Megakaryocytes of patients with MYH9-related thrombocytopenia present an altered proplatelet formation

2009 ◽  
Vol 102 (07) ◽  
pp. 90-96 ◽  
Author(s):  
Alessandro Malara ◽  
Stefania Badalucco ◽  
Valeria Bozzi ◽  
Mauro Torti ◽  
Carlo L. Balduini ◽  
...  

Summary MYH9-related disease (MYH9-RD) is an autosomal-dominant thrombocytopenia caused by mutations of MYH9, the gene for the heavy chain of myosin-IIA. Pathogenesis of thrombocytopenia of MYH9-RD is unknown. Recent studies in mice demonstrated that myosin-IIA is an inhibitor of proplatelet formation (PPF), and suggested that it could be involved in the suppression of PPF exerted by megakaryocyte adhesion to type I collagen, which regulates the timing of platelet release within bone marrow. However, the consequences on PPF of the heterozygous mutations causative of the MYH9-RD have never been investigated. We studied the in-vitro PPF by megakaryocytes obtained from four patients carrying the p.D1424N or the p.R1933X mutations. We demonstrated that MYH9-RD megakaryocytes completely lose the physiologic suppression of proplatelet extension exerted by interaction with type I collagen, thus supporting the hypothesis that a premature platelet release within bone marrow contributes to pathogenesis of MYH9-related thrombocytopenia. Moreover, proplatelets extended by MYH9-RD megakaryocytes presented a significant defect in branching in secondary processes (p=0.001) and formed a significantly lower number of proplatelet tips (p=0.005). Since platelets are assembled at the level of proplatelet tips, this defect could further contribute to pathogenesis of thrombocytopenia of MYH9-RD patients.

2011 ◽  
Vol 106 (10) ◽  
pp. 693-704 ◽  
Author(s):  
Valeria Bozzi ◽  
Emanuele Panza ◽  
Serena Barozzi ◽  
Cristian Gruppi ◽  
Marco Seri ◽  
...  

SummaryMYH9-related disease (MYH9-RD) is an autosomal-dominant thrombocytopenia caused by mutations in the gene for the heavy chain of nonmuscle myosin-IIA (NMMHC-IIA). Recent in vitro studies led to the hypothesis that thrombocytopenia of MYH9-RD derives from an ectopic platelet release by megakaryocytes in the osteoblastic areas of bone marrow (BM), which are enriched in type I collagen, rather than in vascular spaces. SDF-1-driven migration of megakaryocytes within BM to reach the vascular spaces is a key mechanism for platelet biogenesis. Since myosin-IIA is implicated in polarised migration of different cell types, we hypothesised that MYH9 mutations could interfere with this mechanism. We therefore investigated the SDF-1-driven migration of a megakaryoblastic cell line, Dami cells, on type I collagen or fibrinogen by a modified transwell assay. Inhibition of myosin-IIA ATPase activity suppressed the SDF-1-driven migration of Dami cells, while over-expression of NMMHC-IIA increased the efficiency of chemotaxis, indicat- ing a role for NMMHC-IIA in this mechanism. Transfection of cells with three MYH9 mutations frequently responsible for MYH9-RD (p.R702C, p.D1424H, or p.R1933X) resulted in a defective SDF-1-driven migration with respect to the wild-type counterpart and in increased cell spreading onto collagen. Analysis of differential localisation of wild-type and mutant proteins suggested that mutant NMMHC-IIAs had an impaired cytoplasmic re-organisation in functional cytoskeletal structures after cell adhesion to collagen. These findings support the hypothesis that a defect of SDF-1-driven migration of megakaryocytes induced by MYH9 mutations contributes to ectopic platelet release in the BM osteoblastic areas, resulting in ineffective platelet production.


2009 ◽  
Vol 131 (10) ◽  
Author(s):  
Jinjin Ma ◽  
Kristen Goble ◽  
Michael Smietana ◽  
Tatiana Kostrominova ◽  
Lisa Larkin ◽  
...  

