Sexual Health , Humans , Adolescent , Female , Male , Sex Education/methods , Reproductive Health/education , Italy , Surveys and Questionnaires , Longitudinal Studies , Students/statistics & numerical data , Students/psychology , Sexual Behavior/statistics & numerical data , Information Sources
6.
Clin Genet ; 103(1): 109-113, 2023 01.
Article in English | MEDLINE | ID: mdl-36075864

ABSTRACT

Prior studies have demonstrated that patients with chromosome 22q11.2 deletion syndrome (22q11.2DS) have lower platelet counts (PC) compared to non-deleted populations. They also have an increased mean platelet volume. The mechanism for this has been postulated to be haploinsufficiency of the GPIBB gene. We examined platelet parameters, deletion size and factors known to influence counts, including status of thyroid hormone and congenital heart disease (CHD), in a population of 825 patients with 22q11.2DS. We also measured surface expression of GPIB-IX complex by flow cytometry. The major determinant of PC was deletion status of GP1BB, regardless of surface expression or other factors. Patients with nested distal chromosome 22q11.2 deletions (those with GP1BB present) had higher PCs than those with proximal deletions where GP1BB is deleted. Patients with 22q11.2DS also demonstrated an accelerated PC decrease with age, occurring in childhood. These data demonstrate that genes within the proximal deletion segment drive PC differences in 22q11.2DS and suggest that PC reference ranges may need to be adjusted for age and deletion size in 22q11.2DS populations. Bleeding did not correlate with either platelet count or GPIb expression. Further studies into drivers of expression of GPIb and associations with severe thrombocytopenia and immune thrombocytopenia are needed to inform clinical care.


Subject(s)
DiGeorge Syndrome , Humans , DiGeorge Syndrome/genetics
7.
Blood ; 137(19): 2662-2675, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33569577

ABSTRACT

Patients with familial platelet disorder with a predisposition to myeloid malignancy (FPDMM) harbor germline monoallelic mutations in a key hematopoietic transcription factor, RUNX-1. Previous studies of FPDMM have focused on megakaryocyte (Mk) differentiation and platelet production and signaling. However, the effects of RUNX-1 haploinsufficiency on hematopoietic progenitor cells (HPCs) and subsequent megakaryopoiesis remains incomplete. We studied induced pluripotent stem cell (iPSC)-derived HPCs (iHPCs) and Mks (iMks) from both patient-derived lines and a wild-type (WT) line modified to be RUNX-1 haploinsufficient (RUNX-1+/-), each compared with their isogenic WT control. All RUNX-1+/- lines showed decreased iMk yield and depletion of an Mk-biased iHPC subpopulation. To investigate global and local gene expression changes underlying this iHPC shift, single-cell RNA sequencing was performed on sorted FPDMM and control iHPCs. We defined several cell subpopulations in the Mk-biased iHPCs. Analyses of gene sets upregulated in FPDMM iHPCs indicated enrichment for response to stress, regulation of signal transduction, and immune signaling-related gene sets. Immunoblot analyses in FPDMM iMks were consistent with these findings, but also identified augmented baseline c-Jun N-terminal kinase (JNK) phosphorylation, known to be activated by transforming growth factor-ß1 (TGF-ß1) and cellular stressors. These findings were confirmed in adult human CD34+-derived stem and progenitor cells (HSPCs) transduced with lentiviral RUNX1 short hairpin RNA to mimic RUNX-1+/-. In both iHPCs and CD34+-derived HSPCs, targeted inhibitors of JNK and TGF-ß1 pathways corrected the megakaryopoietic defect. We propose that such intervention may correct the thrombocytopenia in patients with FPDMM.


Subject(s)
Core Binding Factor Alpha 2 Subunit/deficiency , Hematopoietic Stem Cells/pathology , Megakaryocytes/pathology , Neoplastic Syndromes, Hereditary/pathology , Adult , Base Sequence , Core Binding Factor Alpha 2 Subunit/genetics , Flow Cytometry , Haploinsufficiency , Humans , Immunophenotyping , Induced Pluripotent Stem Cells/cytology , MAP Kinase Signaling System , Neoplastic Syndromes, Hereditary/genetics , Platelet Glycoprotein GPIb-IX Complex/analysis , RNA, Small Interfering/genetics , Recombinant Proteins/metabolism , Signal Transduction , Single-Cell Analysis , Thrombopoiesis , Transforming Growth Factor beta1/physiology
8.
Curr Opin Hematol ; 28(5): 308-314, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34397590

