1
|
Li M, Wang R, Yan T, Tao X, Gao S, Wang Z, Chai Y, Qiu S, Chen W. Dual effects of DLG5 (disks large homolog 5 gene) modulation on chemotherapy-induced thrombocytopenia and nausea/vomiting via the hippo signalling pathway. Br J Pharmacol 2024. [PMID: 39529470 DOI: 10.1111/bph.17391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/23/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND AND PURPOSE The CAPEOX (combination of oxaliplatin and capecitabine) chemotherapy protocol is widely used for colorectal cancer treatment, but it can lead to chemotherapy-induced adverse effects (CRAEs). EXPERIMENTAL APPROACH To uncover the mechanisms and potential biomarkers for CRAE susceptibility, we performed whole-genome sequencing on normal colorectal tissue (CRT) before adjuvant chemotherapy. This is followed by in vivo and in vitro verifications for selected gene and CRAE pair. KEY RESULTS Our analysis revealed specific germline mutations linked to Grade 2 (or higher) chemotherapy-induced thrombocytopenia (CIT) and nausea/vomiting (CINV). Notably, both CRAEs were associated with mutations in the DLG5 gene. We found that DLG5 mutations related to CIT were associated with increased gene expression, while those associated with CINV were linked to suppressed gene expression, as indicated by the Genotype-Tissue Expression (GTEX) database. In megakaryocytes, overexpression of human DLG5 suppressed the hippo signalling pathway and induced YAP expression. In zebrafish, overexpression of human DLG5 not only reduced platelet production but also inhibited thrombus formation. Subsequent qPCR analysis revealed that DLG5 overexpression affected genes involved in cytoskeleton formation and alpha-granule formation, which could impact the normal generation of proplatelets. CONCLUSION AND IMPLICATIONS We identified a series of germline mutations associated with susceptibility to CIT and CINV. Of particular interest, we demonstrated that induced and suppressed DLG5 expression is respectively related to CIT and CINV. These findings shed light on the involvement of the hippo signalling pathway and DLG5 in the development of CRAEs, providing valuable insights into potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Mingming Li
- The SATCM Key Laboratory for New Resources & Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Rong Wang
- The SATCM Key Laboratory for New Resources & Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Yan
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xia Tao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shouhong Gao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zhipeng Wang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yunsheng Chai
- Department of General Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shi Qiu
- The SATCM Key Laboratory for New Resources & Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wansheng Chen
- The SATCM Key Laboratory for New Resources & Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
2
|
Guinard I, Brassard-Jollive N, Ruch L, Weber J, Eckly A, Boscher J, Léon C. Mechanical confinement prevents ectopic platelet release. Proc Natl Acad Sci U S A 2024; 121:e2407829121. [PMID: 39236232 PMCID: PMC11420179 DOI: 10.1073/pnas.2407829121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/03/2024] [Indexed: 09/07/2024] Open
Abstract
Blood platelets are produced by megakaryocytes (MKs), their parent cells, which are in the bone marrow. Once mature, MK pierces through the sinusoid vessel, and the initial protrusion further elongates as proplatelet or buds to release platelets. The mechanisms controlling the decision to initiate proplatelet and platelet formation are unknown. Here, we show that the mechanical properties of the microenvironment prevent proplatelet and platelet release in the marrow stroma while allowing this process in the bloodstream. Loss of marrow confinement following myelosuppression led to inappropriate proplatelet and platelet release into the extravascular space. We further used an inert viscoelastic hydrogel to evaluate the impact of compressive stress. Transcriptional analysis showed that culture in three-dimensional gel induced upregulation of genes related to the Rho-GTPase pathway. We found higher Rho-GTPase activation, myosin light chain phosphorylation and F-actin under mechanical constraints while proplatelet formation was inhibited. The use of latrunculin-A to decrease F-actin promoted microtubule-dependent budding and proplatelet extension inside the gel. Additionally, ex vivo exposure of intact bone marrow to latrunculin-A triggered proplatelet extensions in the interstitial space. In vivo, this confinement-mediated high intracellular tension is responsible for the formation of the peripheral zone, a unique actin-rich structure. Cytoskeleton reorganization induces the disappearance of the peripheral zone upon reaching a liquid milieu to facilitate proplatelet and platelet formation. Hence, our data provide insight into the mechanisms preventing ectopic platelet release in the marrow stroma. Identifying such pathways is especially important for understanding pathologies altering marrow mechanics such as chemotherapy or myelofibrosis.
Collapse
Affiliation(s)
- Ines Guinard
- University of Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, UMR_S1255 Biologie et Pharmacologie des Plaquettes Sanguines (BPPS), FMTS, Strasbourg F-67065, France
| | - Noémie Brassard-Jollive
- University of Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, UMR_S1255 Biologie et Pharmacologie des Plaquettes Sanguines (BPPS), FMTS, Strasbourg F-67065, France
| | - Laurie Ruch
- University of Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, UMR_S1255 Biologie et Pharmacologie des Plaquettes Sanguines (BPPS), FMTS, Strasbourg F-67065, France
| | - Josiane Weber
- University of Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, UMR_S1255 Biologie et Pharmacologie des Plaquettes Sanguines (BPPS), FMTS, Strasbourg F-67065, France
| | - Anita Eckly
- University of Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, UMR_S1255 Biologie et Pharmacologie des Plaquettes Sanguines (BPPS), FMTS, Strasbourg F-67065, France
| | - Julie Boscher
- University of Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, UMR_S1255 Biologie et Pharmacologie des Plaquettes Sanguines (BPPS), FMTS, Strasbourg F-67065, France
| | - Catherine Léon
- University of Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, UMR_S1255 Biologie et Pharmacologie des Plaquettes Sanguines (BPPS), FMTS, Strasbourg F-67065, France
| |
Collapse
|
3
|
Kim OV, Litvinov RI, Gagne AL, French DL, Brass LF, Weisel JW. Megakaryocyte-induced contraction of plasma clots: cellular mechanisms and structural mechanobiology. Blood 2024; 143:548-560. [PMID: 37944157 PMCID: PMC11033616 DOI: 10.1182/blood.2023021545] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/17/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
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 αIIbβ3-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.
Collapse
Affiliation(s)
- Oleg V. Kim
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Biomedical Engineering and Mechanics, Fralin Biomedical Research Institute, Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA
| | - Rustem I. Litvinov
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Alyssa L. Gagne
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Deborah L. French
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Lawrence F. Brass
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - John W. Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
4
|
Guo K, Machlus KR, Camacho V. The many faces of the megakaryocytes and their biological implications. Curr Opin Hematol 2024; 31:1-5. [PMID: 37910197 PMCID: PMC10842450 DOI: 10.1097/moh.0000000000000793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
PURPOSE OF REVIEW Single-cell RNA sequencing studies have revealed transcriptional heterogeneity within the megakaryocytic lineage and the identified unique subsets. In this review, we discuss the functional and phenotypic plasticity of these subpopulations as well as the impacts on health and disease. RECENT FINDINGS Megakaryocytes (MKs) can be transcriptionally categorized into platelet generating, niche supporting, immune, and cycling cells, which are distinguished by their unique gene expression patterns and cellular markers. Additionally, a significant population of these cells has been established to reside in the nonhematopoietic tissues and they display enhanced immune-related characteristics. Combined with the location in which the megakaryocytes exist, these cells can play unique roles dictated by their current environment and biological needs, including responding to changes in pathogen exposure. SUMMARY Advances in megakaryocyte research has elucidated the existence of multiple subpopulations of MKs that serve different functions. These subpopulations implicate a greater potential for MKs to be regulators of health and suggest new avenues for treatments and therapies in related diseases.
Collapse
Affiliation(s)
- Karen Guo
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Kellie R. Machlus
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Virginia Camacho
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Zhao X, Alibhai D, Walsh TG, Tarassova N, Englert M, Birol SZ, Li Y, Williams CM, Neal CR, Burkard P, Cross SJ, Aitken EW, Waller AK, Beltrán JB, Gunning PW, Hardeman EC, Agbani EO, Nieswandt B, Hers I, Ghevaert C, Poole AW. Highly efficient platelet generation in lung vasculature reproduced by microfluidics. Nat Commun 2023; 14:4026. [PMID: 37419900 PMCID: PMC10329040 DOI: 10.1038/s41467-023-39598-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 06/20/2023] [Indexed: 07/09/2023] Open
Abstract
Platelets, small hemostatic blood cells, are derived from megakaryocytes. Both bone marrow and lung are principal sites of thrombopoiesis although underlying mechanisms remain unclear. Outside the body, however, our ability to generate large number of functional platelets is poor. Here we show that perfusion of megakaryocytes ex vivo through the mouse lung vasculature generates substantial platelet numbers, up to 3000 per megakaryocyte. Despite their large size, megakaryocytes are able repeatedly to passage through the lung vasculature, leading to enucleation and subsequent platelet generation intravascularly. Using ex vivo lung and an in vitro microfluidic chamber we determine how oxygenation, ventilation, healthy pulmonary endothelium and the microvascular structure support thrombopoiesis. We also show a critical role for the actin regulator Tropomyosin 4 in the final steps of platelet formation in lung vasculature. This work reveals the mechanisms of thrombopoiesis in lung vasculature and informs approaches to large-scale generation of platelets.
Collapse
Affiliation(s)
- Xiaojuan Zhao
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK.
| | - Dominic Alibhai
- Wolfson BioimagingFacility, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Tony G Walsh
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Nathalie Tarassova
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Maximilian Englert
- University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, D-97080, Germany
| | - Semra Z Birol
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Yong Li
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Christopher M Williams
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Chris R Neal
- Wolfson BioimagingFacility, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Philipp Burkard
- University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, D-97080, Germany
| | - Stephen J Cross
- Wolfson BioimagingFacility, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Elizabeth W Aitken
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Amie K Waller
- University of Cambridge / NHS Blood and Transplant, Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - José Ballester Beltrán
- University of Cambridge / NHS Blood and Transplant, Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Peter W Gunning
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Edna C Hardeman
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Ejaife O Agbani
- Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Bernhard Nieswandt
- University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, D-97080, Germany
| | - Ingeborg Hers
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Cedric Ghevaert
- University of Cambridge / NHS Blood and Transplant, Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Alastair W Poole
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
6
|
Jiang B, Hartzell M, Yu S, Masab M, Lyckholm L. Venous thromboembolism prophylaxis of a patient with MYH-9 related disease and COVID-19 infection: A case report. World J Hematol 2023; 10:1-8. [DOI: 10.5315/wjh.v10.i1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/06/2022] [Accepted: 12/06/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The May-Hegglin anomaly is among a group of genetic disorders known as MYH9-related disease. Patients with inherited platelet disorders such as May-Hegglin anomaly are at a variably increased risk for bleeding due to a com-bination of platelet dysfunction and thrombocytopenia. Patients admitted to the hospital with coronavirus disease 2019 (COVID-19) infection are at an increased risk for a venous thromboembolism event (VTE). The National Institutes of Health COVID-19 treatment guidelines recommend using a prophylactic dose of heparin as VTE prophylaxis for adults who are receiving high-flow oxygen. We describe a patient admitted for COVID-19 infection with pneumonia and a history of May-Hegglin anomaly. The patient presented a challenge to determine prophylactic anticoagulation as there are no clear guidelines for this patient population.
CASE SUMMARY Herein, we describe the case of a 39-year-old woman admitted with acute hypoxic respiratory failure secondary to COVID-19 pneumonia. She had a history of May-Hegglin anomaly and demonstrated risk for bleeding since childhood, including a life-threatening bleeding event at the age of 9 years requiring blood and platelet transfusions. Her baseline platelet count was 40-50 × 109/L throughout her adult life. Her family history was also notable for May-Hegglin disorder in her mother, maternal uncle, maternal grandfather and her son. Computed tomography/ pulmonary angiography revealed bilateral consolidative opacities consistent with multifocal pneumonia. Complete blood count was notable for platelet count of 54 × 109/L. She was admitted for inpatient respiratory support with high-flow oxygen per nasal cannula and was managed with guideline-directed therapy for COVID-19, including baricitinib and dexamethasone. The Hematology/Oncology consultation team was requested to assist with management of VTE prophylaxis in the setting of active COVID-19 infection and an inherited bleeding disorder. After review of the literature and careful consideration of risks and benefits, it was decided to treat the patient with prophylactic enoxaparin. She was closely monitored in the hospital for bleeding and worsening thrombocytopenia. She had no bleeding or signs of VTE. Her respiratory status improved, and she was discharged home after 5 d of hospitalization with supplemental oxygen by nasal cannula and dexamethasone. At the 6-month follow-up, the patient successfully discontinued her home oxygen use after only a few weeks following discharge.
CONCLUSION The patient presented a challenge to determine prophylactic anticoagulation as anticoagulation guidelines exist for patients with COVID-19, but there are no clear guidelines for management of patients with COVID-19 and inherited bleeding disorders, particularly those with MYH9- related disease. She was discharged after recovery from the COVID-19 infection without bleeding or thrombosis. As there are no published guidelines for this situation, we present a pragmatic, informed approach to a patient with MYH9-related disease who had an indication for anticoagulation.