The incidence of ligament injury has recently been estimated at 400,000/year. The preferred treatment is reconstruction using an allograft, but outcomes are limited by donor availability, biomechanical incompatibility, and immune rejection. The creation of an engineered ligament in vitro solely from patient bone marrow stromal cells (has the potential to greatly enhance outcomes in knee reconstructions. Our laboratory has developed a scaffoldless method to engineer three-dimensional (3D) ligament and bone constructs from rat bone marrow stem cells in vitro. Coculture of these two engineered constructs results in a 3D bone-ligament-bone (BLB) construct with viable entheses, which was successfully used for medial collateral ligament (MCL) replacement in a rat model. 1 month and 2 month implantations were applied to the engineered BLBs. Implantation of 3D BLBs in a MCL replacement application demonstrated that our in vitro engineered tissues grew and remodeled quickly in vivo to an advanced phenotype and partially restored function of the knee. The explanted 3D BLB ligament region stained positively for type I collagen and elastin and was well vascularized after 1 and 2 months in vivo. Tangent moduli of the ligament portion of the 3D BLB 1 month explants increased by a factor of 2.4 over in vitro controls, to a value equivalent to those observed in 14-day-old neonatal rat MCLs. The 3D BLB 1 month explants also exhibited a functionally graded response that closely matched native MCL inhomogeneity, indicating the constructs functionally adapted in vivo.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1609-1609 ◽  
Author(s):  
Isabella Pallotta ◽  
Michael L. Lovett ◽  
David L. Kaplan ◽  
Alessandra Balduini

Abstract Abstract 1609 Background. The mechanisms that regulate megakaryocytic (Mk) development within the bone marrow environment remain poorly understood. The underlying relationships between Mk maturation and bone marrow components are key factors in this process. Mk development occurs in a complex microenvironment where extracellular matrices are fundamental regulatory components. The first events occur in the osteoblastic niche and include commitment of the hemopoietic progenitor cell to Mk, arrest of proliferation and initiation of endomitosis. The second step is Mk maturation and is associated with rapid cytoplasm expansion and intense synthesis of proteins. Finally Mks, which migrate to the vascular niche, convert the bulk of their cytoplasm into multiple long processes called proplatelets that protrude through the vascular endothelium into the sinusoid lumen, where the platelets are released. Hypothesis. The hypothesis for the present work is that a complex in vitro 3D bone marrow-like environment can be used to gain fundamental mechanistic insight into cell signalling and matrix-cell interactions in the bone marrow niche related to Mk development. Methods. We propose the first 3D model for Mk function in the bone marrow environment, by refining a recently proposed bioreactor platform (Lovett et al., 2007). These bioreactors consist of 3 wells (10 mm × 15 mm × 5 mm) within a PDMS block (25 mm × 60 mm × 5 mm) which is plasma bonded to cover glass for imaging. Each bioreactor well was perfused by 23 G stainless steel needles, spanned by porous silk microtubes as blood vessel scaffolds (640 μm inner diameter), positioned approximately 500–750 μm from the bottom of the bioreactor and connected to tubing for media perfusion using a programmable syringe pump. These microtubes were prepared by dipping several times straight lengths of stainless steel wire into 10–14% (w/v) aqueous silk fibroin to obtain blood vessel scaffolds with a wall thickness of around 50 mm. Defined pore sizes of 6–8 μm were obtained by adding 6 w/t % poly(ethylene oxide) (PEO) to the silk fibroin. The perfused silk tubes comprised the vascular niche and were embedded within a cell-seeded hydrogel which comprises the osteoblastic niche. The silk microtubes were coated with a combination of fibrinogen, von Willebrand Factor, type IV collagen and SDF-1 alpha, to better establish the composition of the vascular niche. Control experiments were performed by coating silk microtubes with type I collagen. After staining human umbilical cord blood derived Mks, the cell suspension was added to the hydrogel and Mk migration was analyzed in a time-dependent manner using confocal microscopy analysis. Further, flow effluent through the vascular tubes in the bioreactor was collected at regular time intervals and platelet numbers and function were analyzed by flow cytometry and microscopy. Culture released platelets were counted as CD61+ events with the same scatter properties of human blood platelets. Results. Our results showed that Mks migrated towards the vascular microtube coated with Fibrinogen, von Willebrand Factor, type IV collagen and SDF-1. Mks were also able to complete their maturation in the proximity of the microtube by extending proplatelets. Interestingly, confocal microscopy analysis revealed that Mks were able to extend proplatelets through the vascular microtube wall and release CD61+ platelet-like particles inside the vascular microtube. Cytofluorimentric analysis demonstrated that the particles collected in the flow effluent of the vascular microtube were CD61+ cells with the same scatter properties of human peripheral blood platelets. Finally, upon coating with only type I collagen Mks did not migrate towards the vascular microtube or extend proplatelets to release platelets. Thus, by mimicking the relationship between Mks and the bone marrow environment, a model to reproduce the different steps of Mk development, such as Mk migration, proplatelet formation and platelet release, is established. This is a first significant step towards relevant systems for the study of these cellular processes in detail as well as toward potentially useful in vitro platelet production systems. Conclusions. In this work we developed a new 3D bone marrow system in vitro that could represent a new tool to understand the mechanistic basis for Mk development and function, and the diseases related to these cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2626-2626
Author(s):  
Alessandro Malara ◽  
Cristian Gruppi ◽  
Paola Rebuzzini ◽  
Maria Enrica Tira ◽  
Alessandra Balduini