ABSTRACT

PURPOSE OF REVIEW: Megakaryocytes are rare hematopoietic cells that play an instrumental role in hemostasis, and other important biological processes such as immunity and wound healing. With the advent of cell reprogramming technologies and advances in differentiation protocols, it is now possible to obtain megakaryocytes from any pluripotent stem cell (PSC) via hematopoietic induction. Here, we review recent advances in PSC-derived megakaryocyte (iMK) technology, focusing on platform validation, disease modeling and current limitations. RECENT FINDINGS: A comprehensive study confirmed that iMK can recapitulate many transcriptional and functional aspects of megakaryocyte and platelet biology, including variables associated with complex genetic traits such as sex and race. These findings were corroborated by several pathological models in which iMKs revealed molecular mechanisms behind inherited platelet disorders and assessed the efficacy of novel pharmacological interventions. However, current differentiation protocols generate primarily embryonic iMK, limiting the clinical and translational potential of this system. SUMMARY: iMK are strong candidates to model pathologic mutations involved in platelet defects and develop innovative therapeutic strategies. Future efforts on generating definitive hematopoietic progenitors would improve current platelet generation protocols and expand our capacity to model neonatal and adult megakaryocyte disorders.


Subject(s)
Blood Platelet Disorders , Cell Differentiation , Genetic Diseases, Inborn , Hematopoiesis , Models, Genetic , Pluripotent Stem Cells/metabolism , Animals , Blood Platelet Disorders/genetics , Blood Platelet Disorders/metabolism , Blood Platelet Disorders/therapy , Blood Platelets/metabolism , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/therapy , Humans , Megakaryocytes/metabolism
9.
BMC Biol ; 18(1): 52, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32408895

ABSTRACT

BACKGROUND: Identifying causal variants and genes from human genetic studies of hematopoietic traits is important to enumerate basic regulatory mechanisms underlying these traits, and could ultimately augment translational efforts to generate platelets and/or red blood cells in vitro. To identify putative causal genes from these data, we performed computational modeling using available genome-wide association datasets for platelet and red blood cell traits. RESULTS: Our model identified a joint collection of genomic features enriched at established trait associations and plausible candidate variants. Additional studies associating variation at these loci with change in gene expression highlighted Tropomyosin 1 (TPM1) among our top-ranked candidate genes. CRISPR/Cas9-mediated TPM1 knockout in human induced pluripotent stem cells (iPSCs) enhanced hematopoietic progenitor development, increasing total megakaryocyte and erythroid cell yields. CONCLUSIONS: Our findings may help explain human genetic associations and identify a novel genetic strategy to enhance in vitro hematopoiesis. A similar trait-specific gene prioritization strategy could be employed to help streamline functional validation experiments for virtually any human trait.


Subject(s)
Blood Platelets/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Tropomyosin/metabolism , CRISPR-Cas Systems , Genome-Wide Association Study , Humans , In Vitro Techniques , Tropomyosin/deficiency
10.
Int J Mol Sci ; 22(12)2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34207106

ABSTRACT

Infection with human papillomavirus type 16 (HPV16) is one of the major risk factors for the development of cervical cancer. Our previous studies have demonstrated the involvement of the early oncoprotein E5 of HPV16 (16E5) in the altered isoform switch of fibroblast growth factor receptor 2 (FGFR2) and the consequent expression in human keratinocytes of the mesenchymal FGFR2c isoform, whose aberrant signaling leads to EMT, invasiveness, and dysregulated differentiation. Here, we aimed to establish the possible direct link between these pathological features or the appearance of FGFR2c and the expression of 16E5 in low-grade squamous intraepithelial lesions (LSILs). Molecular analysis showed that the FGFR2c expression displayed a statistically significant positive correlation with that of the viral oncoprotein, whereas the expression values of the epithelial FGR2b variant, as well as those of the differentiation markers keratin 10 (K10), loricrin (LOR) and involucrin (INV), were inversely linked to the 16E5 expression. In contrast, the expression of EMT-related transcription factors Snail1 and ZEB1 overlapped with that of 16E5, becoming a statistically significant positive correlation in the case of Snail2. Parallel analysis performed in human cervical LSIL-derived W12 cells, containing episomal HPV16, revealed that the depletion of 16E5 by siRNA was able to counteract these molecular events, proving to represent an effective strategy to identify the specific role of this viral oncoprotein in determining LSIL oncogenic and more aggressive profiles. Overall, coupling in vitro approaches to the molecular transcript analysis in ectocervical early lesions could significantly contribute to the characterization of specific gene expression profiles prognostic for those LSILs with a greater probability of direct neoplastic progression.