Collapse
Affiliation(s)
- Bei Jiang
- Department of Hematology and Oncology, West Virginia University, Morgantown, WV 26505, United States
| | - Michelle Hartzell
- Department of Internal Medicine, West Virginia University, Morgantown, WV 26505, United States
| | - Stephen Yu
- Department of Internal Medicine, West Virginia University, Morgantown, WV 26505, United States
| | - Muhammad Masab
- Department of Hematology and Oncology, West Virginia University, Morgantown, WV 26505, United States
| | - Laurel Lyckholm
- Department of Hematology and Oncology, West Virginia University, Morgantown, WV 26505, United States
| |
Collapse
|
7
|
Litvinov RI, Weisel JW. Blood clot contraction: Mechanisms, pathophysiology, and disease. Res Pract Thromb Haemost 2023; 7:100023. [PMID: 36760777 PMCID: PMC9903854 DOI: 10.1016/j.rpth.2022.100023] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/09/2022] [Accepted: 11/17/2022] [Indexed: 01/18/2023] Open
Abstract
A State of the Art lecture titled "Blood Clot Contraction: Mechanisms, Pathophysiology, and Disease" was presented at the International Society on Thrombosis and Haemostasis (ISTH) Congress in 2022. This was a systematic description of blood clot contraction or retraction, driven by activated platelets and causing compaction of the fibrin network along with compression of the embedded erythrocytes. The consequences of clot contraction include redistribution of the fibrin-platelet meshwork toward the periphery of the clot and condensation of erythrocytes in the core, followed by their deformation from the biconcave shape into polyhedral cells (polyhedrocytes). These structural signatures of contraction have been found in ex vivo thrombi derived from various locations, which indicated that clots undergo intravital contraction within the blood vessels. In hemostatic clots, tightly packed polyhedrocytes make a nearly impermeable seal that stems bleeding and is impaired in hemorrhagic disorders. In thrombosis, contraction facilitates the local blood flow by decreasing thrombus obstructiveness, reducing permeability, and changing susceptibility to fibrinolytic enzymes. However, in (pro)thrombotic conditions, continuous background platelet activation is followed by platelet exhaustion, refractoriness, and impaired intravital clot contraction, which is associated with weaker thrombi predisposed to embolization. Therefore, assays that detect imperfect in vitro clot contraction have potential diagnostic and prognostic values for imminent or ongoing thrombosis and thrombotic embolism. Collectively, the contraction of blood clots and thrombi is an underappreciated and understudied process that has a pathogenic and clinical significance in bleeding and thrombosis of various etiologies. Finally, we have summarized relevant new data on this topic presented during the 2022 ISTH Congress.
Collapse
Affiliation(s)
- Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
A familial case of MYH9 gene mutation associated with multiple functional and structural platelet abnormalities. Sci Rep 2022; 12:19975. [PMID: 36404341 PMCID: PMC9676191 DOI: 10.1038/s41598-022-24098-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/09/2022] [Indexed: 11/21/2022] Open
Abstract
Mutations in the MYH9 gene result in macrothrombocytopenia often associated with hemorrhages. Here, we studied the function and structure of platelets in three family members with a heterozygous mutation R1933X in the MYH9 gene, characteristic of closely related disorders known as the May-Hegglin anomaly and Sebastian syndrome. The examination included complete blood count, blood smear microscopy, platelet flow cytometry (expression of P-selectin and active integrin αIIbβ3 before and after activation), the kinetics of platelet-driven contraction (retraction) of blood clots, as well as scanning/transmission electron microscopy of platelets. Despite severe thrombocytopenia ranging (36-86) × 109/l, none of the patients had hemorrhages at the time of examination, although they had a history of heavy menstruation, spontaneous ecchymosis, and postpartum hemorrhage. Flow cytometry showed background platelet activation, revealed by overexpression of P-selectin and active αIIbβ3 integrin above normal levels. After TRAP-induced stimulation, the fractions of platelets expressing P-selectin in the proband and her sister were below normal response, indicating partial platelet refractoriness. The initiation of clot contraction was delayed. Electron microscopy revealed giant platelets with multiple filopodia and fusion of α-granules with dilated open canalicular system, containing filamentous and vesicular inclusions. The novel concept implies that the R1933X mutation in the MYH9 gene is associated not only with thrombocytopenia, but also with qualitative structural and functional defects in platelets. Platelet dysfunction includes impaired contractility, which can disrupt the compaction of hemostatic clots, making the clots weak and permeable, therefore predisposing patients with MYH9 gene mutations to the hemorrhagic phenotype.
Collapse
|
9
|
Defective VWF secretion due to the expression of MYH9-RD E1841K mutant in endothelial cells disrupts hemostasis. Blood Adv 2022; 6:4537-4552. [PMID: 35764499 DOI: 10.1182/bloodadvances.2022008011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/08/2022] [Indexed: 11/20/2022] Open
Abstract
Mutations in MYH9, the gene encoding the heavy chain of non-muscle myosin IIa (NMII-A), cause MYH9-related disease (MYH9-RD) that is an autosomal-dominant thrombocytopenia with bleeding tendency. Previously, we showed that NMII-A in endothelial cells (ECs) is critical for hemostasis via regulating von Willebrand factor (VWF) release from Weibel-Palade bodies (WPBs). The aim of this study was to determine the role of the expression of MYH9 mutants in ECs in the pathogenesis of the MYH9-RD bleeding symptom. First, we expressed the 5 most common NMII-A mutants in ECs, and found that E1841K mutant-expressing ECs secreted less VWF than the controls in response to a cAMP signaling agonist. Then, we generated 2 knockin mouse lines, one with Myh9 E1841K in ECs and the other in megakaryocytes. Endothelium-specific E1841K mice exhibited impaired cAMP-induced VWF release and a prolonged bleeding time with normal platelets, while megakaryocyte-specific E1841K mice exhibited macrothrombocytopenia and a prolonged bleeding time with normal VWF release. Finally, we present mechanistic findings that E1841K mutation not only interferes with S1943 phosphorylation and impairs the peripheral distribution of Rab27a positive WPBs in ECs under quiescent condition, but also interferes with S1916 phosphorylation by disrupting the interaction with zyxin and CKIIα, and reduces actin framework formation around WPBs and subsequent VWF secretion under the stimulation by a cAMP agonist. Altogether, our results suggest that impaired cAMP-induced endothelial VWF secretion by E1841K mutant expression may contribute to the MYH9-RD bleeding phenotype.
Collapse
|
10
|
Tilburg J, Becker IC, Italiano JE. Don't you forget about me(gakaryocytes). Blood 2022; 139:3245-3254. [PMID: 34582554 PMCID: PMC9164737 DOI: 10.1182/blood.2020009302] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/08/2021] [Indexed: 11/20/2022] Open
Abstract
Platelets (small, anucleate cell fragments) derive from large precursor cells, megakaryocytes (MKs), that reside in the bone marrow. MKs emerge from hematopoietic stem cells in a complex differentiation process that involves cytoplasmic maturation, including the formation of the demarcation membrane system, and polyploidization. The main function of MKs is the generation of platelets, which predominantly occurs through the release of long, microtubule-rich proplatelets into vessel sinusoids. However, the idea of a 1-dimensional role of MKs as platelet precursors is currently being questioned because of advances in high-resolution microscopy and single-cell omics. On the one hand, recent findings suggest that proplatelet formation from bone marrow-derived MKs is not the only mechanism of platelet production, but that it may also occur through budding of the plasma membrane and in distant organs such as lung or liver. On the other hand, novel evidence suggests that MKs not only maintain physiological platelet levels but further contribute to bone marrow homeostasis through the release of extracellular vesicles or cytokines, such as transforming growth factor β1 or platelet factor 4. The notion of multitasking MKs was reinforced in recent studies by using single-cell RNA sequencing approaches on MKs derived from adult and fetal bone marrow and lungs, leading to the identification of different MK subsets that appeared to exhibit immunomodulatory or secretory roles. In the following article, novel insights into the mechanisms leading to proplatelet formation in vitro and in vivo will be reviewed and the hypothesis of MKs as immunoregulatory cells will be critically discussed.
Collapse
Affiliation(s)
- Julia Tilburg
- Vascular Biology Program, Boston Children's Hospital, Boston, MA
| | | | | |
Collapse
|
11
|
Sun S, Jin C, Si J, Lei Y, Chen K, Cui Y, Liu Z, Liu J, Zhao M, Zhang X, Tang F, Rondina MT, Li Y, Wang QF. Single-cell analysis of ploidy and the transcriptome reveals functional and spatial divergency in murine megakaryopoiesis. Blood 2021; 138:1211-1224. [PMID: 34115843 PMCID: PMC8499048 DOI: 10.1182/blood.2021010697] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/26/2021] [Indexed: 11/20/2022] Open
Abstract
Megakaryocytes (MKs), the platelet progenitor cells, play important roles in hematopoietic stem cell (HSC) maintenance and immunity. However, it is not known whether these diverse programs are executed by a single population or by distinct subsets of cells. Here, we manually isolated primary CD41+ MKs from the bone marrow (BM) of mice and human donors based on ploidy (2N-32N) and performed single-cell RNA sequencing analysis. We found that cellular heterogeneity existed within 3 distinct subpopulations that possess gene signatures related to platelet generation, HSC niche interaction, and inflammatory responses. In situ immunostaining of mouse BM demonstrated that platelet generation and the HSC niche-related MKs were in close physical proximity to blood vessels and HSCs, respectively. Proplatelets, which could give rise to platelets under blood shear forces, were predominantly formed on a platelet generation subset. Remarkably, the inflammatory responses subpopulation, consisting generally of low-ploidy LSP1+ and CD53+ MKs (≤8N), represented ∼5% of total MKs in the BM. These MKs could specifically respond to pathogenic infections in mice. Rapid expansion of this population was accompanied by strong upregulation of a preexisting PU.1- and IRF-8-associated monocytic-like transcriptional program involved in pathogen recognition and clearance as well as antigen presentation. Consistently, isolated primary CD53+ cells were capable of engulfing and digesting bacteria and stimulating T cells in vitro. Together, our findings uncover new molecular, spatial, and functional heterogeneity within MKs in vivo and demonstrate the existence of a specialized MK subpopulation that may act as a new type of immune cell.
Collapse
Affiliation(s)
- Shu Sun
- Chinese Academy of Sciences (CAS) Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, CAS, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chen Jin
- Chinese Academy of Sciences (CAS) Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, CAS, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jia Si
- Chinese Academy of Sciences (CAS) Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, CAS, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ying Lei
- Chinese Academy of Sciences (CAS) Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, CAS, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kunying Chen
- Chinese Academy of Sciences (CAS) Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, CAS, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yueli Cui
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing, China
- Biomedical Institute for Pioneering Investigation via Convergence, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Zhenbo Liu
- Chinese Academy of Sciences (CAS) Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, CAS, Beijing, China
- China National Center for Bioinformation, Beijing, China
| | - Jiang Liu
- Chinese Academy of Sciences (CAS) Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, CAS, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Meng Zhao
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Fuchou Tang
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing, China
- Biomedical Institute for Pioneering Investigation via Convergence, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Matthew T Rondina
- Department of Internal Medicine and Pathology, and the Molecular Medicine Program, University of Utah, Salt Lake City, UT; and
- Geriatric Research Education and Clinical Center, George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT
| | - Yueying Li
- Chinese Academy of Sciences (CAS) Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, CAS, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qian-Fei Wang
- Chinese Academy of Sciences (CAS) Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, CAS, Beijing, China
- China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
12
|
Noh JY. Megakaryopoiesis and Platelet Biology: Roles of Transcription Factors and Emerging Clinical Implications. Int J Mol Sci 2021; 22:ijms22179615. [PMID: 34502524 PMCID: PMC8431765 DOI: 10.3390/ijms22179615] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 12/13/2022] Open
Abstract
Platelets play a critical role in hemostasis and thrombus formation. Platelets are small, anucleate, and short-lived blood cells that are produced by the large, polyploid, and hematopoietic stem cell (HSC)-derived megakaryocytes in bone marrow. Approximately 3000 platelets are released from one megakaryocyte, and thus, it is important to understand the physiologically relevant mechanism of development of mature megakaryocytes. Many genes, including several key transcription factors, have been shown to be crucial for platelet biogenesis. Mutations in these genes can perturb megakaryopoiesis or thrombopoiesis, resulting in thrombocytopenia. Metabolic changes owing to inflammation, ageing, or diseases such as cancer, in which platelets play crucial roles in disease development, can also affect platelet biogenesis. In this review, I describe the characteristics of platelets and megakaryocytes in terms of their differentiation processes. The role of several critical transcription factors have been discussed to better understand the changes in platelet biogenesis that occur during disease or ageing.