Abstract Abstract 2626 Background. The mechanisms by which megakaryocytes (Mks) proliferate, differentiate, and release platelets into circulation are not well understood. Mk maturation and platelet generation occur in the bone marrow and is consequent to Mk migration from the osteoblastic to the vascular niche, where Mks extend proplatelets and newly generated platelets can be released into the bloodstream. Growing evidence indicate that a complex regulatory mechanism, involving megakaryocyte-matrix interactions, may contribute to the quiescent or permissive microenvironment related to Mk differentiation and maturation within the bone marrow. It has been demonstrated that interactions of primary human Mks with matrices supposed to fill the vascular niche, such as fibrinogen or von Willebrand factor, is able to sustain Mk maturation and proplatelet formation, while type I collagen, in the osteoblastic niche, totally suppresses these events and prevents premature platelet release. The negative regulation of proplatelet formation by type I collagen is mediated by the interaction with integrin alpha2beta1, and involves the Rho/ROCK pathway. Hypothesis. The dynamic interaction of Mks with different extra-cellular matrices, that fill the bone marrow spaces, may orchestrate their maturation in specific sites. Despite the improvement in knowledge of biochemical niche, little is known about the mechanical force that regulate Mk-niche interactions. Therefore, in this work, we correlated activation of signaling cascade with generation of contractile force to understand the influences of bone marrow environment on Mk function. Methodology/Principal Findings. To address this hypothesis, we first demonstrated that human Mks express and synthesize cellular fibronectin (cFN), with a predominance of the EDA isoform, and transglutaminase FXIII-A. Thereafter, we proposed that these two molecules are involved in a new regulatory mechanism of Mk-type I collagen interaction in the osteoblastic niche. We propose that Mk adhesion on type I collagen promotes Mk spreading through a mechanism that involves FN, membrane receptors and FXIII-A activity. This mechanism seemed to be mediated by the exposure of cFN to the cell membrane and maintained by FN polymerization catalyzed by FXIII-A. These data address a new role to FN that, upon specific activation, could be released and thereby modulate Mk interaction with extracellular matrices. In this context FXIII-A catalyzes FN cross-linking at cellular sites, stabilizes FN assembly and promotes the organization of extracellular matrix. Consistently, the same mechanism regulated the assembly of plasma FN (pFN) by adherent Mks to type I collagen. Most importantly, our results demonstrated that only Mks adherent to type I collagen, and not to fibrinogen, were able to promote FN assembly. As a result, we observed that Mk adhesion to type I collagen promoted Mk spreading overtime, while Mks on fibrinogen showed a shortened spreading that was replaced by proplatelet formation in sixteen hours of adhesion. Thus, FN assembly regulate the anchoring of Mks to type I collagen with consequent activation of biochemical signalling and generation of contractile force that may prevent proplatelet formation. Conclusions/Significance. In conclusion, this study provides important new elements in the understanding of the regulatory pathways for Mk-matrix interactions within bone marrow environment. In particular, our results demonstrate that fibronectins and FXIII-A modulate Mk spreading on type I collagen by promoting matrix assembly. This work opens new prospective in the study of illnesses, such as primary myelofibrosis or MYH9-related thrombocytopenia, related to defect of Mk-matrix interactions within the bone marrow environment, whose origin is still matter of debate. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1330-1330
Author(s):  
Vittorio Abbonante ◽  
Luca Monti ◽  
Alessandra Balduini