Subject(s)
Epithelial-Mesenchymal Transition , Gene Expression , Human papillomavirus 16/genetics , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/complications , Uterine Cervical Neoplasms/etiology , Uterine Cervical Neoplasms/pathology , Cell Line , Cell Transformation, Viral , Cervix Uteri/pathology , Cervix Uteri/virology , Disease Susceptibility , Epithelial Cells/metabolism , Female , Humans , Keratinocytes/metabolism , Neoplasm Grading , Papillomavirus Infections/virology , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Uterine Cervical Neoplasms/metabolism
11.
Blood ; 129(26): 3486-3494, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28432223

ABSTRACT

Friend leukemia virus integration 1 (FLI1), a critical transcription factor (TF) during megakaryocyte differentiation, is among genes hemizygously deleted in Jacobsen syndrome, resulting in a macrothrombocytopenia termed Paris-Trousseau syndrome (PTSx). Recently, heterozygote human FLI1 mutations have been ascribed to cause thrombocytopenia. We studied induced-pluripotent stem cell (iPSC)-derived megakaryocytes (iMegs) to better understand these clinical disorders, beginning with iPSCs generated from a patient with PTSx and iPSCs from a control line with a targeted heterozygous FLI1 knockout (FLI1+/-). PTSx and FLI1+/- iMegs replicate many of the described megakaryocyte/platelet features, including a decrease in iMeg yield and fewer platelets released per iMeg. Platelets released in vivo from infusion of these iMegs had poor half-lives and functionality. We noted that the closely linked E26 transformation-specific proto-oncogene 1 (ETS1) is overexpressed in these FLI1-deficient iMegs, suggesting FLI1 negatively regulates ETS1 in megakaryopoiesis. Finally, we examined whether FLI1 overexpression would affect megakaryopoiesis and thrombopoiesis. We found increased yield of noninjured, in vitro iMeg yield and increased in vivo yield, half-life, and functionality of released platelets. These studies confirm FLI1 heterozygosity results in pleiotropic defects similar to those noted with other critical megakaryocyte-specific TFs; however, unlike those TFs, FLI1 overexpression improved yield and functionality.


Subject(s)
Jacobsen Distal 11q Deletion Syndrome/pathology , Megakaryocytes/cytology , Proto-Oncogene Protein c-fli-1/blood , Thrombopoiesis , Animals , Blood Platelets/metabolism , Cell Differentiation , Cell Line , Humans , Induced Pluripotent Stem Cells , Mice , Mice, SCID , Proto-Oncogene Mas
12.
Blood ; 130(2): 192-204, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28455282

ABSTRACT

Stem cell-derived platelets have the potential to replace donor platelets for transfusion. Defining the platelet-producing megakaryocytes (MKs) within the heterogeneous MK culture may help to optimize the in vitro generation of platelets. Using 2 human stem cell models of megakaryopoiesis, we identified novel MK populations corresponding to distinct maturation stages. An immature, low granular (LG) MK pool (defined by side scatter on flow cytometry) gives rise to a mature high granular (HG) pool, which then becomes damaged by apoptosis and glycoprotein Ib α chain (CD42b) shedding. We define an undamaged HG/CD42b+ MK subpopulation, which endocytoses fluorescently labeled coagulation factor V (FV) from the media into α-granules and releases functional FV+CD42b+ human platelet-like particles in vitro and when infused into immunodeficient mice. Importantly, these FV+ particles have the same size distribution as infused human donor platelets and are preferentially incorporated into clots after laser injury. Using drugs to protect HG MKs from apoptosis and CD42b shedding, we also demonstrate that apoptosis precedes CD42b shedding and that apoptosis inhibition enriches the FV+ HG/CD42b+ MKs, leading to increased platelet yield in vivo, but not in vitro. These studies identify a transition between distinct MK populations in vitro, including one that is primed for platelet release. Technologies to optimize and select these platelet-ready MKs may be important to efficiently generate functional platelets from in vitro-grown MKs.