Collapse
Affiliation(s)
- Ji-Yoon Noh
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| |
Collapse
|
13
|
Mbiandjeu S, Balduini A, Malara A. Megakaryocyte Cytoskeletal Proteins in Platelet Biogenesis and Diseases. Thromb Haemost 2021; 122:666-678. [PMID: 34218430 DOI: 10.1055/s-0041-1731717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Thrombopoiesis governs the formation of blood platelets in bone marrow by converting megakaryocytes into long, branched proplatelets on which individual platelets are assembled. The megakaryocyte cytoskeleton responds to multiple microenvironmental cues, including chemical and mechanical stimuli, sustaining the platelet shedding. During the megakaryocyte's life cycle, cytoskeletal networks organize cell shape and content, connect them physically and biochemically to the bone marrow vascular niche, and enable the release of platelets into the bloodstream. While the basic building blocks of the cytoskeleton have been studied extensively, new sets of cytoskeleton regulators have emerged as critical components of the dynamic protein network that supports platelet production. Understanding how the interaction of individual molecules of the cytoskeleton governs megakaryocyte behavior is essential to improve knowledge of platelet biogenesis and develop new therapeutic strategies for inherited thrombocytopenias caused by alterations in the cytoskeletal genes.
Collapse
Affiliation(s)
- Serge Mbiandjeu
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | | |
Collapse
|
14
|
Garcia A, Dunoyer-Geindre S, Nolli S, Strassel C, Reny JL, Fontana P. miR-204-5p and Platelet Function Regulation: Insight into a Mechanism Mediated by CDC42 and GPIIbIIIa. Thromb Haemost 2021; 121:1206-1219. [PMID: 33940656 PMCID: PMC8421094 DOI: 10.1055/a-1497-9649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Several platelet-derived microRNAs are associated with platelet reactivity (PR) and clinical outcome in cardiovascular patients. We previously showed an association between miR-204-5p and PR in stable cardiovascular patients, but data on functional mechanisms are lacking. AIMS To validate miR-204-5p as a regulator of PR in platelet-like structures (PLS) derived from human megakaryocytes and to address mechanistic issues. METHODS Human hematopoietic stem cells were differentiated into megakaryocytes, enabling the transfection of miR-204-5p and the recovery of subsequent PLS. The morphology of transfected megakaryocytes and PLS was characterized using flow cytometry and microscopy. The functional impact of miR-204-5p was assessed using a flow assay, the quantification of the activated form of the GPIIbIIIa receptor, and a fibrinogen-binding assay. Quantitative polymerase chain reaction and western blot were used to evaluate the impact of miR-204-5p on a validated target, CDC42. The impact of CDC42 modulation was investigated using a silencing strategy. RESULTS miR-204-5p transfection induced cytoskeletal changes in megakaryocytes associated with the retracted protrusion of proPLS, but it had no impact on the number of PLS released. Functional assays showed that the PLS produced by megakaryocytes transfected with miR-204-5p were more reactive than controls. This phenotype is mediated by the regulation of GPIIbIIIa expression, a key contributor in platelet-fibrinogen interaction. Similar results were obtained after CDC42 silencing, suggesting that miR-204-5p regulates PR, at least in part, via CDC42 downregulation. CONCLUSION We functionally validated miR-204-5p as a regulator of the PR that occurs through CDC42 downregulation and regulation of fibrinogen receptor expression.
Collapse
Affiliation(s)
- Alix Garcia
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Séverine Nolli
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Jean-Luc Reny
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of General Internal Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Pierre Fontana
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of Angiology and Haemostasis, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
15
|
Heib T, Hermanns HM, Manukjan G, Englert M, Kusch C, Becker IC, Gerber A, Wackerbarth LM, Burkard P, Dandekar T, Balkenhol J, Jahn D, Beck S, Meub M, Dütting S, Stigloher C, Sauer M, Cherpokova D, Schulze H, Brakebusch C, Nieswandt B, Nagy Z, Pleines I. RhoA/Cdc42 signaling drives cytoplasmic maturation but not endomitosis in megakaryocytes. Cell Rep 2021; 35:109102. [PMID: 33979620 DOI: 10.1016/j.celrep.2021.109102] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/20/2021] [Accepted: 04/18/2021] [Indexed: 12/15/2022] Open
Abstract
Megakaryocytes (MKs), the precursors of blood platelets, are large, polyploid cells residing mainly in the bone marrow. We have previously shown that balanced signaling of the Rho GTPases RhoA and Cdc42 is critical for correct MK localization at bone marrow sinusoids in vivo. Using conditional RhoA/Cdc42 double-knockout (DKO) mice, we reveal here that RhoA/Cdc42 signaling is dispensable for the process of polyploidization in MKs but essential for cytoplasmic MK maturation. Proplatelet formation is virtually abrogated in the absence of RhoA/Cdc42 and leads to severe macrothrombocytopenia in DKO animals. The MK maturation defect is associated with downregulation of myosin light chain 2 (MLC2) and β1-tubulin, as well as an upregulation of LIM kinase 1 and cofilin-1 at both the mRNA and protein level and can be linked to impaired MKL1/SRF signaling. Our findings demonstrate that MK endomitosis and cytoplasmic maturation are separately regulated processes, and the latter is critically controlled by RhoA/Cdc42.
Collapse
Affiliation(s)
- Tobias Heib
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Heike M Hermanns
- Department of Internal Medicine II, Hepatology Research Laboratory, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Georgi Manukjan
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Maximilian Englert
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Charly Kusch
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Isabelle Carlotta Becker
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Annika Gerber
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Lou Martha Wackerbarth
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Philipp Burkard
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Johannes Balkenhol
- Department of Internal Medicine II, Hepatology Research Laboratory, University Hospital Würzburg, 97080 Würzburg, Germany; Department of Bioinformatics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Daniel Jahn
- Department of Internal Medicine II, Hepatology Research Laboratory, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Sarah Beck
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Mara Meub
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Sebastian Dütting
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Christian Stigloher
- Imaging Core Facility, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Deya Cherpokova
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Harald Schulze
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Cord Brakebusch
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany.
| | - Zoltan Nagy
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Irina Pleines
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany; Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany.
| |
Collapse
|
16
|
Bornert A, Boscher J, Pertuy F, Eckly A, Stegner D, Strassel C, Gachet C, Lanza F, Léon C. Cytoskeletal-based mechanisms differently regulate in vivo and in vitro proplatelet formation. Haematologica 2021; 106:1368-1380. [PMID: 32327502 PMCID: PMC8094084 DOI: 10.3324/haematol.2019.239111] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Indexed: 12/23/2022] Open
Abstract
Platelets are produced by bone marrow megakaryocytes through cytoplasmic protrusions, named native proplatelets (nPPT), into blood vessels. Proplatelets also refer to protrusions observed in megakaryocyte culture (cultured proplatelets [cPPT]) which are morphologically different. Contrary to cPPT, the mechanisms of nPPT formation are poorly understood. We show here in living mice that nPPT elongation is in equilibrium between protrusion and retraction forces mediated by myosin-IIA. We also found, using wild-type and b1-tubulin-deficient mice, that microtubule behavior differs between cPPT and nPPT, being absolutely required in vitro, while less critical in vivo. Remarkably, microtubule depolymerization in myosin-deficient mice did not affect nPPT elongation. We then calculated that blood Stokes’ forces may be sufficient to promote nPPT extension, independently of myosin and microtubules. Together, we propose a new mechanism for nPPT extension that might explain contradictions between severely affected cPPT production and moderate platelet count defects in some patients and animal models.
Collapse
Affiliation(s)
- Alicia Bornert
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - Julie Boscher
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - Fabien Pertuy
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - Anita Eckly
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - David Stegner
- University Hospital Würzburg and Rudolf Virchow Center for Experimental Biomedicine, Germany
| | - Catherine Strassel
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - Christian Gachet
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - François Lanza
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - Catherine Léon
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| |
Collapse
|
17
|
Rani GF, Preham O, Ashwin H, Brown N, Hitchcock IS, Kaye PM. Dissecting pathways to thrombocytopenia in a mouse model of visceral leishmaniasis. Blood Adv 2021; 5:1627-1637. [PMID: 33710338 PMCID: PMC7993103 DOI: 10.1182/bloodadvances.2020004082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/08/2021] [Indexed: 12/24/2022] Open
Abstract
Visceral leishmaniasis is an important yet neglected parasitic disease caused by infection with Leishmania donovani or L infantum. Disease manifestations include fever, weight loss, hepatosplenomegaly, immune dysregulation, and extensive hematological complications. Thrombocytopenia is a dominant hematological feature seen in both humans and experimental models, but the mechanisms behind this infection-driven thrombocytopenia remain poorly understood. Using a murine model of experimental visceral leishmaniasis (EVL), we demonstrated a progressive decrease in platelets from day 14 after infection, culminating in severe thrombocytopenia by day 28. Plasma thrombopoietin (TPO) levels were reduced in infected mice, at least in part because of the alterations in the liver microenvironment associated with granulomatous inflammation. Bone marrow (BM) megakaryocyte cytoplasmic maturation was significantly reduced. In addition to a production deficit, we identified significant increases in platelet clearance. L donovani-infected splenectomized mice were protected from thrombocytopenia compared with sham operated infected mice and had a greater response to exogenous TPO. Furthermore, infection led to higher levels of platelet opsonization and desialylation, both associated with platelet clearance in spleen and liver, respectively. Critically, these changes could be reversed rapidly by drug treatment to reduce parasite load or by administration of TPO agonists. In summary, our findings demonstrate that the mechanisms underpinning thrombocytopenia in EVL are multifactorial and reversible, with no obvious residual damage to the BM microenvironment.
Collapse
Affiliation(s)
| | - Olivier Preham
- York Biomedical Research Institute, Hull York Medical School, and
| | - Helen Ashwin
- York Biomedical Research Institute, Hull York Medical School, and
| | - Najmeeyah Brown
- York Biomedical Research Institute, Hull York Medical School, and
| | - Ian S Hitchcock
- Department of Biology, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Paul M Kaye
- York Biomedical Research Institute, Hull York Medical School, and
| |
Collapse
|
18
|
Vainchenker W, Arkoun B, Basso-Valentina F, Lordier L, Debili N, Raslova H. Role of Rho-GTPases in megakaryopoiesis. Small GTPases 2021; 12:399-415. [PMID: 33570449 PMCID: PMC8583283 DOI: 10.1080/21541248.2021.1885134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Megakaryocytes (MKs) are the bone marrow (BM) cells that generate blood platelets by a process that requires: i) polyploidization responsible for the increased MK size and ii) cytoplasmic organization leading to extension of long pseudopods, called proplatelets, through the endothelial barrier to allow platelet release into blood. Low level of localized RHOA activation prevents actomyosin accumulation at the cleavage furrow and participates in MK polyploidization. In the platelet production, RHOA and CDC42 play opposite, but complementary roles. RHOA inhibits both proplatelet formation and MK exit from BM, whereas CDC42 drives the development of the demarcation membranes and MK migration in BM. Moreover, the RhoA or Cdc42 MK specific knock-out in mice and the genetic alterations in their down-stream effectors in human induce a thrombocytopenia demonstrating their key roles in platelet production. A better knowledge of Rho-GTPase signalling is thus necessary to develop therapies for diseases associated with platelet production defects. Abbreviations: AKT: Protein Kinase BARHGEF2: Rho/Rac Guanine Nucleotide Exchange Factor 2ARP2/3: Actin related protein 2/3BM: Bone marrowCDC42: Cell division control protein 42 homologCFU-MK: Colony-forming-unit megakaryocyteCIP4: Cdc42-interacting protein 4mDIA: DiaphanousDIAPH1; Protein diaphanous homolog 1ECT2: Epithelial Cell Transforming Sequence 2FLNA: Filamin AGAP: GTPase-activating proteins or GTPase-accelerating proteinsGDI: GDP Dissociation InhibitorGEF: Guanine nucleotide exchange factorHDAC: Histone deacetylaseLIMK: LIM KinaseMAL: Megakaryoblastic leukaemiaMARCKS: Myristoylated alanine-rich C-kinase substrateMKL: Megakaryoblastic leukaemiaMLC: Myosin light chainMRTF: Myocardin Related Transcription FactorOTT: One-Twenty Two ProteinPACSIN2: Protein Kinase C And Casein Kinase Substrate In Neurons 2PAK: P21-Activated KinasePDK: Pyruvate Dehydrogenase kinasePI3K: Phosphoinositide 3-kinasePKC: Protein kinase CPTPRJ: Protein tyrosine phosphatase receptor type JRAC: Ras-related C3 botulinum toxin substrate 1RBM15: RNA Binding Motif Protein 15RHO: Ras homologousROCK: Rho-associated protein kinaseSCAR: Suppressor of cAMP receptorSRF: Serum response factorSRC: SarcTAZ: Transcriptional coactivator with PDZ motifTUBB1: Tubulin β1VEGF: Vascular endothelial growth factorWAS: Wiskott Aldrich syndromeWASP: Wiskott Aldrich syndrome proteinWAVE: WASP-family verprolin-homologous proteinWIP: WASP-interacting proteinYAP: Yes-associated protein
Collapse
Affiliation(s)
- William Vainchenker
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France.,GrEX, Sorbonne Paris Cité, Paris, France
| | - Brahim Arkoun
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France.,GrEX, Sorbonne Paris Cité, Paris, France
| | - Francesca Basso-Valentina
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France.,Université Sorbonne Paris Cité/Université Paris Dideront, Paris, France
| | - Larissa Lordier
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France
| | - Najet Debili
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France
| | - Hana Raslova
- INSERM, UMR 1287, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.,Université Paris Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Gustave Roussy, Villejuif, France
| |
Collapse
|
19
|
Megakaryocyte migration defects due to nonmuscle myosin IIA mutations underlie thrombocytopenia in MYH9-related disease. Blood 2021; 135:1887-1898. [PMID: 32315395 DOI: 10.1182/blood.2019003064] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/29/2020] [Indexed: 12/17/2022] Open
Abstract
Megakaryocytes (MKs), the precursor cells for platelets, migrate from the endosteal niche of the bone marrow (BM) toward the vasculature, extending proplatelets into sinusoids, where circulating blood progressively fragments them into platelets. Nonmuscle myosin IIA (NMIIA) heavy chain gene (MYH9) mutations cause macrothrombocytopenia characterized by fewer platelets with larger sizes leading to clotting disorders termed myosin-9-related disorders (MYH9-RDs). MYH9-RD patient MKs have proplatelets with thicker and fewer branches that produce fewer and larger proplatelets, which is phenocopied in mouse Myh9-RD models. Defective proplatelet formation is considered to be the principal mechanism underlying the macrothrombocytopenia phenotype. However, MYH9-RD patient MKs may have other defects, as NMII interactions with actin filaments regulate physiological processes such as chemotaxis, cell migration, and adhesion. How MYH9-RD mutations affect MK migration and adhesion in BM or NMIIA activity and assembly prior to proplatelet production remain unanswered. NMIIA is the only NMII isoform expressed in mature MKs, permitting exploration of these questions without complicating effects of other NMII isoforms. Using mouse models of MYH9-RD (NMIIAR702C+/-GFP+/-, NMIIAD1424N+/-, and NMIIAE1841K+/-) and in vitro assays, we investigated MK distribution in BM, chemotaxis toward stromal-derived factor 1, NMIIA activity, and bipolar filament assembly. Results indicate that different MYH9-RD mutations suppressed MK migration in the BM without compromising bipolar filament formation but led to divergent adhesion phenotypes and NMIIA contractile activities depending on the mutation. We conclude that MYH9-RD mutations impair MK chemotaxis by multiple mechanisms to disrupt migration toward the vasculature, impairing proplatelet release and causing macrothrombocytopenia.