Abstract Abstract 1330 Background. The mechanisms that regulate the megakaryocyte (MK) development within the bone marrow environment remain poorly understood. The underlying relationships between MK maturation and bone marrow components are key factors in this process. MK development occurs in a complex microenvironment where extracellular matrices are fundamental regulatory components. Since MK maturation is critical to blood functions, it appears evident that a fine regulation of the different steps of MK development is needed. On this basis, the search for new receptors, that may regulate MK functions, is still open. Discoidin domain receptor 1 (DDR1) is a tyrosine-kinase collagen receptor that is stimulated by fibrillar and base membrane collagens and mediates cell adhesion and migration in different tissues. Interestingly, DDR1 effects vary depending on cellular type and environment. Regarding the mechanisms, dissociation of actin from myosin IIa enhances DDR1 interaction with myosin IIa and thereby promotes cell migration. Further, DDR1 interacts with the non-receptor tyrosine-kinase Syk and the Syk-mediated cell migration inhibition is blocked in the presence of DDR1. Expression and function of DDR1 on human MKs are still completely unknown. Hypothesis. The hypothesis for the present work is that DDR1 may represent a new collagen receptor of human MKs that regulates their functions in the bone marrow-matrix environment. Methods. MKs were differentiated from human cord blood hemopoietic progenitors. DDR1 expression in mature MKs was evaluated by RT-PCR, immunofluorescence and western blotting analysis. DDR1 activation and interaction with other molecules was evaluated by immunoprecipitation. For cell migration experiments, MKs were seeded in the upper well of transwell migration chambers. MK migration, towards SDF-1 and through filters coated with type I collagen containing or not a DDR1 blocking molecule (DDR1:Fc), was measured after 16 hours. To study the phosphorylation status of DDR1 downstream molecules, western blotting analysis was performed. Results. Our results showed that human MKs express DDR1 at both mRNA and protein levels. MK DDR1 was activated by incubation with type I collagen as demonstrated by immunoprecipitation and probing with an anti-phosphotyrosine antibody. Moreover, upon type I collagen stimulation, MK DDR1 associated with myosin IIA as demonstrated by co-immunoprecipitation. In order to study the role of DDR1 in mediating MK interactions with type I collagen, we took advantage of the recombinant soluble protein DDR1:Fc that was previously shown to block DDR1 activation. Immunoprecipitation analysis demonstrated that DDR1 activation was inhibited in MKs that were let to adhere for 16 hours on type I collagen treated with DDR1:Fc as compared to control. Interestingly, no differences in MLC-2 phosphorylation were observed in MKs adherent on either type I collagen preparations. Consistently, DDR1:Fc did not affect neither MK adhesion nor MK spreading on type I collagen demonstrating that the engagement of other collagen receptor, such as alpha2beta1 integrin, had occurred. However, an important increase of Src and Syk non receptor kinase activation was observed in MKs upon adhesion on DDR1:Fc treated type I collagen with respect to controls. Importantly, a significant decrease (about 40%) of MK migration, through type I collagen, was observed when DDR1 was blocked as compared to controls. These results demonstrated that Syk-mediated migration inhibition is blocked by activation of DDR1 in human MKs. Conclusions. In conclusion, this study provides evidence that the new collagen receptor DDR1 may participate in the regulation of human MK interactions and functions on type I collagen. Although preliminary, these data demonstrate that the bone marrow-matrix environment regulate MK functions through multi-faceted mechanisms that are still to be completely unraveled. Disclosures: No relevant conflicts of interest to declare.


2010 ◽  
Vol 6 (4) ◽  
pp. e1000847 ◽  
Author(s):  
Roberto G. Pozner ◽  
Agustín E. Ure ◽  
Carolina Jaquenod de Giusti ◽  
Lina P. D'Atri ◽  
Joseph E. Italiano ◽  
...  

PLoS ONE ◽  
2020 ◽  
Vol 15 (12) ◽  
pp. e0243840
Author(s):  
Gabriel Santos Rosalem ◽  
Libardo Andrés Gonzáles Torres ◽  
Estevam Barbosa de Las Casas ◽  
Fernando Augusto Siqueira Mathias ◽  
Jeronimo Conceição Ruiz ◽  
...  