Subject(s)
Blood Platelets/cytology , Bone Marrow Cells/immunology , Factor V/genetics , Megakaryocyte Progenitor Cells/cytology , Megakaryocytes/cytology , Animals , Apoptosis/drug effects , Arterioles/drug effects , Arterioles/immunology , Arterioles/injuries , Biomarkers/blood , Blood Platelets/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Differentiation , Cell Lineage/immunology , Endocytosis , Factor V/immunology , Factor V/pharmacology , Flow Cytometry , Gene Expression , Humans , Immunophenotyping , Lasers , Megakaryocyte Progenitor Cells/immunology , Megakaryocytes/immunology , Mice , Mice, SCID , Platelet Glycoprotein GPIb-IX Complex/genetics , Platelet Glycoprotein GPIb-IX Complex/immunology
13.
Blood ; 127(10): 1227-33, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26787738

ABSTRACT

Platelets are anucleate cytoplasmic discs derived from megakaryocytes that circulate in the blood and have major roles in hemostasis, thrombosis, inflammation, and vascular biology. Platelet transfusions are required to prevent the potentially life-threatening complications of severe thrombocytopenia seen in a variety of medical settings including cancer therapy, trauma, and sepsis. Platelets used in the clinic are currently donor-derived which is associated with concerns over sufficient availability, quality, and complications due to immunologic and/or infectious issues. To overcome our dependence on donor-derived platelets for transfusion, efforts have been made to generate in vitro-based platelets. Work in this area has advanced our understanding of the complex processes that megakaryocytes must undergo to generate platelets both in vivo and in vitro. This knowledge has also defined the challenges that must be overcome to bring in vitro-based platelet manufacturing to a clinical reality. This review will focus on our understanding of committed megakaryocytes and platelet release in vivo and in vitro, and how this knowledge can guide the development of in vitro-derived platelets for clinical application.


Subject(s)
Blood Platelets/metabolism , Cell Culture Techniques/methods , Cell Differentiation , Megakaryocytes/metabolism , Platelet Transfusion , Animals , Blood Donors , Blood Platelets/cytology , Humans , Megakaryocytes/cytology
14.
Blood ; 127(6): 675-80, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26634302

ABSTRACT

Human platelet alloantigens (HPAs) reside on functionally important platelet membrane glycoproteins and are caused by single nucleotide polymorphisms in the genes that encode them. Antibodies that form against HPAs are responsible for several clinically important alloimmune bleeding disorders, including fetal and neonatal alloimmune thrombocytopenia and posttransfusion purpura. The HPA-1a/HPA-1b alloantigen system, also known as the Pl(A1)/Pl(A2) polymorphism, is the most frequently implicated HPA among whites, and a single Leu33Pro amino acid polymorphism within the integrin ß3 subunit is responsible for generating the HPA-1a/HPA-1b alloantigenic epitopes. HPA-1b/b platelets, like those bearing other low-frequency platelet-specific alloantigens, are relatively rare in the population and difficult to obtain for purposes of transfusion therapy and diagnostic testing. We used CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR associated protein 9) gene-editing technology to transform Leu33 (+) megakaryocytelike DAMI cells and induced pluripotent stem cells (iPSCs) to the Pro33 allotype. CD41(+) megakaryocyte progenitors derived from these cells expressed the HPA-1b (Pl(A2)) alloantigenic epitope, as reported by diagnostic NciI restriction enzyme digestion, DNA sequencing, and western blot analysis using HPA-1b-specific human maternal alloantisera. Application of CRISPR/Cas9 technology to genetically edit this and other clinically-important HPAs holds great potential for production of designer platelets for diagnostic, investigative, and, ultimately, therapeutic use.


Subject(s)
Antigens, Human Platelet/genetics , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/immunology , Clustered Regularly Interspaced Short Palindromic Repeats , Isoantigens/genetics , Antigens, Human Platelet/immunology , Base Sequence , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/immunology , Epitopes/genetics , Epitopes/immunology , Humans , Integrin beta3/genetics , Integrin beta3/immunology , Isoantibodies/genetics , Isoantibodies/immunology , Isoantigens/immunology , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Polymorphism, Single Nucleotide
15.
Blood ; 127(23): 2791-803, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27084890