Collapse
|
20
|
Wang H, He J, Xu C, Chen X, Yang H, Shi S, Liu C, Zeng Y, Wu D, Bai Z, Wang M, Wen Y, Su P, Xia M, Huang B, Ma C, Bian L, Lan Y, Cheng T, Shi L, Liu B, Zhou J. Decoding Human Megakaryocyte Development. Cell Stem Cell 2020; 28:535-549.e8. [PMID: 33340451 DOI: 10.1016/j.stem.2020.11.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 09/25/2020] [Accepted: 11/10/2020] [Indexed: 12/25/2022]
Abstract
Despite our growing understanding of embryonic immune development, rare early megakaryocytes (MKs) remain relatively understudied. Here we used single-cell RNA sequencing of human MKs from embryonic yolk sac (YS) and fetal liver (FL) to characterize the transcriptome, cellular heterogeneity, and developmental trajectories of early megakaryopoiesis. In the YS and FL, we found heterogeneous MK subpopulations with distinct developmental routes and patterns of gene expression that could reflect early functional specialization. Intriguingly, we identified a subpopulation of CD42b+CD14+ MKs in vivo that exhibit high expression of genes associated with immune responses and can also be derived from human embryonic stem cells (hESCs) in vitro. Furthermore, we identified THBS1 as an early marker for MK-biased embryonic endothelial cells. Overall, we provide important insights and invaluable resources for dissection of the molecular and cellular programs underlying early human megakaryopoiesis.
Collapse
Affiliation(s)
- Hongtao Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Jian He
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing 100071, China
| | - Changlu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Xiaoyuan Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Hua Yang
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin 300052, China
| | - Shujuan Shi
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin 300052, China
| | - Cuicui Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Yang Zeng
- Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Dan Wu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Zhijie Bai
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing 100071, China
| | - Mengge Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Yuqi Wen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Pei Su
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Meijuan Xia
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Baiming Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Chunyu Ma
- Department of Gynecology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Lihong Bian
- Department of Gynecology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Yu Lan
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China
| | - Lihong Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China.
| | - Bing Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing 100071, China; Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China.
| | - Jiaxi Zhou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin 300020, China.
| |
Collapse
|
21
|
Zingariello M, Rosti V, Vannucchi AM, Guglielmelli P, Mazzarini M, Barosi G, Genova ML, Migliaccio AR. Shared and Distinctive Ultrastructural Abnormalities Expressed by Megakaryocytes in Bone Marrow and Spleen From Patients With Myelofibrosis. Front Oncol 2020; 10:584541. [PMID: 33312951 PMCID: PMC7701330 DOI: 10.3389/fonc.2020.584541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/19/2020] [Indexed: 12/21/2022] Open
Abstract
Numerous studies have documented ultrastructural abnormalities in malignant megakaryocytes from bone marrow (BM) of myelofibrosis patients but the morphology of these cells in spleen, an important extramedullary site in this disease, was not investigated as yet. By transmission-electron microscopy, we compared the ultrastructural features of megakaryocytes from BM and spleen of myelofibrosis patients and healthy controls. The number of megakaryocytes was markedly increased in both BM and spleen. However, while most of BM megakaryocytes are immature, those from spleen appear mature with well-developed demarcation membrane systems (DMS) and platelet territories and are surrounded by platelets. In BM megakaryocytes, paucity of DMS is associated with plasma (thick with protrusions) and nuclear (dilated with large pores) membrane abnormalities and presence of numerous glycosomes, suggesting a skewed metabolism toward insoluble polyglucosan accumulation. By contrast, the membranes of the megakaryocytes from the spleen were normal but these cells show mitochondria with reduced crests, suggesting deficient aerobic energy-metabolism. These distinctive morphological features suggest that malignant megakaryocytes from BM and spleen express distinctive metabolic impairments that may play different roles in the pathogenesis of myelofibrosis.
Collapse
Affiliation(s)
- Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry Biotechnology and Advanced Diagnosis, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alessandro M Vannucchi
- CRIMM; Center Research and Innovation of Myeloproliferative Neoplasms, AOUC, University of Florence, Florence, Italy
| | - Paola Guglielmelli
- CRIMM; Center Research and Innovation of Myeloproliferative Neoplasms, AOUC, University of Florence, Florence, Italy
| | - Maria Mazzarini
- Biomedical and Neuromotor Sciences, Alma Mater University Bologna, Bologna, Italy
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry Biotechnology and Advanced Diagnosis, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Maria Luisa Genova
- Biomedical and Neuromotor Sciences, Alma Mater University Bologna, Bologna, Italy
| | - Anna Rita Migliaccio
- Biomedical and Neuromotor Sciences, Alma Mater University Bologna, Bologna, Italy.,Myeloproliferative Neoplasm-Research Consortium, New York, NY, United States
| |
Collapse
|
22
|
Eckly A, Scandola C, Oprescu A, Michel D, Rinckel JY, Proamer F, Hoffmann D, Receveur N, Léon C, Bear JE, Ghalloussi D, Harousseau G, Bergmeier W, Lanza F, Gaits-Iacovoni F, de la Salle H, Gachet C. Megakaryocytes use in vivo podosome-like structures working collectively to penetrate the endothelial barrier of bone marrow sinusoids. J Thromb Haemost 2020; 18:2987-3001. [PMID: 32702204 DOI: 10.1111/jth.15024] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/18/2020] [Accepted: 07/16/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND Blood platelets are anucleate cell fragments that prevent bleeding and minimize blood vessel injury. They are formed from the cytoplasm of megakaryocytes located in the bone marrow. For successful platelet production, megakaryocyte fragments must pass through the sinusoid endothelial barrier by a cell biology process unique to these giant cells as compared with erythrocytes and leukocytes. Currently, the mechanisms by which megakaryocytes interact and progress through the endothelial cells are not understood, resulting in a significant gap in our knowledge of platelet production. OBJECTIVE The aim of this study was to investigate how megakaryocytes interact and progress through the endothelial cells of mouse bone marrow sinusoids. METHODS We used a combination of fluorescence, electron, and three-dimensional microscopy to characterize the cellular events between megakaryocytes and endothelial cells. RESULTS We identified protrusive, F-actin-based podosome-like structures, called in vivo-MK podosomes, which initiate the formation of pores through endothelial cells. These structures present a collective and spatial organization through their interconnection via a contractile network of actomyosin, essential to regulate the endothelial openings. This ensures proper passage of megakaryocyte-derived processes into the blood circulation to promote thrombopoiesis. CONCLUSION This study provides novel insight into the in vivo function of podosomes of megakaryocytes with critical importance to platelet production.
Collapse
Affiliation(s)
- Anita Eckly
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - Cyril Scandola
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - Antoine Oprescu
- INSERM U1048, I2MC, Université Paul Sabatier, Toulouse, France
| | - Deborah Michel
- INSERM U1048, I2MC, Université Paul Sabatier, Toulouse, France
| | - Jean-Yves Rinckel
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - Fabienne Proamer
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - David Hoffmann
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - Nicolas Receveur
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - Catherine Léon
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - James E Bear
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Dorsaf Ghalloussi
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Gabriel Harousseau
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Francois Lanza
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | | | - Henri de la Salle
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - Christian Gachet
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| |
Collapse
|
23
|
Bächer C, Bender M, Gekle S. Flow-accelerated platelet biogenesis is due to an elasto-hydrodynamic instability. Proc Natl Acad Sci U S A 2020; 117:18969-18976. [PMID: 32719144 PMCID: PMC7431004 DOI: 10.1073/pnas.2002985117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Blood platelets are formed by fragmentation of long membrane extensions from bone marrow megakaryocytes in the blood flow. Using lattice-Boltzmann/immersed boundary simulations we propose a biological Rayleigh-Plateau instability as the biophysical mechanism behind this fragmentation process. This instability is akin to the surface tension-induced breakup of a liquid jet but is driven by active cortical processes including actomyosin contractility and microtubule sliding. Our fully three-dimensional simulations highlight the crucial role of actomyosin contractility, which is required to trigger the instability, and illustrate how the wavelength of the instability determines the size of the final platelets. The elasto-hydrodynamic origin of the fragmentation explains the strong acceleration of platelet biogenesis in the presence of an external flow, which we observe in agreement with experiments. Our simulations then allow us to disentangle the influence of specific flow conditions: While a homogeneous flow with uniform velocity leads to the strongest acceleration, a shear flow with a linear velocity gradient can cause fusion events of two developing platelet-sized swellings during fragmentation. A fusion event may lead to the release of larger structures which are observable as preplatelets in experiments. Together, our findings strongly indicate a mainly physical origin of fragmentation and regulation of platelet size in flow-accelerated platelet biogenesis.
Collapse
Affiliation(s)
- Christian Bächer
- Biofluid Simulation and Modeling, Theoretische Physik VI, University of Bayreuth, 95447 Bayreuth, Germany;
| | - Markus Bender
- Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center, 97080 Würzburg, Germany
| | - Stephan Gekle
- Biofluid Simulation and Modeling, Theoretische Physik VI, University of Bayreuth, 95447 Bayreuth, Germany;
| |
Collapse
|
24
|
Asensio-Juárez G, Llorente-González C, Vicente-Manzanares M. Linking the Landscape of MYH9-Related Diseases to the Molecular Mechanisms that Control Non-Muscle Myosin II-A Function in Cells. Cells 2020; 9:E1458. [PMID: 32545517 PMCID: PMC7348894 DOI: 10.3390/cells9061458] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
The MYH9 gene encodes the heavy chain (MHCII) of non-muscle myosin II A (NMII-A). This is an actin-binding molecular motor essential for development that participates in many crucial cellular processes such as adhesion, cell migration, cytokinesis and polarization, maintenance of cell shape and signal transduction. Several types of mutations in the MYH9 gene cause an array of autosomal dominant disorders, globally known as MYH9-related diseases (MYH9-RD). These include May-Hegglin anomaly (MHA), Epstein syndrome (EPS), Fechtner syndrome (FTS) and Sebastian platelet syndrome (SPS). Although caused by different MYH9 mutations, all patients present macrothrombocytopenia, but may later display other pathologies, including loss of hearing, renal failure and presenile cataracts. The correlation between the molecular and cellular effects of the different mutations and clinical presentation are beginning to be established. In this review, we correlate the defects that MYH9 mutations cause at a molecular and cellular level (for example, deficient filament formation, altered ATPase activity or actin-binding) with the clinical presentation of the syndromes in human patients. We address why these syndromes are tissue restricted, and the existence of possible compensatory mechanisms, including residual activity of mutant NMII-A and/ or the formation of heteropolymers or co-polymers with other NMII isoforms.