Bone marrow (BM) is an organ responsible for crucial processes in living organs, e. g., hematopoiesis. In recent years, Organ-on-a-Chip (OoC) devices have been used to satisfy the need for in vitro systems that better mimic the phenomena occurring in the BM microenvironment. Given the growing interest in these systems and the diversity of developed devices, an integrative systematic literature review is required. We have performed this review, following the PRISMA method aiming to identify the main characteristics and assess the effectiveness of the devices that were developed to represent the BM. A search was performed in the Scopus, PubMed, Web of Science and Science Direct databases using the keywords ((“bone marrow” OR “hematopoietic stem cells” OR “haematopoietic stem cells”) AND (“organ in a” OR “lab on a chip” OR “microfluidic” OR “microfluidic*” OR (“bioreactor” AND “microfluidic*”))). Original research articles published between 2009 and 2020 were included in the review, giving a total of 21 papers. The analysis of these papers showed that their main purpose was to study BM cells biology, mimic BM niches, model pathological BM, and run drug assays. Regarding the fabrication protocols, we have observed that polydimethylsiloxane (PDMS) material and soft lithography method were the most commonly used. To reproduce the microenvironment of BM, most devices used the type I collagen and alginate. Peristaltic and syringe pumps were mostly used for device perfusion. Regarding the advantages compared to conventional methods, there were identified three groups of OoC devices: perfused 3D BM; co-cultured 3D BM; and perfused co-cultured 3D BM. Cellular behavior and mimicking their processes and responses were the mostly commonly studied parameters. The results have demonstrated the effectiveness of OoC devices for research purposes compared to conventional cell cultures. Furthermore, the devices have a wide range of applicability and the potential to be explored.


2020 ◽  
Author(s):  
Xiaoling Zhang ◽  
Liangzhi Du ◽  
Ningbo Zhao ◽  
Lizhe Zhu ◽  
Lei Wang ◽  
...  

Abstract Background: In recent years, researchers have found that exosomes, an important component of intercellular signal transduction and exchange, have great significance in bone tissue repair. In this study, to further promote the development of oral implants, preliminary in vitro experiments were conducted to verify the different concentrations of exosomes from bone marrow mesenchymal stem cells (BMSC-exos) for osteogenesis on the surfaces of titanium sheets.Methods: In this experiment, rabbit bone marrow mesenchymal stem cells(BMSCs) were seeded on the surfaces of 10 mm × 10 mm × 1 mm square titanium sheets and were divided into four groups to investigate their adsorption, proliferation and osteogenesis after treatment with different concentrations of BMSC-exos: 1. BMSCs + titanium + 0 µg/ml BMSC-exos; 2. BMSCs + titanium + 10 µg/ml BMSC-exos; 3. BMSCs + titanium + 25 µg/ml BMSC-exos; and 4. BMSCs + titanium + 50 µg/ml BMSC-exos.Results: Compared with the control group, BMSCs’ adsorption, extension, proliferation and osteogenesis on titanium sheets were significantly increased in the Exosomes group.Conclusions: Exosomes can promote the bone formation of BMSCs on titanium plates by promoting adsorption, extension, proliferation, production of alkaline phosphatase(ALP) and type I collagen and mineralization during the osteogenesis process.


Author(s):  
Arthur J. Wasserman ◽  
Kathy C. Kloos ◽  
David E. Birk

Type I collagen is the predominant collagen in the cornea with type V collagen being a quantitatively minor component. However, the content of type V collagen (10-20%) in the cornea is high when compared to other tissues containing predominantly type I collagen. The corneal stroma has a homogeneous distribution of these two collagens, however, immunochemical localization of type V collagen requires the disruption of type I collagen structure. This indicates that these collagens may be arranged as heterpolymeric fibrils. This arrangement may be responsible for the control of fibril diameter necessary for corneal transparency. The purpose of this work is to study the in vitro assembly of collagen type V and to determine whether the interactions of these collagens influence fibril morphology.


2021 ◽  
Vol 10 (14) ◽  
pp. 3141
Author(s):  
Hyerin Jung ◽  
Yeri Alice Rim ◽  
Narae Park ◽  
Yoojun Nam ◽  
Ji Hyeon Ju

Osteogenesis imperfecta (OI) is a genetic disease characterized by bone fragility and repeated fractures. The bone fragility associated with OI is caused by a defect in collagen formation due to mutation of COL1A1 or COL1A2. Current strategies for treating OI are not curative. In this study, we generated induced pluripotent stem cells (iPSCs) from OI patient-derived blood cells harboring a mutation in the COL1A1 gene. Osteoblast (OB) differentiated from OI-iPSCs showed abnormally decreased levels of type I collagen and osteogenic differentiation ability. Gene correction of the COL1A1 gene using CRISPR/Cas9 recovered the decreased type I collagen expression in OBs differentiated from OI-iPSCs. The osteogenic potential of OI-iPSCs was also recovered by the gene correction. This study suggests a new possibility of treatment and in vitro disease modeling using patient-derived iPSCs and gene editing with CRISPR/Cas9.


Sign in / Sign up

Export Citation Format

Share Document