ABSTRACT

Inherited bleeding, thrombotic, and platelet disorders (BPDs) are diseases that affect ∼300 individuals per million births. With the exception of hemophilia and von Willebrand disease patients, a molecular analysis for patients with a BPD is often unavailable. Many specialized tests are usually required to reach a putative diagnosis and they are typically performed in a step-wise manner to control costs. This approach causes delays and a conclusive molecular diagnosis is often never reached, which can compromise treatment and impede rapid identification of affected relatives. To address this unmet diagnostic need, we designed a high-throughput sequencing platform targeting 63 genes relevant for BPDs. The platform can call single nucleotide variants, short insertions/deletions, and large copy number variants (though not inversions) which are subjected to automated filtering for diagnostic prioritization, resulting in an average of 5.34 candidate variants per individual. We sequenced 159 and 137 samples, respectively, from cases with and without previously known causal variants. Among the latter group, 61 cases had clinical and laboratory phenotypes indicative of a particular molecular etiology, whereas the remainder had an a priori highly uncertain etiology. All previously detected variants were recapitulated and, when the etiology was suspected but unknown or uncertain, a molecular diagnosis was reached in 56 of 61 and only 8 of 76 cases, respectively. The latter category highlights the need for further research into novel causes of BPDs. The ThromboGenomics platform thus provides an affordable DNA-based test to diagnose patients suspected of having a known inherited BPD.


Subject(s)
Blood Platelet Disorders/genetics , Genetic Predisposition to Disease , Hemorrhage/genetics , High-Throughput Nucleotide Sequencing/methods , Thrombosis/genetics , Case-Control Studies , DNA Copy Number Variations , Female , Genetic Association Studies/methods , Humans , Male , Mutation , Polymorphism, Single Nucleotide , Sequence Analysis, DNA/methods
16.
Arterioscler Thromb Vasc Biol ; 37(11): 2007-2013, 2017 11.
Article in English | MEDLINE | ID: mdl-28982668

ABSTRACT

Platelets, derived from megakaryocytes, are anucleate cytoplasmic discs that circulate in the blood stream and play major roles in hemostasis, inflammation, and vascular biology. Platelet transfusions are used in a variety of medical settings to prevent life-threatening thrombocytopenia because of cancer therapy, other causes of acquired or inherited thrombocytopenia, and trauma. Currently, platelets used for transfusion purposes are donor derived. However, there is a drive to generate nondonor sources of platelets to help supplement donor-derived platelets. Efforts have been made by many laboratories to generate in vitro platelets and optimize their production and quality. In vitro-derived platelets have the potential to be a safer, more uniform product, and genetic manipulation could allow for better treatment of patients who become refractory to donor-derived units. This review focuses on potential clinical applications of in vitro-derived megakaryocytes and platelets, current methods to generate and expand megakaryocytes from pluripotent stem cell sources, and the use of these cells for disease modeling.


Subject(s)
Blood Platelets/physiology , Induced Pluripotent Stem Cells/physiology , Megakaryocytes/physiology , Thrombopoiesis , Blood Platelets/metabolism , Cell Line , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Hematologic Diseases/blood , Hematologic Diseases/genetics , Hematologic Diseases/therapy , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Megakaryocytes/metabolism , Megakaryocytes/transplantation , Phenotype , Platelet Transfusion
17.
Blood ; 125(23): 3627-36, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25852052

ABSTRACT

Thrombopoiesis is the process by which megakaryocytes release platelets that circulate as uniform small, disc-shaped anucleate cytoplasmic fragments with critical roles in hemostasis and related biology. The exact mechanism of thrombopoiesis and the maturation pathways of platelets released into the circulation remain incompletely understood. We showed that ex vivo-generated murine megakaryocytes infused into mice release platelets within the pulmonary vasculature. Here we now show that infused human megakaryocytes also release platelets within the lungs of recipient mice. In addition, we observed a population of platelet-like particles (PLPs) in the infusate, which include platelets released during ex vivo growth conditions. By comparing these 2 platelet populations to human donor platelets, we found marked differences: platelets derived from infused megakaryocytes closely resembled infused donor platelets in morphology, size, and function. On the other hand, the PLP was a mixture of nonplatelet cellular fragments and nonuniform-sized, preactivated platelets mostly lacking surface CD42b that were rapidly cleared by macrophages. These data raise a cautionary note for the clinical use of human platelets released under standard ex vivo conditions. In contrast, human platelets released by intrapulmonary-entrapped megakaryocytes appear more physiologic in nature and nearly comparable to donor platelets for clinical application.