Collapse
Affiliation(s)
| | | | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain; (G.A.-J.); (C.L.-G.)
| |
Collapse
|
25
|
Xu WH, Guo HH, Chen SJ, Wang YZ, Lin ZH, Huang XD, Tang HJ, He YH, Sun JJ, Gan L. Transcriptome analysis revealed changes of multiple genes involved in muscle hardness in grass carp (Ctenopharyngodon idellus) fed with faba bean meal. Food Chem 2020; 314:126205. [PMID: 31968291 DOI: 10.1016/j.foodchem.2020.126205] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/24/2019] [Accepted: 01/11/2020] [Indexed: 11/26/2022]
Abstract
An 8-week feeding trial and transcriptome analysis were conducted to investigate the potential mechanism of muscle-hardening caused by faba bean in grass carp (Ctenopharyngodon idellus). Ordinary grass carp (fed with practical diet) and crisp grass carp (fed with faba bean meal) groups were designed. Lower water holding capacity and higher some texture parameters were observed in the muscle of crisp grass carp compared with another group. 19.62 GB clean reads were generated, and total 1354 genes exhibiting differentially expression were identified (FDR < 0.05). Genes function enrichment revealed up-regulated genes in crisp grass carp mainly in response to myofibroblast proliferation, while down-regulated genes in response to immune regulation. Consistent with this, the tight junction pathway and the NF-κB signaling pathway were likewise significantly enriched. In summary, this study identified several candidate genes and putative signaling pathways deserving further investigation to the mechanism of muscle-hardening in fish fed with faba bean.
Collapse
Affiliation(s)
- Wei-Hua Xu
- College of Marine Science, South China Agricultural University, Guangzhou 510642, China; School of Marine Sciences & Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, South China Agricultural University, Guangzhou 510642, China
| | - Hong-Hong Guo
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, GuangZhou 510275, China
| | - Shi-Jun Chen
- College of Marine Science, South China Agricultural University, Guangzhou 510642, China; School of Marine Sciences & Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, South China Agricultural University, Guangzhou 510642, China
| | - Yan-Zhi Wang
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, GuangZhou 510275, China
| | - Zhuo-Heng Lin
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, GuangZhou 510275, China
| | - Xian-De Huang
- College of Marine Science, South China Agricultural University, Guangzhou 510642, China; School of Marine Sciences & Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, South China Agricultural University, Guangzhou 510642, China
| | - Hui-Juan Tang
- College of Marine Science, South China Agricultural University, Guangzhou 510642, China; School of Marine Sciences & Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, South China Agricultural University, Guangzhou 510642, China
| | - Yu-Hui He
- College of Marine Science, South China Agricultural University, Guangzhou 510642, China; School of Marine Sciences & Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, South China Agricultural University, Guangzhou 510642, China
| | - Ji-Jia Sun
- College of Marine Science, South China Agricultural University, Guangzhou 510642, China; School of Marine Sciences & Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, South China Agricultural University, Guangzhou 510642, China
| | - Lian Gan
- College of Marine Science, South China Agricultural University, Guangzhou 510642, China; School of Marine Sciences & Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
26
|
Scandola C, Erhardt M, Rinckel JY, Proamer F, Gachet C, Eckly A. Use of electron microscopy to study megakaryocytes. Platelets 2020; 31:589-598. [PMID: 31903825 DOI: 10.1080/09537104.2019.1708885] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Electron microscopy (EM) has a long history in megakaryocyte (MK) cellular biology. This chapter shows how the electron microscope, since its first appearance almost 90 years ago, has occupied center stage in the studies of MK morphology and function. It describes some of the more productive EM techniques that have shaped our understanding of the physiology of thrombopoiesis. These include the standard transmission and scanning EM techniques as well as the new imaging methods, correlative microscopy and volume EM which provide information on the 3D organization of MKs on different scales: single organelles, whole cells and tissues. For each technique, we list the advantages and limitations, the resolution that can be achieved, the technical difficulties and the applications in MK biology.
Collapse
Affiliation(s)
- Cyril Scandola
- INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Université de Strasbourg , Strasbourg, France
| | | | - Jean-Yves Rinckel
- INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Université de Strasbourg , Strasbourg, France
| | - Fabienne Proamer
- INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Université de Strasbourg , Strasbourg, France
| | - Christian Gachet
- INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Université de Strasbourg , Strasbourg, France
| | - Anita Eckly
- INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Université de Strasbourg , Strasbourg, France
| |
Collapse
|
27
|
Ai Q, Zhao L, Yin J, Jiang L, Jin Q, Hu X, Chen S. A novel de novo MYH9 mutation in MYH9-related disease: A case report and review of literature. Medicine (Baltimore) 2020; 99:e18887. [PMID: 31977897 PMCID: PMC7004752 DOI: 10.1097/md.0000000000018887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
INTRODUCTION MYH9-related disease (MYH9-RD) is a rare autosomal dominant disorder caused by mutations in MYH9, which is responsible for encoding nonmuscle myosin heavy chains IIA (NMMHCIIA). MYH9-RD is clinically characterized by congenital macrothrombocytopenia, granulocyte inclusions variably associated with the risk of developing progressive sensorineural deafness, cataracts and nephropathy. PATIENT CONCERNS A 5-year-old boy had a history of a mild bleeding tendency and chronic thrombocytopenia, first identified at four months of age. No other family members were noted to have similar clinical features or hematologic disorders. DIAGNOSES The boy was diagnosed with MYH9-RD. Light microscopic examination of peripheral blood films (Wright-Giemsa stain) showed marked platelet macrocytosis with giant platelets and basophilic Döhle-like inclusions in 83% of the neutrophils. Immunofluorescence analysis disclosed a type II pattern, manifested by neutrophils which contained several circle-to-oval shaped cytoplasmic NMMMHCA-positive granules. Sequencing analysis of MYH9-RD genes was carried out and revealed a novel missense mutation of c.97T>G (p.W33G) in the patient but not in his parents. INTERVENTION No treatment is necessary. Recognition of MYH9-RD is important to Avoiding unnecessary and potentially harmful treatments. OUTCOMES The patient's condition remained stable during the follow-up. CONCLUSIONS As a result of identifying this missense mutation in this particular case, we have added c.97T>G (p.W33G) to the broad spectrum of potential MYH9 mutations.
Collapse
Affiliation(s)
- Qi Ai
- Department of Hematology and Oncology
| | | | - Jing Yin
- Department of Immunology, Tianjin Children's Hospital, Tianjin, China
| | | | | | | | - Sen Chen
- Department of Hematology and Oncology
| |
Collapse
|
28
|
Zhang Y, Li L, Zhou Q, Li W, Li M, Guo G, Yu B, Kou J. An inhibitor of myosin II, blebbistatin, suppresses development of arterial thrombosis. Biomed Pharmacother 2019; 122:109775. [PMID: 31918291 DOI: 10.1016/j.biopha.2019.109775] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/23/2022] Open
Abstract
Arterial thrombosis (AT) causes various ischemia-related diseases, which impose a serious medical burden worldwide. As an inhibitor of myosin II, blebbistatin has an important role in thrombosis development. We investigated the effect of blebbistatin on carotid artery ligation (CAL)-induced carotid AT and its potential underlying mechanism. A model of carotid AT in mice was generated by CAL. Mice were divided into three groups: CAL model, blebbistatin-treated, and sham-operation. After 7 days, blood vessels were harvested from mice in each group. The procoagulant activity of tissue factor (TF) was tested by a chromogenic assay, and thrombus severity assessed by histopathology scores. Expression of non-muscle myosin heavy chain II A (NMMHCIIA), TF, glycogen synthase kinase 3β (GSK3β), and nuclear factor-kappa B (NF-κB) was detected by immunohistochemical and immunofluorescence staining. mRNA expression was measured by quantitative polymerase chain reaction. Blebbistatin (1 mg/kg) inhibited development of carotid AT, reduced infiltration of inflammatory cells, and prevented vascular-tissue damage, relative to the model group. Furthermore, blebbistatin also reduced the procoagulant activity of TF. Immunohistochemical and immunofluorescence data demonstrated that, compared with the model group, blebbistatin intervention reduced expression of NMMHCIIA, TF, GSK3β, p65, and p-p65 in carotid-artery endothelia in the CAL-induced AT model, but it increased levels of p-GSK3β. Blebbistatin could inhibit expression of NMMHCIIA mRNA in the CAL model. Overall, our data demonstrated that blebbistatin could inhibit TF expression and AT development in arterial endothelia (at least in part) via GSK3β/NF-κB signaling.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Long Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Qianliu Zhou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Wang Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Min Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Gengshuo Guo
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China.
| |
Collapse
|
29
|
Ghalloussi D, Dhenge A, Bergmeier W. New insights into cytoskeletal remodeling during platelet production. J Thromb Haemost 2019; 17:1430-1439. [PMID: 31220402 PMCID: PMC6760864 DOI: 10.1111/jth.14544] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 06/12/2019] [Indexed: 12/16/2022]
Abstract
The past decade has brought unprecedented advances in our understanding of megakaryocyte (MK) biology and platelet production, processes that are strongly dependent on the cytoskeleton. Facilitated by technological innovations, such as new high-resolution imaging techniques (in vitro and in vivo) and lineage-specific gene knockout and reporter mouse strains, we are now able to visualize and characterize the molecular machinery required for MK development and proplatelet formation in live mice. Whole genome and RNA sequencing analysis of patients with rare platelet disorders, combined with targeted genetic interventions in mice, has led to the identification and characterization of numerous new genes important for MK development. Many of the genes important for proplatelet formation code for proteins that control cytoskeletal dynamics in cells, such as Rho GTPases and their downstream targets. In this review, we discuss how the final stages of MK development are controlled by the cellular cytoskeletons, and we compare changes in MK biology observed in patients and mice with mutations in cytoskeleton regulatory genes.
Collapse
Affiliation(s)
- Dorsaf Ghalloussi
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ankita Dhenge
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Wolfgang Bergmeier
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
30
|
Strassel C, Magiera MM, Dupuis A, Batzenschlager M, Hovasse A, Pleines I, Guéguen P, Eckly A, Moog S, Mallo L, Kimmerlin Q, Chappaz S, Strub JM, Kathiresan N, de la Salle H, Van Dorsselaer A, Ferec C, Py JY, Gachet C, Schaeffer-Reiss C, Kile BT, Janke C, Lanza F. An essential role for α4A-tubulin in platelet biogenesis. Life Sci Alliance 2019; 2:2/1/e201900309. [PMID: 30760556 PMCID: PMC6374996 DOI: 10.26508/lsa.201900309] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 01/23/2019] [Accepted: 01/23/2019] [Indexed: 11/24/2022] Open
Abstract
Alpha4A-tubulin is the predominant α-tubulin isotype in platelets. Mutations in α4A-tubulin cause abnormal platelet biogenesis and marginal band formation in mice and in a patient, establishing an essential role of this tubulin isotype. During platelet biogenesis, microtubules (MTs) are arranged into submembranous structures (the marginal band) that encircle the cell in a single plane. This unique MT array has no equivalent in any other mammalian cell, and the mechanisms responsible for this particular mode of assembly are not fully understood. One possibility is that platelet MTs are composed of a particular set of tubulin isotypes that carry specific posttranslational modifications. Although β1-tubulin is known to be essential, no equivalent roles of α-tubulin isotypes in platelet formation or function have so far been reported. Here, we identify α4A-tubulin as a predominant α-tubulin isotype in platelets. Similar to β1-tubulin, α4A-tubulin expression is up-regulated during the late stages of megakaryocyte differentiation. Missense mutations in the α4A-tubulin gene cause macrothrombocytopenia in mice and humans. Defects in α4A-tubulin lead to changes in tubulin tyrosination status of the platelet tubulin pool. Ultrastructural defects include reduced numbers and misarranged MT coils in the platelet marginal band. We further observed defects in megakaryocyte maturation and proplatelet formation in Tuba4a-mutant mice. We have, thus, discovered an α-tubulin isotype with specific and essential roles in platelet biogenesis.
Collapse
Affiliation(s)
- Catherine Strassel
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale, Etablissement Français du Sang Grand Est, Unité Mixte de Recherche-S 1255, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Maria M Magiera
- Institut Curie, Paris-Sciences-et-Lettres Research University, CNRS UMR3348, Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Arnaud Dupuis
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale, Etablissement Français du Sang Grand Est, Unité Mixte de Recherche-S 1255, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Morgane Batzenschlager
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale, Etablissement Français du Sang Grand Est, Unité Mixte de Recherche-S 1255, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Agnès Hovasse
- Laboratoire de Spectrométrie de Masse BioOrganique, Institut Pluridisciplinaire Hubert Curien, CNRS UMR7178, Université de Strasbourg, Strasbourg, France
| | - Irina Pleines
- ACRF Australian Cancer Research Foundation Chemical Biology Division, the Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Paul Guéguen
- Laboratoire de génétique moléculaire et d'histocompatibilité, Centre Hospitalier Régional et Universitaire Morvan, INSERM U1078, EFS Bretagne, Brest, France
| | - Anita Eckly
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale, Etablissement Français du Sang Grand Est, Unité Mixte de Recherche-S 1255, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Sylvie Moog
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale, Etablissement Français du Sang Grand Est, Unité Mixte de Recherche-S 1255, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Léa Mallo
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale, Etablissement Français du Sang Grand Est, Unité Mixte de Recherche-S 1255, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Quentin Kimmerlin
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale, Etablissement Français du Sang Grand Est, Unité Mixte de Recherche-S 1255, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Stéphane Chappaz
- ACRF Australian Cancer Research Foundation Chemical Biology Division, the Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Jean-Marc Strub
- Laboratoire de Spectrométrie de Masse BioOrganique, Institut Pluridisciplinaire Hubert Curien, CNRS UMR7178, Université de Strasbourg, Strasbourg, France
| | - Natarajan Kathiresan
- Institut Curie, Paris-Sciences-et-Lettres Research University, CNRS UMR3348, Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Henri de la Salle
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale, Etablissement Français du Sang Grand Est, Unité Mixte de Recherche-S 1255, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Alain Van Dorsselaer
- Laboratoire de Spectrométrie de Masse BioOrganique, Institut Pluridisciplinaire Hubert Curien, CNRS UMR7178, Université de Strasbourg, Strasbourg, France
| | - Claude Ferec
- Laboratoire de génétique moléculaire et d'histocompatibilité, Centre Hospitalier Régional et Universitaire Morvan, INSERM U1078, EFS Bretagne, Brest, France
| | - Jean-Yves Py
- EFS Centre-Pays de la Loire, site d'Orléans, France
| | - Christian Gachet
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale, Etablissement Français du Sang Grand Est, Unité Mixte de Recherche-S 1255, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Christine Schaeffer-Reiss
- Laboratoire de Spectrométrie de Masse BioOrganique, Institut Pluridisciplinaire Hubert Curien, CNRS UMR7178, Université de Strasbourg, Strasbourg, France
| | - Benjamin T Kile
- ACRF Australian Cancer Research Foundation Chemical Biology Division, the Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Carsten Janke
- Institut Curie, Paris-Sciences-et-Lettres Research University, CNRS UMR3348, Orsay, France .,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - François Lanza
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale, Etablissement Français du Sang Grand Est, Unité Mixte de Recherche-S 1255, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| |
Collapse
|
31
|
Tutwiler V, Peshkova AD, Le Minh G, Zaitsev S, Litvinov RI, Cines DB, Weisel JW. Blood clot contraction differentially modulates internal and external fibrinolysis. J Thromb Haemost 2019; 17:361-370. [PMID: 30582674 DOI: 10.1111/jth.14370] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Indexed: 01/16/2023]
Abstract
Essentials Clot contraction influences the rate of fibrinolysis in vitro. Internal fibrinolysis is enhanced ∼2-fold in contracted vs. uncontracted blood clots. External fibrinolysis is ∼4-fold slower in contracted vs. uncontracted blood clots. Contraction can modulate lytic resistance and potentially the clinical outcome of thrombosis. SUMMARY: Background Fibrinolysis involves dissolution of polymeric fibrin networks that is required to restore blood flow through vessels obstructed by thrombi. The efficiency of lysis depends in part on the susceptibility of fibrin to enzymatic digestion, which is governed by the structure and spatial organization of fibrin fibers. How platelet-driven clot contraction affects the efficacy of fibrinolysis has received relatively little study. Objective Here, we examined the effects of clot contraction on the rate of internal fibrinolysis emanating from within the clot to simulate (patho)physiological conditions and external fibrinolysis initiated from the clot exterior to simulate therapeutic thrombolysis. Methods Clot contraction was prevented by inhibiting platelet myosin IIa activity, actin polymerization or platelet-fibrin(ogen) binding. Internal fibrinolysis was measured by optical tracking of clot size. External fibrinolysis was determined by the release of radioactive fibrin degradation products. Results and Conclusions Clot contraction enhanced the rate of internal fibrinolysis ∼2-fold. In contrast, external fibrinolysis was ~4-fold slower in contracted clots. This dichotomy in the susceptibility of contracted and uncontracted clots to internal vs. external lysis suggests that the rate of lysis is dependent upon the interplay between accessibility of fibrin fibers to fibrinolytic agents, including clot permeability, and the spatial proximity of the fibrin fibers that modulate the effects of the fibrinolytic enzymes. Understanding how compaction of blood clots influences clot lysis might have important implications for prevention and treatment of thrombotic disorders.
Collapse
Affiliation(s)
- Valerie Tutwiler
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Alina D Peshkova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Giang Le Minh
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Sergei Zaitsev
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Douglas B Cines
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
32
|
|
33
|
Variants in exons 5 and 6 of ACTB cause syndromic thrombocytopenia. Nat Commun 2018; 9:4250. [PMID: 30315159 PMCID: PMC6185941 DOI: 10.1038/s41467-018-06713-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 09/20/2018] [Indexed: 01/08/2023] Open
Abstract
Germline mutations in the ubiquitously expressed ACTB, which encodes β-cytoplasmic actin (CYA), are almost exclusively associated with Baraitser-Winter Cerebrofrontofacial syndrome (BWCFF). Here, we report six patients with previously undescribed heterozygous variants clustered in the 3′-coding region of ACTB. Patients present with clinical features distinct from BWCFF, including mild developmental disability, microcephaly, and thrombocytopenia with platelet anisotropy. Using patient-derived fibroblasts, we demonstrate cohort specific changes to β-CYA filament populations, which include the enhanced recruitment of thrombocytopenia-associated actin binding proteins (ABPs). These perturbed interactions are supported by in silico modeling and are validated in disease-relevant thrombocytes. Co-examination of actin and microtubule cytoskeleton constituents in patient-derived megakaryocytes and thrombocytes indicates that these β-CYA mutations inhibit the final stages of platelet maturation by compromising microtubule organization. Our results define an ACTB-associated clinical syndrome with a distinct genotype-phenotype correlation and delineate molecular mechanisms underlying thrombocytopenia in this patient cohort. Genetic variants in ACTB and ACTG1 have been associated with Baraitser-Winter Cerebrofrontofacial syndrome. Here, the authors report of a syndromic thrombocytopenia caused by variants in ACTB exons 5 or 6 that compromise the organization and coupling of the cytoskeleton, leading to impaired platelet maturation.
Collapse
|
34
|
Platelet communication with the vascular wall: role of platelet-derived microparticles and non-coding RNAs. Clin Sci (Lond) 2018; 132:1875-1888. [PMID: 30185611 DOI: 10.1042/cs20180580] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 12/24/2022]
Abstract
Platelets play an important role in vascular homeostasis through their interaction with circulating blood cells as well as the vascular wall. Platelet-mediated communication with other cells can take the form of direct cell-cell interactions via membrane receptors or indirectly through the release of different soluble factors stored in their granules as well as through the release of microparticles. The latter carry different proteins and RNAs which are transferred to the target cells. The aim of this review is to discuss the role of platelet-derived factors, adhesion molecules as well as RNAs as mediators of the cross-talk between platelets and the vessel wall.
Collapse
|
35
|
Pleines I, Cherpokova D, Bender M. Rho GTPases and their downstream effectors in megakaryocyte biology. Platelets 2018; 30:9-16. [DOI: 10.1080/09537104.2018.1478071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Irina Pleines
- Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Deya Cherpokova
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Markus Bender
- Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| |
Collapse
|
36
|
Balduini A, Raslova H, Di Buduo CA, Donada A, Ballmaier M, Germeshausen M, Balduini CL. Clinic, pathogenic mechanisms and drug testing of two inherited thrombocytopenias, ANKRD26-related Thrombocytopenia and MYH9-related diseases. Eur J Med Genet 2018; 61:715-722. [PMID: 29545013 DOI: 10.1016/j.ejmg.2018.01.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 01/08/2018] [Accepted: 01/27/2018] [Indexed: 12/21/2022]
Abstract
Inherited thrombocytopenias (ITs) are a heterogeneous group of disorders characterized by low platelet count resulting in impaired hemostasis. Patients can have spontaneous hemorrhages and/or excessive bleedings provoked by hemostatic challenges as trauma or surgery. To date, ITs encompass 32 different rare monogenic disorders caused by mutations of 30 genes. This review will focus on the major discoveries that have been made in the last years on the diagnosis, treatment and molecular mechanisms of ANKRD26-Related Thrombocytopenia and MYH9-Related Diseases. Furthermore, we will discuss the use a Thrombopoietin mimetic as a novel approach to treat the thrombocytopenia in these patients. We will propose the use of a new 3D bone marrow model to study the mechanisms of action of these drugs and to test their efficacy and safety in patients. The overall purpose of this review is to point out that important progresses have been made in understanding the pathogenesis of ANKRD26-Related Thrombocytopenia and MYH9-Related Diseases and new therapeutic approaches have been proposed and tested. Future advancement in this research will rely in the development of more physiological models to study the regulation of human platelet biogenesis, disease mechanisms and specific pharmacologic targets.
Collapse
Affiliation(s)
- Alessandra Balduini
- University of Pavia, Pavia, Italy; IRCCS Policlinico San Matteo Foundation, Pavia, Italy.
| | - Hana Raslova
- INSERM UMR 1170, Gustave Roussy Cancer Campus, Université Paris-Saclay, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Villejuif, France
| | - Christian A Di Buduo
- University of Pavia, Pavia, Italy; IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Alessandro Donada
- INSERM UMR 1170, Gustave Roussy Cancer Campus, Université Paris-Saclay, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Villejuif, France
| | | | | | - Carlo L Balduini
- University of Pavia, Pavia, Italy; IRCCS Policlinico San Matteo Foundation, Pavia, Italy.
| |
Collapse
|
37
|
Italiano JE, Hartwig JH. Megakaryocyte and Platelet Structure. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00124-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
38
|
Aguilar A, Boscher J, Pertuy F, Gachet C, Léon C. Three-Dimensional Culture in a Methylcellulose-Based Hydrogel to Study the Impact of Stiffness on Megakaryocyte Differentiation. Methods Mol Biol 2018; 1812:139-153. [PMID: 30171577 DOI: 10.1007/978-1-4939-8585-2_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The differentiation and maturation of megakaryocytes (MKs) occurs in a 3D environment where the cells must constantly adapt to the external physical and mechanical constraints during their development and migration to sinusoid vessels. In this chapter, we present a method for culture of mouse MKs from bone marrow hematopoietic progenitor cells in a methylcellulose 3D medium with a stiffness mimicking that of bone marrow. In addition, we describe how the MKs can be recovered to allow for analysis of their differentiation and maturation state by transmission electron microscopy, immunofluorescence or flow cytometry techniques and to evaluate their ability to form proplatelets. This approach allows (1) generation of MKs with a morphology that more closely resembles the MKs that differentiate in vivo, (2) recovery of megakaryocyte phenotypes sometimes observed in vivo but not found in classical liquid cultures, and (3) study of mechanotransduction pathways induced by the stiffness of the medium.
Collapse
Affiliation(s)
- Alicia Aguilar
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, F-67000, Strasbourg, France
| | - Julie Boscher
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, F-67000, Strasbourg, France
| | - Fabien Pertuy
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, F-67000, Strasbourg, France
| | - Christian Gachet
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, F-67000, Strasbourg, France
| | - Catherine Léon
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, F-67000, Strasbourg, France.
| |
Collapse
|
39
|
Bozzi V, Panza E, Barozzi S, Gruppi C, Seri M, Balduini C, Pecci A. Mutations responsible for MYH9-related thrombocytopenia impair SDF-1-driven migration of megakaryoblastic cells. Thromb Haemost 2017; 106:693-704. [DOI: 10.1160/th11-02-0126] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 07/11/2011] [Indexed: 01/01/2023]
Abstract
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.
Collapse
|
40
|
Léon C, Dupuis A, Gachet C, Lanza F. The contribution of mouse models to the understanding of constitutional thrombocytopenia. Haematologica 2017; 101:896-908. [PMID: 27478199 DOI: 10.3324/haematol.2015.139394] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/04/2016] [Indexed: 11/09/2022] Open
Abstract
Constitutional thrombocytopenias result from platelet production abnormalities of hereditary origin. Long misdiagnosed and poorly studied, knowledge about these rare diseases has increased considerably over the last twenty years due to improved technology for the identification of mutations, as well as an improvement in obtaining megakaryocyte culture from patient hematopoietic stem cells. Simultaneously, the manipulation of mouse genes (transgenesis, total or conditional inactivation, introduction of point mutations, random chemical mutagenesis) have helped to generate disease models that have contributed greatly to deciphering patient clinical and laboratory features. Most of the thrombocytopenias for which the mutated genes have been identified now have a murine model counterpart. This review focuses on the contribution that these mouse models have brought to the understanding of hereditary thrombocytopenias with respect to what was known in humans. Animal models have either i) provided novel information on the molecular and cellular pathways that were missing from the patient studies; ii) improved our understanding of the mechanisms of thrombocytopoiesis; iii) been instrumental in structure-function studies of the mutated gene products; and iv) been an invaluable tool as preclinical models to test new drugs or develop gene therapies. At present, the genetic determinants of thrombocytopenia remain unknown in almost half of all cases. Currently available high-speed sequencing techniques will identify new candidate genes, which will in turn allow the generation of murine models to confirm and further study the abnormal phenotype. In a complementary manner, programs of random mutagenesis in mice should also identify new candidate genes involved in thrombocytopenia.
Collapse
Affiliation(s)
- Catherine Léon
- UMR_S949, INSERM, Strasbourg, France Etablissement Français du Sang-Alsace (EFS-Alsace), Strasbourg, France Université de Strasbourg, France Fédération de Médecine Translationnelle de Strasbourg (FMTS), France
| | - Arnaud Dupuis
- UMR_S949, INSERM, Strasbourg, France Etablissement Français du Sang-Alsace (EFS-Alsace), Strasbourg, France Université de Strasbourg, France Fédération de Médecine Translationnelle de Strasbourg (FMTS), France
| | - Christian Gachet
- UMR_S949, INSERM, Strasbourg, France Etablissement Français du Sang-Alsace (EFS-Alsace), Strasbourg, France Université de Strasbourg, France Fédération de Médecine Translationnelle de Strasbourg (FMTS), France
| | - François Lanza
- UMR_S949, INSERM, Strasbourg, France Etablissement Français du Sang-Alsace (EFS-Alsace), Strasbourg, France Université de Strasbourg, France Fédération de Médecine Translationnelle de Strasbourg (FMTS), France
| |
Collapse
|
41
|
Dütting S, Gaits-Iacovoni F, Stegner D, Popp M, Antkowiak A, van Eeuwijk JMM, Nurden P, Stritt S, Heib T, Aurbach K, Angay O, Cherpokova D, Heinz N, Baig AA, Gorelashvili MG, Gerner F, Heinze KG, Ware J, Krohne G, Ruggeri ZM, Nurden AT, Schulze H, Modlich U, Pleines I, Brakebusch C, Nieswandt B. A Cdc42/RhoA regulatory circuit downstream of glycoprotein Ib guides transendothelial platelet biogenesis. Nat Commun 2017. [PMID: 28643773 PMCID: PMC5481742 DOI: 10.1038/ncomms15838] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Blood platelets are produced by large bone marrow (BM) precursor cells, megakaryocytes (MKs), which extend cytoplasmic protrusions (proplatelets) into BM sinusoids. The molecular cues that control MK polarization towards sinusoids and limit transendothelial crossing to proplatelets remain unknown. Here, we show that the small GTPases Cdc42 and RhoA act as a regulatory circuit downstream of the MK-specific mechanoreceptor GPIb to coordinate polarized transendothelial platelet biogenesis. Functional deficiency of either GPIb or Cdc42 impairs transendothelial proplatelet formation. In the absence of RhoA, increased Cdc42 activity and MK hyperpolarization triggers GPIb-dependent transmigration of entire MKs into BM sinusoids. These findings position Cdc42 (go-signal) and RhoA (stop-signal) at the centre of a molecular checkpoint downstream of GPIb that controls transendothelial platelet biogenesis. Our results may open new avenues for the treatment of platelet production disorders and help to explain the thrombocytopenia in patients with Bernard-Soulier syndrome, a bleeding disorder caused by defects in GPIb-IX-V.
Collapse
Affiliation(s)
- Sebastian Dütting
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.,Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Frederique Gaits-Iacovoni
- INSERM UMR1048, Institut des Maladies Métaboliques et Cardiovasculaires-I2MC, UMR1048, Institut National de la Santé et de la Recherche Médicale, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France
| | - David Stegner
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Michael Popp
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.,Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Adrien Antkowiak
- INSERM UMR1048, Institut des Maladies Métaboliques et Cardiovasculaires-I2MC, UMR1048, Institut National de la Santé et de la Recherche Médicale, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France
| | - Judith M M van Eeuwijk
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.,Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Paquita Nurden
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.,Institut Hospitalo-Universitaire LIRYC, Plateforme Technologique d'Innovation Biomédicale, Hôpital Xavier Arnozan, Avenue du Haut Lévêque, 33604 Pessac, France
| | - Simon Stritt
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.,Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Tobias Heib
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.,Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Katja Aurbach
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.,Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Oguzhan Angay
- Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Deya Cherpokova
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.,Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Niels Heinz
- Research Group for Gene Modification in Stem Cells, LOEWE Center for Cell and Gene Therapy Frankfurt/Main and the Paul-Ehrlich-Institute, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany
| | - Ayesha A Baig
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.,Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Maximilian G Gorelashvili
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.,Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Frank Gerner
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.,Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Jerry Ware
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, Arkansass 72205, USA
| | - Georg Krohne
- Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Zaverio M Ruggeri
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, California 92037, USA
| | - Alan T Nurden
- Institut Hospitalo-Universitaire LIRYC, Plateforme Technologique d'Innovation Biomédicale, Hôpital Xavier Arnozan, Avenue du Haut Lévêque, 33604 Pessac, France
| | - Harald Schulze
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Ute Modlich
- Research Group for Gene Modification in Stem Cells, LOEWE Center for Cell and Gene Therapy Frankfurt/Main and the Paul-Ehrlich-Institute, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany
| | - Irina Pleines
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.,Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Cord Brakebusch
- BRIC, Biomedical Institute, University of Copenhagen, Nørregade 10, 1165 Copenhagen, Denmark
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.,Rudolf Virchow Center, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| |
Collapse
|
42
|
Zhang Y, Li L, Zhao Y, Han H, Hu Y, Liang D, Yu B, Kou J. The Myosin II Inhibitor, Blebbistatin, Ameliorates FeCl3-induced Arterial Thrombosis via the GSK3β-NF-κB Pathway. Int J Biol Sci 2017; 13:630-639. [PMID: 28539835 PMCID: PMC5441179 DOI: 10.7150/ijbs.18485] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 03/07/2017] [Indexed: 12/13/2022] Open
Abstract
Arterial thrombosis and its related diseases are major healthcare problems worldwide. Blebbistatin is an inhibitor of myosin II, which plays an important role in thrombosis. The aim of our study is to explore the effect and potential mechanism of blebbistatin on arterial thrombosis. A ferric chloride (FeCl3) solution at a concentration of 5% was used to induce carotid artery thrombosis in mice. Immunohistochemistry and immunofluorescence staining were used to detect the expression or activation of non-muscle myosin heavy chain IIA (NMMHC IIA), tissue factor (TF), GSK3β and NF-κB. Blebbistatin (1 mg/kg, i.p.) significantly reduced carotid artery thrombosis induced by FeCl3 solution in mice, inhibited NMMHC IIA expression and impeded TF expression via the GSK3β-NF-κB signalling pathway in mouse arterial vascular tissues. The present study demonstrates that blebbistatin may impede TF expression partly via the Akt/GSK3β-NF-κB signalling pathways in the endothelium in a FeCl3 model, shedding new insights into the pathogenesis of arterial thrombosis and providing new clues for the development of antithrombotic drugs.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Long Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Yazheng Zhao
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Han Han
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Yang Hu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Di Liang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| |
Collapse
|
43
|
Palazzo A, Bluteau O, Messaoudi K, Marangoni F, Chang Y, Souquere S, Pierron G, Lapierre V, Zheng Y, Vainchenker W, Raslova H, Debili N. The cell division control protein 42-Src family kinase-neural Wiskott-Aldrich syndrome protein pathway regulates human proplatelet formation. J Thromb Haemost 2016; 14:2524-2535. [PMID: 27685868 DOI: 10.1111/jth.13519] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Indexed: 12/31/2022]
Abstract
Essentials The role of the cytoskeleton during megakaryocyte differentiation was examined. Human megakaryocytes are derived from in vitro cultured CD34+ cells. Cell division control protein 42 (CDC42) positively regulates proplatelet formation (PPF). Neural Wiskott-Aldrich syndrome protein, the main effector of CDC42 with Src positively regulates PPF. SUMMARY Background Cytoskeletal rearrangements are essential for platelet release. The RHO small GTPase family, as regulators of the actin cytoskeleton, play an important role in proplatelet formation (PPF). In the neuronal system, CDC42 is involved in axon formation, a process that combines elongation and branching as for PPF. Objective To analyze the role of CDC42 and its effectors of the Wiskott-Aldrich syndrome protein (WASP) family in PPF. Methods Human megakaryocytes (MKs) were obtained from CD34+ cells. Inhibition of CDC42 in MKs was performed with the chemical inhibitor CASIN or with an active or a dominant-negative form of CDC42. The knock-down of N-WASP was obtained with a small hairpin RNA strategy Results Herein, we show that CDC42 activity increased during MK differentiation. The use of the chemical inhibitor CASIN or of an active or a dominant-negative form of CDC42 demonstrated that CDC42 positively regulated PPF in vitro. We determined that N-WASP, but not WASP, regulated PPF. We found that N-WASP knockdown led to a marked decrease in PPF, owing to a defect in the demarcation membrane system (DMS). This was associated with RHOA activation, and a concomitant augmentation in the phosphorylation of mysosin light chain 2. Phosphorylation of N-WASP, creating a primed form of N-WASP, increased during MK differentiation. Phosphorylation inhibition by two Src family kinase inhibitors decreased PPF. Conclusions We conclude that N-WASP positively regulates DMS development and PPF, and that the Src family kinases in association with CDC42 regulate PPF through N-WASP.
Collapse
Affiliation(s)
- A Palazzo
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée Ligue Contre le Cancer, Laboratoire d'Excellence GR-Ex, Villejuif, France
- Université Paris-Saclay, UMR 1170, Villejuif, France
- Gustave Roussy, UMR 1170, Villejuif, France
| | - O Bluteau
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée Ligue Contre le Cancer, Laboratoire d'Excellence GR-Ex, Villejuif, France
- Université Paris-Saclay, UMR 1170, Villejuif, France
- Gustave Roussy, UMR 1170, Villejuif, France
| | - K Messaoudi
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée Ligue Contre le Cancer, Laboratoire d'Excellence GR-Ex, Villejuif, France
- Université Paris-Saclay, UMR 1170, Villejuif, France
- Gustave Roussy, UMR 1170, Villejuif, France
| | - F Marangoni
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée Ligue Contre le Cancer, Laboratoire d'Excellence GR-Ex, Villejuif, France
- Université Paris-Saclay, UMR 1170, Villejuif, France
- Gustave Roussy, UMR 1170, Villejuif, France
| | - Y Chang
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée Ligue Contre le Cancer, Laboratoire d'Excellence GR-Ex, Villejuif, France
- Université Paris-Saclay, UMR 1170, Villejuif, France
- Gustave Roussy, UMR 1170, Villejuif, France
| | - S Souquere
- Gustave Roussy, Centre Nationale de la Recherche Scientifique, UMR 8122, Gustave Roussy, Villejuif, France
| | - G Pierron
- Gustave Roussy, Centre Nationale de la Recherche Scientifique, UMR 8122, Gustave Roussy, Villejuif, France
| | - V Lapierre
- Gustave Roussy, Unité de Thérapie Cellulaire, Villejuif, France
| | - Y Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - W Vainchenker
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée Ligue Contre le Cancer, Laboratoire d'Excellence GR-Ex, Villejuif, France
- Université Paris-Saclay, UMR 1170, Villejuif, France
- Gustave Roussy, UMR 1170, Villejuif, France
| | - H Raslova
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée Ligue Contre le Cancer, Laboratoire d'Excellence GR-Ex, Villejuif, France
- Université Paris-Saclay, UMR 1170, Villejuif, France
- Gustave Roussy, UMR 1170, Villejuif, France
| | - N Debili
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée Ligue Contre le Cancer, Laboratoire d'Excellence GR-Ex, Villejuif, France
- Université Paris-Saclay, UMR 1170, Villejuif, France
- Gustave Roussy, UMR 1170, Villejuif, France
| |
Collapse
|
44
|
Fan X, Yang H, Kumar S, Tumelty KE, Pisarek-Horowitz A, Rasouly HM, Sharma R, Chan S, Tyminski E, Shamashkin M, Belghasem M, Henderson JM, Coyle AJ, Salant DJ, Berasi SP, Lu W. SLIT2/ROBO2 signaling pathway inhibits nonmuscle myosin IIA activity and destabilizes kidney podocyte adhesion. JCI Insight 2016; 1:e86934. [PMID: 27882344 DOI: 10.1172/jci.insight.86934] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The repulsive guidance cue SLIT2 and its receptor ROBO2 are required for kidney development and podocyte foot process structure, but the SLIT2/ROBO2 signaling mechanism regulating podocyte function is not known. Here we report that a potentially novel signaling pathway consisting of SLIT/ROBO Rho GTPase activating protein 1 (SRGAP1) and nonmuscle myosin IIA (NMIIA) regulates podocyte adhesion downstream of ROBO2. We found that the myosin II regulatory light chain (MRLC), a subunit of NMIIA, interacts directly with SRGAP1 and forms a complex with ROBO2/SRGAP1/NMIIA in the presence of SLIT2. Immunostaining demonstrated that SRGAP1 is a podocyte protein and is colocalized with ROBO2 on the basal surface of podocytes. In addition, SLIT2 stimulation inhibits NMIIA activity, decreases focal adhesion formation, and reduces podocyte attachment to collagen. In vivo studies further showed that podocyte-specific knockout of Robo2 protects mice from hypertension-induced podocyte detachment and albuminuria and also partially rescues the podocyte-loss phenotype in Myh9 knockout mice. Thus, we have identified SLIT2/ROBO2/SRGAP1/NMIIA as a potentially novel signaling pathway in kidney podocytes, which may play a role in regulating podocyte adhesion and attachment. Our findings also suggest that SLIT2/ROBO2 signaling might be a therapeutic target for kidney diseases associated with podocyte detachment and loss.
Collapse
Affiliation(s)
- Xueping Fan
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Hongying Yang
- Centers for Therapeutic Innovation, Pfizer Inc., Boston, Massachusetts, USA
| | - Sudhir Kumar
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Kathleen E Tumelty
- Centers for Therapeutic Innovation, Pfizer Inc., Boston, Massachusetts, USA
| | - Anna Pisarek-Horowitz
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Hila Milo Rasouly
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Richa Sharma
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Stefanie Chan
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Edyta Tyminski
- Centers for Therapeutic Innovation, Pfizer Inc., Boston, Massachusetts, USA
| | - Michael Shamashkin
- Centers for Therapeutic Innovation, Pfizer Inc., Boston, Massachusetts, USA
| | - Mostafa Belghasem
- Department of Pathology and Laboratory Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Joel M Henderson
- Department of Pathology and Laboratory Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Anthony J Coyle
- Centers for Therapeutic Innovation, Pfizer Inc., Boston, Massachusetts, USA
| | - David J Salant
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Stephen P Berasi
- Centers for Therapeutic Innovation, Pfizer Inc., Boston, Massachusetts, USA
| | - Weining Lu
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
45
|
Importance of environmental stiffness for megakaryocyte differentiation and proplatelet formation. Blood 2016; 128:2022-2032. [PMID: 27503502 DOI: 10.1182/blood-2016-02-699959] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 07/20/2016] [Indexed: 12/31/2022] Open
Abstract
Megakaryocyte (MK) differentiation occurs within the bone marrow (BM), a complex 3-dimensional (3D) environment of low stiffness exerting local external constraints. To evaluate the influence of the 3D mechanical constraints that MKs may encounter in vivo, we differentiated mouse BM progenitors in methylcellulose (MC) hydrogels tuned to mimic BM stiffness. We found that MKs grown in a medium of 30- to 60-Pa stiffness more closely resembled those in the BM in terms of demarcation membrane system (DMS) morphological aspect and exhibited higher ploidy levels, as compared with MKs in liquid culture. Following resuspension in a liquid medium, MC-grown MKs displayed twice as much proplatelet formation as cells grown in liquid culture. Thus, the MC gel, by mimicking external constraints, appeared to positively influence MK differentiation. To determine whether MKs adapt to extracellular stiffness through mechanotransduction involving actomyosin-based modulation of the intracellular tension, myosin-deficient (Myh9-/-) progenitors were grown in MC gels. Absence of myosin resulted in abnormal cell deformation and strongly decreased proplatelet formation, similarly to features observed for Myh9-/- MKs differentiated in situ but not in vitro. Moreover, megakaryoblastic leukemia 1 (MKL1), a well-known actor in mechanotransduction, was found to be preferentially relocated within the nucleus of MC-differentiated MKs, whereas its inhibition prevented MC-mediated increased proplatelet formation. Altogether, these data show that a 3D medium mimicking BM stiffness contributes, through the myosin IIA and MKL1 pathways, to a more favorable in vitro environment for MK differentiation, which ultimately translates into increased proplatelet production.
Collapse
|
46
|
Antkowiak A, Viaud J, Severin S, Zanoun M, Ceccato L, Chicanne G, Strassel C, Eckly A, Leon C, Gachet C, Payrastre B, Gaits-Iacovoni F. Cdc42-dependent F-actin dynamics drive structuration of the demarcation membrane system in megakaryocytes. J Thromb Haemost 2016; 14:1268-84. [PMID: 26991240 DOI: 10.1111/jth.13318] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 03/03/2016] [Indexed: 02/04/2023]
Abstract
UNLABELLED Essentials Information about the formation of the demarcation membrane system (DMS) is still lacking. We investigated the role of the cytoskeleton in DMS structuration in megakaryocytes. Cdc42/Pak-dependent F-actin remodeling regulates DMS organization for proper megakaryopoiesis. These data highlight the mandatory role of F-actin in platelet biogenesis. SUMMARY Background Blood platelet biogenesis results from the maturation of megakaryocytes (MKs), which involves the development of a complex demarcation membrane system (DMS). Therefore, MK differentiation is an attractive model for studying membrane remodeling. Objectives We sought to investigate the mechanism of DMS structuration in relationship to the cytoskeleton. Results Using three-dimensional (3D) confocal imaging, we have identified consecutive stages of DMS organization that rely on F-actin dynamics to polarize membranes and nuclei territories. Interestingly, microtubules are not involved in this process. We found that the mechanism underlying F-actin-dependent DMS formation required the activation of the guanosine triphosphate hydrolase Cdc42 and its p21-activated kinase effectors (Pak1/2/3). Förster resonance energy transfer demonstrated that active Cdc42 was associated with endomembrane dynamics throughout terminal maturation. Inhibition of Cdc42 or Pak1/2/3 severely destructured the DMS and blocked proplatelet formation. Even though this process does not require containment within the hematopoietic niche, because DMS structuration was observed upon thrombopoietin-treatment in suspension, integrin outside-in signaling was required for Pak activation and probably resulted from secretion of extracellular matrix by MKs. Conclusions These data indicate a functional link, mandatory for MK differentiation, between actin dynamics, regulated by Cdc42/Pak1/2/3, and DMS maturation.
Collapse
Affiliation(s)
- A Antkowiak
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - J Viaud
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - S Severin
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - M Zanoun
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - L Ceccato
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - G Chicanne
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - C Strassel
- INSERM, UMR_S949, Université de Strasbourg, Etablissement Français du Sang-Alsace, Toulouse, France
| | - A Eckly
- INSERM, UMR_S949, Université de Strasbourg, Etablissement Français du Sang-Alsace, Toulouse, France
| | - C Leon
- INSERM, UMR_S949, Université de Strasbourg, Etablissement Français du Sang-Alsace, Toulouse, France
| | - C Gachet
- INSERM, UMR_S949, Université de Strasbourg, Etablissement Français du Sang-Alsace, Toulouse, France
| | - B Payrastre
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
- Laboratoire d'Hématologie, CHU de Toulouse, Toulouse, France
| | - F Gaits-Iacovoni
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| |
Collapse
|
47
|
Litvinov RI, Weisel JW. What Is the Biological and Clinical Relevance of Fibrin? Semin Thromb Hemost 2016; 42:333-43. [PMID: 27056152 DOI: 10.1055/s-0036-1571342] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
As our knowledge of the structure and functions of fibrinogen and fibrin has increased tremendously, several key findings have given some people a superficial impression that the biological and clinical significance of these clotting proteins may be less than earlier thought. Most strikingly, studies of fibrinogen knockout mice demonstrated that many of these mice survive to weaning and beyond, suggesting that fibrin(ogen) may not be entirely necessary. Humans with afibrinogenemia also survive. Furthermore, in recent years, the major emphasis in the treatment of arterial thrombosis has been on inhibition of platelets, rather than fibrin. In contrast to the initially apparent conclusions from these results, it has become increasingly clear that fibrin is essential for hemostasis; is a key factor in thrombosis; and plays an important biological role in infection, inflammation, immunology, and wound healing. In addition, fibrinogen replacement therapy has become a preferred, major treatment for severe bleeding in trauma and surgery. Finally, fibrin is a unique biomaterial and is used as a sealant or glue, a matrix for cells, a scaffold for tissue engineering, and a carrier and/or a vector for targeted drug delivery.
Collapse
Affiliation(s)
- Rustem I Litvinov
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John W Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
48
|
Newell-Litwa KA, Horwitz R, Lamers ML. Non-muscle myosin II in disease: mechanisms and therapeutic opportunities. Dis Model Mech 2015; 8:1495-515. [PMID: 26542704 PMCID: PMC4728321 DOI: 10.1242/dmm.022103] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The actin motor protein non-muscle myosin II (NMII) acts as a master regulator of cell morphology, with a role in several essential cellular processes, including cell migration and post-synaptic dendritic spine plasticity in neurons. NMII also generates forces that alter biochemical signaling, by driving changes in interactions between actin-associated proteins that can ultimately regulate gene transcription. In addition to its roles in normal cellular physiology, NMII has recently emerged as a critical regulator of diverse, genetically complex diseases, including neuronal disorders, cancers and vascular disease. In the context of these disorders, NMII regulatory pathways can be directly mutated or indirectly altered by disease-causing mutations. NMII regulatory pathway genes are also increasingly found in disease-associated copy-number variants, particularly in neuronal disorders such as autism and schizophrenia. Furthermore, manipulation of NMII-mediated contractility regulates stem cell pluripotency and differentiation, thus highlighting the key role of NMII-based pharmaceuticals in the clinical success of stem cell therapies. In this Review, we discuss the emerging role of NMII activity and its regulation by kinases and microRNAs in the pathogenesis and prognosis of a diverse range of diseases, including neuronal disorders, cancer and vascular disease. We also address promising clinical applications and limitations of NMII-based inhibitors in the treatment of these diseases and the development of stem-cell-based therapies.
Collapse
Affiliation(s)
- Karen A Newell-Litwa
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Rick Horwitz
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Marcelo L Lamers
- Department of Morphological Sciences, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-010, Brazil
| |
Collapse
|
49
|
Regulation of actin polymerization by tropomodulin-3 controls megakaryocyte actin organization and platelet biogenesis. Blood 2015; 126:520-30. [PMID: 25964668 DOI: 10.1182/blood-2014-09-601484] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 05/04/2015] [Indexed: 01/02/2023] Open
Abstract
The actin cytoskeleton is important for platelet biogenesis. Tropomodulin-3 (Tmod3), the only Tmod isoform detected in platelets and megakaryocytes (MKs), caps actin filament (F-actin) pointed ends and binds tropomyosins (TMs), regulating actin polymerization and stability. To determine the function of Tmod3 in platelet biogenesis, we studied Tmod3(-/-) embryos, which are embryonic lethal by E18.5. Tmod3(-/-) embryos often show hemorrhaging at E14.5 with fewer and larger platelets, indicating impaired platelet biogenesis. MK numbers are moderately increased in Tmod3(-/-) fetal livers, with only a slight increase in the 8N population, suggesting that MK differentiation is not significantly affected. However, Tmod3(-/-) MKs fail to develop a normal demarcation membrane system (DMS), and cytoplasmic organelle distribution is abnormal. Moreover, cultured Tmod3(-/-) MKs exhibit impaired proplatelet formation with a wide range of proplatelet bud sizes, including abnormally large proplatelet buds containing incorrect numbers of von Willebrand factor-positive granules. Tmod3(-/-) MKs exhibit F-actin disturbances, and Tmod3(-/-) MKs spreading on collagen fail to polymerize F-actin into actomyosin contractile bundles. Tmod3 associates with TM4 and the F-actin cytoskeleton in wild-type MKs, and confocal microscopy reveals that Tmod3, TM4, and F-actin partially colocalize near the membrane of proplatelet buds. In contrast, the abnormally large proplatelets from Tmod3(-/-) MKs show increased F-actin and redistribution of F-actin and TM4 from the cortex to the cytoplasm, but normal microtubule coil organization. We conclude that F-actin capping by Tmod3 regulates F-actin organization in mouse fetal liver-derived MKs, thereby controlling MK cytoplasmic morphogenesis, including DMS formation and organelle distribution, as well as proplatelet formation and sizing.
Collapse
|
50
|
Badirou I, Pan J, Souquere S, Legrand C, Pierron G, Wang A, Eckly A, Roy A, Gachet C, Vainchenker W, Chang Y, Léon C. Distinct localizations and roles of non-muscle myosin II during proplatelet formation and platelet release. J Thromb Haemost 2015; 13:851-9. [PMID: 25736522 DOI: 10.1111/jth.12887] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/24/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND At the end of maturation, megakaryocytes (MKs) form long cytoplasmic extensions called proplatelets (PPT). Enormous changes in cytoskeletal structures cause PPT to extend further, to re-localize organelles such as mitochondria and to fragment, leading to platelet release. Two non-muscle myosin IIs (NMIIs) are expressed in MKs; however, only NMII-A (MYH9), but not NMII-B (MYH10), is expressed in mature MKs and is implicated in PPT formation. OBJECTIVES To provide in vivo evidence on the specific role of NMII-A and IIB in MK PPT formation. METHODS We studied two transgenic mouse models in which non-muscle myosin heavy chain (NMHC) II-A was genetically replaced either by II-B or by a chimeric NMHCII that combined the head domain of II-A with the rod and tail domains of II-B. RESULTS AND CONCLUSIONS This work demonstrates that the kinetic properties of NM-IIA, depending on the N-terminal domain, render NMII-A the better NMII candidate to control PPT formation. Furthermore, the carboxyl-terminal domain determines myosin II localization in the constriction region of PPT and is responsible for the specific role of NMII in platelet release.
Collapse
Affiliation(s)
- I Badirou
- Institut National de la Santé et de la Recherche Médicale, Villejuif, France; Université Paris-Sud, Le Kremlin-Bicêtre, France; Institut Gustave Roussy, Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|