Subject(s)
Blood Platelets , Macrophages , Megakaryocytes , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Cell Line , Humans , Macrophages/metabolism , Macrophages/pathology , Megakaryocytes/metabolism , Megakaryocytes/pathology , Megakaryocytes/transplantation , Mice , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombopoiesis
18.
Blood ; 125(6): 930-40, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25490895

ABSTRACT

To explore how RUNX1 mutations predispose to leukemia, we generated induced pluripotent stem cells (iPSCs) from 2 pedigrees with germline RUNX1 mutations. The first, carrying a missense R174Q mutation, which acts as a dominant-negative mutant, is associated with thrombocytopenia and leukemia, and the second, carrying a monoallelic gene deletion inducing a haploinsufficiency, presents only as thrombocytopenia. Hematopoietic differentiation of these iPSC clones demonstrated profound defects in erythropoiesis and megakaryopoiesis and deregulated expression of RUNX1 targets. iPSC clones from patients with the R174Q mutation specifically generated an increased amount of granulomonocytes, a phenotype reproduced by an 80% RUNX1 knockdown in the H9 human embryonic stem cell line, and a genomic instability. This phenotype, found only with a lower dosage of RUNX1, may account for development of leukemia in patients. Altogether, RUNX1 dosage could explain the differential phenotype according to RUNX1 mutations, with a haploinsufficiency leading to thrombocytopenia alone in a majority of cases whereas a more complete gene deletion predisposes to leukemia.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Hematopoiesis , Leukemia/genetics , Mutation , Thrombocytopenia/genetics , Cell Line , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Deletion , Genetic Predisposition to Disease , Genomic Instability , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Leukemia/metabolism , Leukemia/pathology , Molecular Sequence Data , Mutation, Missense , Thrombocytopenia/metabolism , Thrombocytopenia/pathology
19.
J Surg Oncol ; 116(2): 184-194, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28475823

ABSTRACT

BACKGROUND AND OBJECTIVES: Gastric cancers are usually characterized using Lauren's classification into intestinal and diffuse types. We previously documented the down-modulation of miR31, miR148a, miR204, and miR375 in gastric cancers. We aimed this manuscript to investigate these miRs with the end-points of diagnosis, Lauren's classification and prognosis. METHODS: A total of 117 resected non-cardial adenocarcinomas were evaluated for miRs' expressions. The performance of miRs' expressions for cancer diagnosis was tested using ROC curves. Logistic regression was conducted with the end-point of Lauren's classification. Kaplan-Meier and Cox analyses were performed for OS, DFS, and DSS. miRs' targets were reviewed using PRISMA method and BCL-2 was further investigated in cell lines. RESULTS: ROC curves documented that miRs' down-modulation was significant in differentiating cancer versus normal tissues. Diffuse type cancers were associated with female sex, young age, and miR375 higher expression. We confirmed BCL-2 as a miR204 target. However, survival analyses confirmed the pathologic criteria (advanced stages, LNR, and low LNH) as the significant variables correlated to worse prognosis. CONCLUSIONS: The down-modulation of miR31, miR148a, miR204, and miR375 is significantly associated with non-cardial gastric cancers and miR375 is specifically linked to Lauren's classification. Nevertheless, standard pathological features display as the independent variables associated with worse prognosis.


Subject(s)
Adenocarcinoma/classification , Adenocarcinoma/genetics , MicroRNAs/genetics , Stomach Neoplasms/classification , Stomach Neoplasms/genetics , Adenocarcinoma/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Cell Line, Tumor , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Male , MicroRNAs/metabolism , Middle Aged , Prognosis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , ROC Curve , Repressor Proteins/genetics , Repressor Proteins/metabolism , Retrospective Studies , Sensitivity and Specificity , Sex Factors , Stomach Neoplasms/metabolism
20.
Blood ; 123(5): 753-7, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24335497

ABSTRACT

Megakaryocyte-specific transgene expression in patient-derived induced pluripotent stem cells (iPSCs) offers a new approach to study and potentially treat disorders affecting megakaryocytes and platelets. By using a Gp1ba promoter, we developed a strategy for achieving a high level of protein expression in human megakaryocytes. The feasibility of this approach was demonstrated in iPSCs derived from two patients with Glanzmann thrombasthenia (GT), an inherited platelet disorder caused by mutations in integrin αIIbß3. Hemizygous insertion of Gp1ba promoter-driven human αIIb complementary DNA into the AAVS1 locus of iPSCs led to high αIIb messenger RNA and protein expression and correction of surface αIIbß3 in megakaryocytes. Agonist stimulation of these cells displayed recovery of integrin αIIbß3 activation. Our findings demonstrate a novel approach to studying human megakaryocyte biology as well as functional correction of the GT defect, offering a potential therapeutic strategy for patients with diseases that affect platelet function.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Megakaryocytes/metabolism , Membrane Glycoproteins/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Thrombasthenia/genetics , Transgenes , Gene Expression , Humans , Platelet Glycoprotein GPIb-IX Complex , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL