1
|
Italiano JE, Payne C, Bekendam RH. Looking Under the Hood at the Cytoskeletal Engine of Platelet Production. Arterioscler Thromb Vasc Biol 2025; 45:186-197. [PMID: 39665140 DOI: 10.1161/atvbaha.124.320392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Blood platelets are anucleate cells essential for normal blood hemostasis. To maintain a normal platelet count of 150 000 to 400 000 per μL of blood, 1011 platelets must be released each day from precursor cells called megakaryocytes. In this review, we aim to provide an overview of platelet production and evaluate the proposed mechanisms of platelet generation. We will discuss novel cytoskeletal mechanisms of platelet production, including microtubule and actin-based systems. We present new evidence that supports a cytoplasmic trigger for platelet production, discuss centrosome clustering as a new mechanism to trigger proplatelet production, and review new data supporting the bone marrow as the major location of platelet production.
Collapse
Affiliation(s)
- Joseph E Italiano
- Vascular Biology Program, Boston Children's Hospital, MA (J.E.I., C.P., R.H.B.)
- Department of Surgery, Harvard Medical School, Boston, MA (J.E.I., C.P., R.H.B.)
| | - Clementine Payne
- Vascular Biology Program, Boston Children's Hospital, MA (J.E.I., C.P., R.H.B.)
- Department of Surgery, Harvard Medical School, Boston, MA (J.E.I., C.P., R.H.B.)
| | - Roelof H Bekendam
- Vascular Biology Program, Boston Children's Hospital, MA (J.E.I., C.P., R.H.B.)
- Department of Surgery, Harvard Medical School, Boston, MA (J.E.I., C.P., R.H.B.)
- Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, MA (R.H.B.)
| |
Collapse
|
2
|
Yao HHY, Kahr WHA. Molecular basis of platelet granule defects. J Thromb Haemost 2024:S1538-7836(24)00706-2. [PMID: 39617187 DOI: 10.1016/j.jtha.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 01/02/2025]
Abstract
Platelets are small, discoid, anucleate blood cells that play key roles in clotting and other functions involved in health and disease. Platelets are derived from bone marrow-resident megakaryocytes, which undergo a complex developmental process where they increase dramatically in size and produce an abundance of organelles destined for platelets. These organelles include mitochondria, lysosomes, peroxisomes, and 2 unique types of secretory organelles: α- and dense (δ-) granules. δ-Granules contain small molecules, including adenosine triphosphate, adenosine diphosphate, serotonin, and ions, such as calcium and zinc (Ca2+ and Zn2+). α-Granules contain a variety of cargo proteins, which, when secreted by activated platelets, are involved in processes such as hemostasis (eg, fibrinogen and von Willebrand factor), angiogenesis, inflammation, and wound healing. Investigations of patients with inherited conditions resulting in decreased/abnormal platelet secretory granules have led to the identification of proteins, protein complexes, and cellular processes involved in their production by megakaryocytes. Notably, studies of ARPC1B deficiency, Hermansky-Pudlak, and Chediak-Higashi syndromes have linked several genes/proteins to δ-granule biogenesis. Studies of multisystemic arthrogryposis, renal dysfunction, and cholestasis syndrome revealed the requirement of 2 proteins, VPS33B and VPS16B, in α-granule formation. Identification of the genetic cause of gray platelet syndrome established that NBEAL2 is an additional protein needed for α-granule cargo retention. These discoveries enabled studies using animal models, cell culture, and molecular analysis to gain insights into the roles of proteins and cellular processes involved in platelet secretory granule production, which are discussed in this review.
Collapse
Affiliation(s)
- Helen H Y Yao
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Walter H A Kahr
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Division of Haematology/Oncology, Department of Paediatrics, University of Toronto, The Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
3
|
Carminita E, Becker IC, Italiano JE. What It Takes To Be a Platelet: Evolving Concepts in Platelet Production. Circ Res 2024; 135:540-549. [PMID: 39088641 DOI: 10.1161/circresaha.124.323579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
Platelets are among the most abundant cells within the circulation. Given that the platelet lifespan is 7 to 10 days in humans, a constant production of around 100 billion platelets per day is required. Platelet production from precursor cells called megakaryocytes is one of the most enigmatic processes in human biology. Although it has been studied for over a century, there is still controversy about the exact mechanisms leading to platelet release into circulation. The formation of proplatelet extensions from megakaryocytes into bone marrow sinusoids is the best-described mechanism explaining the origin of blood platelets. However, using powerful imaging techniques, several emerging studies have recently raised challenging questions in the field, suggesting that small platelet-sized structures called buds might also contribute to the circulating platelet pool. How and whether these structures differ from microvesicles or membrane blebs, which have previously been described to be released from megakaryocytes, is still a matter of discussion. In this review, we will summarize what the past and present have revealed about platelet production and whether mature blood platelets might emerge via different mechanisms.
Collapse
Affiliation(s)
- Estelle Carminita
- Vascular Biology Program, Boston Children's Hospital, Boston, MA (E.C., I.C.B., J.E.I.)
- Harvard Medical School, Boston, MA (E.C., I.C.B.)
| | - Isabelle C Becker
- Vascular Biology Program, Boston Children's Hospital, Boston, MA (E.C., I.C.B., J.E.I.)
- Harvard Medical School, Boston, MA (E.C., I.C.B.)
| | - Joseph E Italiano
- Vascular Biology Program, Boston Children's Hospital, Boston, MA (E.C., I.C.B., J.E.I.)
| |
Collapse
|
4
|
Tsuji C, Bradshaw M, Allen MF, Jackson ML, Mantell J, Borucu U, Poole AW, Verkade P, Hers I, Paul DM, Dodding MP. CryoET reveals actin filaments within platelet microtubules. Nat Commun 2024; 15:5967. [PMID: 39013865 PMCID: PMC11252303 DOI: 10.1038/s41467-024-50424-8] [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: 01/02/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
Crosstalk between the actin and microtubule cytoskeletons is important for many cellular processes. Recent studies have shown that microtubules and F-actin can assemble to form a composite structure where F-actin occupies the microtubule lumen. Whether these cytoskeletal hybrids exist in physiological settings and how they are formed is unclear. Here, we show that the short-crossover Class I actin filament previously identified inside microtubules in human HAP1 cells is cofilin-bound F-actin. Lumenal F-actin can be reconstituted in vitro, but cofilin is not essential. Moreover, actin filaments with both cofilin-bound and canonical morphologies reside within human platelet microtubules under physiological conditions. We propose that stress placed upon the microtubule network during motor-driven microtubule looping and sliding may facilitate the incorporation of actin into microtubules.
Collapse
Affiliation(s)
- Chisato Tsuji
- School of Biochemistry, Faculty of Health and Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK
| | - Marston Bradshaw
- School of Physiology, Pharmacology and Neuroscience, Faculty of Health and Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK
| | - Megan F Allen
- School of Physiology, Pharmacology and Neuroscience, Faculty of Health and Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK
| | - Molly L Jackson
- School of Physiology, Pharmacology and Neuroscience, Faculty of Health and Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK
| | - Judith Mantell
- School of Biochemistry, Faculty of Health and Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK
| | - Ufuk Borucu
- GW4 Facility for High-Resolution Electron Cryo-Microscopy, University of Bristol, Bristol, UK
| | - Alastair W Poole
- School of Physiology, Pharmacology and Neuroscience, Faculty of Health and Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK
| | - Paul Verkade
- School of Biochemistry, Faculty of Health and Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK
| | - Ingeborg Hers
- School of Physiology, Pharmacology and Neuroscience, Faculty of Health and Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK.
| | - Danielle M Paul
- School of Physiology, Pharmacology and Neuroscience, Faculty of Health and Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK.
| | - Mark P Dodding
- School of Biochemistry, Faculty of Health and Life Sciences, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK.
| |
Collapse
|
5
|
Becker IC, Wilkie AR, Nikols E, Carminita E, Roweth HG, Tilburg J, Sciaudone AR, Noetzli LJ, Fatima F, Couldwell G, Ray A, Mogilner A, Machlus KR, Italiano JE. Cell cycle-dependent centrosome clustering precedes proplatelet formation. SCIENCE ADVANCES 2024; 10:eadl6153. [PMID: 38896608 PMCID: PMC11186502 DOI: 10.1126/sciadv.adl6153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
Platelet-producing megakaryocytes (MKs) primarily reside in the bone marrow, where they duplicate their DNA content with each cell cycle resulting in polyploid cells with an intricate demarcation membrane system. While key elements of the cytoskeletal reorganizations during proplatelet formation have been identified, what initiates the release of platelets into vessel sinusoids remains largely elusive. Using a cell cycle indicator, we observed a unique phenomenon, during which amplified centrosomes in MKs underwent clustering following mitosis, closely followed by proplatelet formation, which exclusively occurred in G1 of interphase. Forced cell cycle arrest in G1 increased proplatelet formation not only in vitro but also in vivo following short-term starvation of mice. We identified that inhibition of the centrosomal protein kinesin family member C1 (KIFC1) impaired clustering and subsequent proplatelet formation, while KIFC1-deficient mice exhibited reduced platelet counts. In summary, we identified KIFC1- and cell cycle-mediated centrosome clustering as an important initiator of proplatelet formation from MKs.
Collapse
Affiliation(s)
- Isabelle C. Becker
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Adrian R. Wilkie
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Emma Nikols
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
| | - Estelle Carminita
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Harvey G. Roweth
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
- Brigham and Women’s Hospital, 4 Blackfan Circle, Boston, MA 02115, USA
| | - Julia Tilburg
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | | | - Leila J. Noetzli
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
- Brigham and Women’s Hospital, 4 Blackfan Circle, Boston, MA 02115, USA
| | - Farheen Fatima
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
| | | | - Anjana Ray
- Brigham and Women’s Hospital, 4 Blackfan Circle, Boston, MA 02115, USA
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences, New York University, 251 Mercer Street, New York, NY 10012, USA
| | - Kellie R. Machlus
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Joseph E. Italiano
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| |
Collapse
|
6
|
Becker IC, Wilkie AR, Unger BA, Sciaudone AR, Fatima F, Tsai IT, Xu K, Machlus KR, Italiano JE. Dynamic actin/septin network in megakaryocytes coordinates proplatelet elaboration. Haematologica 2024; 109:915-928. [PMID: 37675512 PMCID: PMC10905084 DOI: 10.3324/haematol.2023.283369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/18/2023] [Indexed: 09/08/2023] Open
Abstract
Megakaryocytes (MK) undergo extensive cytoskeletal rearrangements as they give rise to platelets. While cortical microtubule sliding has been implicated in proplatelet formation, the role of the actin cytoskeleton in proplatelet elongation is less understood. It is assumed that actin filament reorganization is important for platelet generation given that mouse models with mutations in actin-associated proteins exhibit thrombocytopenia. However, due to the essential role of the actin network during MK development, a differential understanding of the contribution of the actin cytoskeleton on proplatelet release is lacking. Here, we reveal that inhibition of actin polymerization impairs the formation of elaborate proplatelets by hampering proplatelet extension and bead formation along the proplatelet shaft, which was mostly independent of changes in cortical microtubule sliding. We identify Cdc42 and its downstream effectors, septins, as critical regulators of intracellular actin dynamics in MK, inhibition of which, similarly to inhibition of actin polymerization, impairs proplatelet movement and beading. Super-resolution microscopy revealed a differential association of distinctive septins with the actin and microtubule cytoskeleton, respectively, which was disrupted upon septin inhibition and diminished intracellular filamentous actin dynamics. In vivo, septins, similarly to F-actin, were subject to changes in expression upon enforcing proplatelet formation through prior platelet depletion. In summary, we demonstrate that a Cdc42/septin axis is not only important for MK maturation and polarization, but is further required for intracellular actin dynamics during proplatelet formation.
Collapse
Affiliation(s)
- Isabelle C Becker
- Vascular Biology Program, Boston Children's Hospital, 1 Blackfan Circle, Boston, MA, 02115; Department of Surgery, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115
| | - Adrian R Wilkie
- Vascular Biology Program, Boston Children's Hospital, 1 Blackfan Circle, Boston, MA, 02115; Department of Surgery, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115
| | - Bret A Unger
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720
| | | | - Farheen Fatima
- Vascular Biology Program, Boston Children's Hospital, 1 Blackfan Circle, Boston, MA, 02115; Department of Surgery, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115
| | - I-Ting Tsai
- Vascular Biology Program, Boston Children's Hospital, 1 Blackfan Circle, Boston, MA, 02115; Department of Surgery, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115
| | - Ke Xu
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720
| | - Kellie R Machlus
- Vascular Biology Program, Boston Children's Hospital, 1 Blackfan Circle, Boston, MA, 02115; Department of Surgery, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115
| | - Joseph E Italiano
- Vascular Biology Program, Boston Children's Hospital, 1 Blackfan Circle, Boston, MA, 02115; Department of Surgery, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115.
| |
Collapse
|
7
|
Manole CG, Soare C, Ceafalan LC, Voiculescu VM. Platelet-Rich Plasma in Dermatology: New Insights on the Cellular Mechanism of Skin Repair and Regeneration. Life (Basel) 2023; 14:40. [PMID: 38255655 PMCID: PMC10817627 DOI: 10.3390/life14010040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
The skin's recognised functions may undergo physiological alterations due to ageing, manifesting as varying degrees of facial wrinkles, diminished tautness, density, and volume. Additionally, these functions can be disrupted (patho)physiologically through various physical and chemical injuries, including surgical trauma, accidents, or chronic conditions like ulcers associated with diabetes mellitus, venous insufficiency, or obesity. Advancements in therapeutic interventions that boost the skin's innate regenerative abilities could significantly enhance patient care protocols. The application of Platelet-Rich Plasma (PRP) is widely recognized for its aesthetic and functional benefits to the skin. Yet, the endorsement of PRP's advantages often borders on the dogmatic, with its efficacy commonly ascribed solely to the activation of fibroblasts by the factors contained within platelet granules. PRP therapy is a cornerstone of regenerative medicine which involves the autologous delivery of conditioned plasma enriched by platelets. This is achieved by centrifugation, removing erythrocytes while retaining platelets and their granules. Despite its widespread use, the precise sequences of cellular activation, the specific cellular players, and the molecular machinery that drive PRP-facilitated healing are still enigmatic. There is still a paucity of definitive and robust studies elucidating these mechanisms. In recent years, telocytes (TCs)-a unique dermal cell population-have shown promising potential for tissue regeneration in various organs, including the dermis. TCs' participation in neo-angiogenesis, akin to that attributed to PRP, and their role in tissue remodelling and repair processes within the interstitia of several organs (including the dermis), offer intriguing insights. Their potential to contribute to, or possibly orchestrate, the skin regeneration process following PRP treatment has elicited considerable interest. Therefore, pursuing a comprehensive understanding of the cellular and molecular mechanisms at work, particularly those involving TCs, their temporal involvement in structural recovery following injury, and the interconnected biological events in skin wound healing and regeneration represents a compelling field of study.
Collapse
Affiliation(s)
- Catalin G. Manole
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Ultrastructural Pathology Laboratory, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| | - Cristina Soare
- Department of Oncological Dermatology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Laura Cristina Ceafalan
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Cell Biology, Neurosciences and Experimental Myology Laboratory, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
| | - Vlad M. Voiculescu
- Department of Oncological Dermatology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
8
|
Quiogue AR, Sumiyoshi E, Fries A, Chuang CH, Bowerman B. Microtubules oppose cortical actomyosin-driven membrane ingression during C. elegans meiosis I polar body extrusion. PLoS Genet 2023; 19:e1010984. [PMID: 37782660 PMCID: PMC10569601 DOI: 10.1371/journal.pgen.1010984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/12/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023] Open
Abstract
During C. elegans oocyte meiosis I cytokinesis and polar body extrusion, cortical actomyosin is locally remodeled to assemble a contractile ring that forms within and remains part of a much larger and actively contractile cortical actomyosin network. This network both mediates contractile ring dynamics and generates shallow ingressions throughout the oocyte cortex during polar body extrusion. Based on our analysis of requirements for CLS-2, a member of the CLASP family of proteins that stabilize microtubules, we recently proposed that a balance of actomyosin-mediated tension and microtubule-mediated stiffness limits membrane ingression throughout the oocyte during meiosis I polar body extrusion. Here, using live cell imaging and fluorescent protein fusions, we show that CLS-2 is part of a group of kinetochore proteins, including the scaffold KNL-1 and the kinase BUB-1, that also co-localize during meiosis I to structures called linear elements, which are present within the assembling oocyte spindle and also are distributed throughout the oocyte in proximity to, but appearing to underlie, the actomyosin cortex. We further show that KNL-1 and BUB-1, like CLS-2, promote the proper organization of sub-cortical microtubules and also limit membrane ingression throughout the oocyte. Moreover, nocodazole or taxol treatment to destabilize or stabilize oocyte microtubules leads to, respectively, excess or decreased membrane ingression throughout the oocyte. Furthermore, taxol treatment, and genetic backgrounds that elevate the levels of cortically associated microtubules, both suppress excess membrane ingression in cls-2 mutant oocytes. We propose that linear elements influence the organization of sub-cortical microtubules to generate a stiffness that limits cortical actomyosin-driven membrane ingression throughout the oocyte during meiosis I polar body extrusion. We discuss the possibility that this regulation of sub-cortical microtubule dynamics facilitates actomyosin contractile ring dynamics during C. elegans oocyte meiosis I cell division.
Collapse
Affiliation(s)
- Alyssa R. Quiogue
- Institute of Molecular Biology, University of Oregon, Eugen, Oregon, United States of America
| | - Eisuke Sumiyoshi
- Institute of Molecular Biology, University of Oregon, Eugen, Oregon, United States of America
| | - Adam Fries
- Institute of Molecular Biology, University of Oregon, Eugen, Oregon, United States of America
- Imaging Core, Office of the Vice President for Research University of Oregon, Eugene, Oregon, United States of America
| | - Chien-Hui Chuang
- Institute of Molecular Biology, University of Oregon, Eugen, Oregon, United States of America
| | - Bruce Bowerman
- Institute of Molecular Biology, University of Oregon, Eugen, Oregon, United States of America
| |
Collapse
|
9
|
Quiogue AR, Sumiyoshi E, Fries A, Chuang CH, Bowerman B. Cortical microtubules oppose actomyosin-driven membrane ingression during C. elegans meiosis I polar body extrusion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.26.542508. [PMID: 37292632 PMCID: PMC10245968 DOI: 10.1101/2023.05.26.542508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
During C. elegans oocyte meiosis I, cortical actomyosin is locally remodeled to assemble a contractile ring near the spindle. In contrast to mitosis, when most cortical actomyosin converges into a contractile ring, the small oocyte ring forms within and remains part of a much larger and actively contractile cortical actomyosin network. This network both mediates contractile ring dynamics and generates shallow ingressions throughout the oocyte cortex during polar body extrusion. Based on our analysis of requirements for CLS-2, a member of the CLASP family of proteins that stabilize microtubules, we recently proposed that a balance of actomyosin-mediated tension and microtubule-mediated stiffness are required for contractile ring assembly within the oocyte cortical actomyosin network. Here, using live cell imaging and fluorescent protein fusions, we show that CLS-2 is part of a complex of kinetochore proteins, including the scaffold KNL-1 and the kinase BUB-1, that also co-localize to patches distributed throughout the oocyte cortex during meiosis I. By reducing their function, we further show that KNL-1 and BUB-1, like CLS-2, are required for cortical microtubule stability, to limit membrane ingression throughout the oocyte, and for meiotic contractile ring assembly and polar body extrusion. Moreover, nocodazole or taxol treatment to destabilize or stabilize oocyte microtubules, respectively, leads to excess or decreased membrane ingression throughout the oocyte and defective polar body extrusion. Finally, genetic backgrounds that elevate cortical microtubule levels suppress the excess membrane ingression in cls-2 mutant oocytes. These results support our hypothesis that CLS-2, as part of a sub-complex of kinetochore proteins that also co-localize to patches throughout the oocyte cortex, stabilizes microtubules to stiffen the oocyte cortex and limit membrane ingression throughout the oocyte, thereby facilitating contractile ring dynamics and the successful completion of polar body extrusion during meiosis I.
Collapse
Affiliation(s)
| | | | - Adam Fries
- Institute of Molecular Biology
- Imaging Core, Office of the Vice President for Research, University of Oregon, Eugene, OR USA 97403
| | | | | |
Collapse
|
10
|
Lai J, Li Y, Ran M, Huang Q, Huang F, Zhu L, Wu Y, Zou W, Xie X, Tang Y, Yang F, Wu A, Ge G, Wu J. Xanthotoxin, a novel inducer of platelet formation, promotes thrombocytopoiesis via IL-1R1 and MEK/ERK signaling. Biomed Pharmacother 2023; 163:114811. [PMID: 37156117 DOI: 10.1016/j.biopha.2023.114811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/20/2023] [Accepted: 04/30/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND Thrombocytopenia is a common hematological disease caused by many factors. It usually complicates critical diseases and increases morbidity and mortality. The treatment of thrombocytopenia remains a great challenge in clinical practice, however, its treatment options are limited. In this study, the active monomer xanthotoxin (XAT) was screened out to explore its medicinal value and provide novel therapeutic strategies for the clinical treatment of thrombocytopenia. METHODS The effects of XAT on megakaryocyte differentiation and maturation were detected by flow cytometry, Giemsa and phalloidin staining. RNA-seq identified differentially expressed genes and enriched pathways. The signaling pathway and transcription factors were verified through WB and immunofluorescence staining. Tg (cd41: eGFP) transgenic zebrafish and mice with thrombocytopenia were used to evaluate the biological activity of XAT on platelet formation and the related hematopoietic organ index in vivo. RESULTS XAT promoted the differentiation and maturation of Meg-01 cells in vitro. Meanwhile, XAT could stimulate platelet formation in transgenic zebrafish and recover platelet production and function in irradiation-induced thrombocytopenia mice. Further RNA-seq prediction and WB verification revealed that XAT activates the IL-1R1 target and MEK/ERK signaling pathway, and upregulates the expression of transcription factors related to the hematopoietic lineage to promote megakaryocyte differentiation and platelet formation. CONCLUSION XAT accelerates megakaryocyte differentiation and maturation to promote platelet production and recovery through triggering IL-1R1 and activating the MEK/ERK signaling pathway, providing a new pharmacotherapy strategy for thrombocytopenia.
Collapse
Affiliation(s)
- Jia Lai
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Yueyue Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Mei Ran
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Qianqian Huang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Feihong Huang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Linjie Zhu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Yuesong Wu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Wenjun Zou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiang Xie
- School of Basic Medical Sciences, Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou 646000, China
| | - Yong Tang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Fei Yang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Anguo Wu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jianming Wu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
11
|
Asgari A, Jurasz P. Role of Nitric Oxide in Megakaryocyte Function. Int J Mol Sci 2023; 24:ijms24098145. [PMID: 37175857 PMCID: PMC10179655 DOI: 10.3390/ijms24098145] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Megakaryocytes are the main members of the hematopoietic system responsible for regulating vascular homeostasis through their progeny platelets, which are generally known for maintaining hemostasis. Megakaryocytes are characterized as large polyploid cells that reside in the bone marrow but may also circulate in the vasculature. They are generated directly or through a multi-lineage commitment step from the most primitive progenitor or Hematopoietic Stem Cells (HSCs) in a process called "megakaryopoiesis". Immature megakaryocytes enter a complicated development process defined as "thrombopoiesis" that ultimately results in the release of extended protrusions called proplatelets into bone marrow sinusoidal or lung microvessels. One of the main mediators that play an important modulatory role in hematopoiesis and hemostasis is nitric oxide (NO), a free radical gas produced by three isoforms of nitric oxide synthase within the mammalian cells. In this review, we summarize the effect of NO and its signaling on megakaryopoiesis and thrombopoiesis under both physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Amir Asgari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
| | - Paul Jurasz
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G-2H7, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G-2S2, Canada
- Mazankowski Alberta Heart Institute, Edmonton, AB T6G-2R7, Canada
| |
Collapse
|
12
|
Tilburg J, Stone AP, Billingsley JM, Scoville DK, Pavenko A, Liang Y, Italiano JE, Machlus KR. Spatial transcriptomics of murine bone marrow megakaryocytes at single-cell resolution. Res Pract Thromb Haemost 2023; 7:100158. [PMID: 37255850 PMCID: PMC10225915 DOI: 10.1016/j.rpth.2023.100158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 03/20/2023] [Accepted: 04/07/2023] [Indexed: 06/01/2023] Open
Abstract
Background While megakaryocytes are known for making platelets, recent single-cell RNA sequencing data have revealed subpopulations of megakaryocytes with predicted immunoregulatory and bone marrow niche-supporting roles. Although these studies uncovered interesting information regarding the transcriptional variation of megakaryocytes, the generation, localization, and regulation of these subsets have not yet been studied and therefore remain incompletely understood. Considering the complex organization of the bone marrow, we reasoned that the application of spatial transcriptomic approaches could help dissect megakaryocyte heterogeneity within a spatiotemporal context. Objectives The aim of this study was to combine spatial context and transcriptomics to assess the heterogeneity of murine bone marrow megakaryocytes in situ at a single-cell level. Methods Bone marrow sections were obtained from femurs of C57BL/6J mice. Using the murine whole transcriptome array on the Nanostring GeoMx digital spatial profiling platform, we profiled 44 individual megakaryocytes (CD41+ by immunofluorescence) in situ throughout the bone marrow, both adjacent and nonadjacent to the endothelium (directly in contact with vascular endothelial-cadherin-positive cells). Results Principal component analysis revealed no association between transcriptomic profile and adjacency to the vasculature. However, there was a significant effect of proximal vs distal regions of the bone. Two and 3 genes were found overexpressed in the proximal and distal sides, respectively. Of note, proplatelet basic protein and platelet factor 4, 2 genes associated with platelet production, had higher expression in proximal megakaryocytes. Conclusion This study indicates a possible effect of spatial location on megakaryocyte heterogeneity and substantiate further interest in investigating megakaryocyte subpopulations in the context of their spatial orientation.
Collapse
Affiliation(s)
- Julia Tilburg
- Department of Surgery, Harvard Medical School and Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Andrew P. Stone
- Department of Surgery, Harvard Medical School and Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - James M. Billingsley
- Harvard Chan Bioinformatics Core, Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, USA
| | | | - Anna Pavenko
- Nanostring Technologies Inc, Seattle, Washington, USA
| | - Yan Liang
- Nanostring Technologies Inc, Seattle, Washington, USA
| | - Joseph E. Italiano
- Department of Surgery, Harvard Medical School and Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Kellie R. Machlus
- Department of Surgery, Harvard Medical School and Vascular Biology Program, Boston Children’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
13
|
The Application of Ethnomedicine in Modulating Megakaryocyte Differentiation and Platelet Counts. Int J Mol Sci 2023; 24:ijms24043168. [PMID: 36834579 PMCID: PMC9961075 DOI: 10.3390/ijms24043168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/28/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
Megakaryocytes (MKs), a kind of functional hematopoietic stem cell, form platelets to maintain platelet balance through cell differentiation and maturation. In recent years, the incidence of blood diseases such as thrombocytopenia has increased, but these diseases cannot be fundamentally solved. The platelets produced by MKs can treat thrombocytopenia-associated diseases in the body, and myeloid differentiation induced by MKs has the potential to improve myelosuppression and erythroleukemia. Currently, ethnomedicine is extensively used in the clinical treatment of blood diseases, and the recent literature has reported that many phytomedicines can improve the disease status through MK differentiation. This paper reviewed the effects of botanical drugs on megakaryocytic differentiation covering the period 1994-2022, and information was obtained from PubMed, Web of Science and Google Scholar. In conclusions, we summarized the role and molecular mechanism of many typical botanical drugs in promoting megakaryocyte differentiation in vivo, providing evidence as much as possible for botanical drugs treating thrombocytopenia and other related diseases in the future.
Collapse
|
14
|
Tsuji C, Dodding MP. Lumenal components of cytoplasmic microtubules. Biochem Soc Trans 2022; 50:1953-1962. [PMID: 36524962 DOI: 10.1042/bst20220851] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 07/30/2023]
Abstract
The lumen of cytoplasmic microtubules is a poorly explored expanse of intracellular space. Although typically represented in textbooks as a hollow tube, studies over several decades have shown that the microtubule lumen is occupied by a range of morphologically diverse components. These are predominantly globular particles of varying sizes which appear to exist either in isolation, bind to the microtubule wall, or form discontinuous columns that extend through the lumenal space. Actin filaments with morphologies distinct from the canonical cytoplasmic forms have also now been found within the microtubule lumen. In this review, we examine the historic literature that observed these lumenal components in tissues from diverse species and integrate it with recent cryo-electron tomography studies that have begun to identify lumenal proteins. We consider their cell and tissue distribution, possible mechanisms of incorporation, and potential functions. It is likely that continuing work in this area will open a new frontier in cytoskeletal biology.
Collapse
Affiliation(s)
- Chisato Tsuji
- School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol BS8 1TD, U.K
| | - Mark P Dodding
- School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol BS8 1TD, U.K
| |
Collapse
|
15
|
Freitag M, Schwertz H. A New Role of NAP1L1 in Megakaryocytes and Human Platelets. Int J Mol Sci 2022; 23:ijms232314694. [PMID: 36499021 PMCID: PMC9737020 DOI: 10.3390/ijms232314694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/26/2022] Open
Abstract
Platelets (PLTs) are anucleate and considered incapable of nuclear functions. Contrastingly, nuclear proteins were detected in human PLTs. For most of these proteins, it is unclear if nuclear or alternatively assigned functions are performed, a question we wanted to address for nuclear assembly protein 1like 1 (NAP1L1). Using a wide array of molecular methods, including RNAseq, co-IP, overexpression and functional assays, we explored expression pattern and functionality of NAP1L1 in PLTs, and CD34+-derived megakaryocytes (MKs). NAP1L1 is expressed in PLTs and MKs. Co-IP experiments revealed that dihydrolipolylysine-residue acetyltransferase (DLAT encoded protein PDC-E2, ODP2) dynamically interacts with NAP1L1. PDC-E2 is part of the mitochondrial pyruvate-dehydrogenase (PDH) multi-enzyme complex, playing a crucial role in maintaining cellular respiration, and promoting ATP-synthesis via the respiratory chain. Since altered mitochondrial function is a hallmark of infectious syndromes, we analyzed PDH activity in PLTs from septic patients demonstrating increased activity, paralleling NAP1L1 expression levels. MKs PDH activity decreased following an LPS-challenge. Furthermore, overexpression of NAP1L1 significantly altered the ability of MKs to form proplatelet extensions, diminishing thrombopoiesis. These results indicate that NAP1L1 performs in other than nucleosome-assembly functions in PTLs and MKs, binding a key mitochondrial protein as a potential chaperone, and gatekeeper, influencing PDH activity and thrombopoiesis.
Collapse
Affiliation(s)
- Martin Freitag
- Department of Cardiac Surgery, Heart Center Leipzig-University Hospital, 04289 Leipzig, Germany
| | - Hansjörg Schwertz
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Division of Occupational Medicine, University of Utah, Salt Lake City, UT 84112, USA
- Occupational Medicine at Billings Clinic Bozeman, Bozeman, MT 59715, USA
- Correspondence: or
| |
Collapse
|
16
|
Zhang Y, Benz P, Stehle D, Yang S, Kurz H, Feil S, Nagel G, Feil R, Gao S, Bender M. Optogenetic manipulation of cyclic guanosine monophosphate to probe phosphodiesterase activities in megakaryocytes. Open Biol 2022; 12:220058. [PMID: 35975649 PMCID: PMC9382455 DOI: 10.1098/rsob.220058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Cyclic guanosine monophosphate (cGMP) signalling plays a fundamental role in many cell types, including platelets. cGMP has been implicated in platelet formation, but mechanistic detail about its spatio-temporal regulation in megakaryocytes (MKs) is lacking. Optogenetics is a technique which allows spatio-temporal manipulation of molecular events in living cells or organisms. We took advantage of this method and expressed a photo-activated guanylyl cyclase, Blastocladiella emersonii Cyclase opsin (BeCyclop), after viral-mediated gene transfer in bone marrow (BM)-derived MKs to precisely light-modulate cGMP levels. BeCyclop-MKs showed a significantly increased cGMP concentration after illumination, which was strongly dependent on phosphodiesterase (PDE) 5 activity. This finding was corroborated by real-time imaging of cGMP signals which revealed that pharmacological PDE5 inhibition also potentiated nitric oxide-triggered cGMP generation in BM MKs. In summary, we established for the first-time optogenetics in primary MKs and show that PDE5 is the predominant PDE regulating cGMP levels in MKs. These findings also demonstrate that optogenetics allows for the precise manipulation of MK biology.
Collapse
Affiliation(s)
- Yujing Zhang
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, 97080 Würzburg, Germany
| | - Pascal Benz
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, 97080 Würzburg, Germany
| | - Daniel Stehle
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Shang Yang
- Department of Neurophysiology, Institute of Physiology, Biocenter, University of Würzburg, 97070 Würzburg, Germany
| | - Hendrikje Kurz
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, 97080 Würzburg, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Georg Nagel
- Department of Neurophysiology, Institute of Physiology, Biocenter, University of Würzburg, 97070 Würzburg, Germany
| | - Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Shiqiang Gao
- Department of Neurophysiology, Institute of Physiology, Biocenter, University of Würzburg, 97070 Würzburg, Germany
| | - Markus Bender
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, 97080 Würzburg, Germany
| |
Collapse
|
17
|
De Wispelaere K, Freson K. The Analysis of the Human Megakaryocyte and Platelet Coding Transcriptome in Healthy and Diseased Subjects. Int J Mol Sci 2022; 23:ijms23147647. [PMID: 35886993 PMCID: PMC9317744 DOI: 10.3390/ijms23147647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 12/10/2022] Open
Abstract
Platelets are generated and released into the bloodstream from their precursor cells, megakaryocytes that reside in the bone marrow. Though platelets have no nucleus or DNA, they contain a full transcriptome that, during platelet formation, is transported from the megakaryocyte to the platelet. It has been described that transcripts in platelets can be translated into proteins that influence platelet response. The platelet transcriptome is highly dynamic and has been extensively studied using microarrays and, more recently, RNA sequencing (RNA-seq) in relation to diverse conditions (inflammation, obesity, cancer, pathogens and others). In this review, we focus on bulk and single-cell RNA-seq studies that have aimed to characterize the coding transcriptome of healthy megakaryocytes and platelets in humans. It has been noted that bulk RNA-seq has limitations when studying in vitro-generated megakaryocyte cultures that are highly heterogeneous, while single-cell RNA-seq has not yet been applied to platelets due to their very limited RNA content. Next, we illustrate how these methods can be applied in the field of inherited platelet disorders for gene discovery and for unraveling novel disease mechanisms using RNA from platelets and megakaryocytes and rare disease bioinformatics. Next, future perspectives are discussed on how this field of coding transcriptomics can be integrated with other next-generation technologies to decipher unexplained inherited platelet disorders in a multiomics approach.
Collapse
|
18
|
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: 5.0] [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
|
19
|
Kemble S, Dalby A, Lowe GC, Nicolson PLR, Watson SP, Senis Y, Thomas SG, Harrison P. Analysis of preplatelets and their barbell platelet derivatives by imaging flow cytometry. Blood Adv 2022; 6:2932-2946. [PMID: 35042240 PMCID: PMC9092408 DOI: 10.1182/bloodadvances.2021006073] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/21/2021] [Indexed: 11/20/2022] Open
Abstract
Circulating large "preplatelets" undergo fission via barbell platelet intermediates into two smaller, mature platelets. In this study, we determine whether preplatelets and/or barbells are equivalent to reticulated/immature platelets by using ImageStream flow cytometry and super-resolution microscopy. Immature platelets, preplatelets, and barbells were quantified in healthy and thrombocytopenic mice, healthy human volunteers, and patients with immune thrombocytopenia or undergoing chemotherapy. Preplatelets and barbells were 1.9% ± 0.18%/1.7% ± 0.48% (n = 6) and 3.3% ± 1.6%/0.5% ± 0.27% (n = 12) of total platelet counts in murine and human whole blood, respectively. Both preplatelets and barbells exhibited high expression of major histocompatibility complex class I with high thiazole orange and Mitotracker fluorescence. Tracking dye experiments confirmed that preplatelets transform into barbells and undergo fission ex vivo to increase platelet counts, with dependence on the cytoskeleton and normal mitochondrial respiration. Samples from antibody-induced thrombocytopenia in mice and patients with immune thrombocytopenia had increased levels of both preplatelets and barbells correlating with immature platelet levels. Furthermore, barbells were absent after chemotherapy in patients. In mice, in vivo biotinylation confirmed that barbells, but not all large platelets, were immature. This study demonstrates that a subpopulation of large platelets are immature preplatelets that can transform into barbells and undergo fission during maturation.
Collapse
Affiliation(s)
| | - Amanda Dalby
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
| | - Gillian C. Lowe
- West Midlands Haemophilia Comprehensive Care Centre, University Hospitals Birmingham Foundation Trust, Birmingham, United Kingdom; and
| | - Phillip L. R. Nicolson
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
- West Midlands Haemophilia Comprehensive Care Centre, University Hospitals Birmingham Foundation Trust, Birmingham, United Kingdom; and
| | - Steve P. Watson
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
| | - Yotis Senis
- 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, Université de Strasbourg, Strasbourg, France
| | - Steven G. Thomas
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
| | | |
Collapse
|
20
|
Abstract
This review discusses our understanding of platelet diversity with implications for the roles of platelets in hemostasis and thrombosis and identifies advanced technologies set to provide new insights. We use the term diversity to capture intrasubject platelet variability that can be intrinsic or governed by the environment and lead to a heterogeneous response pattern of aggregation, clot promotion, and external communication. Using choice examples, we discuss how the use of advanced technologies can provide new insights into the underlying causes of platelet molecular, structural, and functional diversity. As sources of diversity, we discuss the proliferating megakaryocytes with different allele-specific expression patterns, the asymmetrical formation of proplatelets, changes in platelets induced by aging and priming, interplatelet heterogeneity in thrombus organization and stability, and platelet-dependent communications. We provide indications how current knowledge gaps can be addressed using promising technologies, such as next-generation sequencing, proteomic approaches, advanced imaging techniques, multicolor flow and mass cytometry, multifunctional microfluidics assays, and organ-on-a-chip platforms. We then argue how this technology base can aid in characterizing platelet populations and in identifying platelet biomarkers relevant for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (J.W.M.H.)
| | - Jonathan West
- Faculty of Medicine and Centre for Hybrid Biodevices, University of Southampton, United Kingdom (J.W.)
| |
Collapse
|
21
|
Kimmerlin Q, Strassel C, Eckly A, Lanza F. The tubulin code in platelet biogenesis. Semin Cell Dev Biol 2022; 137:63-73. [PMID: 35148939 DOI: 10.1016/j.semcdb.2022.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 01/12/2022] [Accepted: 01/31/2022] [Indexed: 11/28/2022]
Abstract
Blood platelets are small non-nucleated cellular fragments that prevent and stop hemorrhages. They are produced in the bone marrow by megakaryocytes through megakaryopoiesis. This intricate process involves profound microtubule rearrangements culminating in the formation of a unique circular sub-membranous microtubule array, the marginal band, which supports the typical disc-shaped morphology of platelets. Mechanistically, these processes are thought to be controlled by a specific tubulin code. In this review, we summarize the current knowledge on the key isotypes, notably β1-, α4A- and α8-tubulin, and putative post-translational modifications, involved in platelet and marginal band formation. Additionally, we provide a provisional list of microtubule-associated proteins (MAPs) involved in these processes and a survey of tubulin variants identified in patients presenting defective platelet production. A comprehensive characterization of the platelet tubulin code and the identification of essential MAPs may be expected in the near future to shed new light on a very specialized microtubule assembly process with applications in platelet diseases and transfusion.
Collapse
Affiliation(s)
- Quentin Kimmerlin
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, Strasbourg, France.
| | - Catherine Strassel
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, Strasbourg, France.
| | - Anita Eckly
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, Strasbourg, France.
| | - François Lanza
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, Strasbourg, France.
| |
Collapse
|
22
|
O’Sullivan LR, Cahill MR, Young PW. The Importance of Alpha-Actinin Proteins in Platelet Formation and Function, and Their Causative Role in Congenital Macrothrombocytopenia. Int J Mol Sci 2021; 22:9363. [PMID: 34502272 PMCID: PMC8431150 DOI: 10.3390/ijms22179363] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/04/2022] Open
Abstract
The actin cytoskeleton plays a central role in platelet formation and function. Alpha-actinins (actinins) are actin filament crosslinking proteins that are prominently expressed in platelets and have been studied in relation to their role in platelet activation since the 1970s. However, within the past decade, several groups have described mutations in ACTN1/actinin-1 that cause congenital macrothrombocytopenia (CMTP)-accounting for approximately 5% of all cases of this condition. These findings are suggestive of potentially novel functions for actinins in platelet formation from megakaryocytes in the bone marrow and/or platelet maturation in circulation. Here, we review some recent insights into the well-known functions of actinins in platelet activation before considering possible roles for actinins in platelet formation that could explain their association with CMTP. We describe what is known about the consequences of CMTP-linked mutations on actinin-1 function at a molecular and cellular level and speculate how these changes might lead to the alterations in platelet count and morphology observed in CMTP patients. Finally, we outline some unanswered questions in this area and how they might be addressed in future studies.
Collapse
Affiliation(s)
- Leanne R. O’Sullivan
- School of Biochemistry & Cell Biology, University College Cork, T12 XF62 Cork, Ireland;
| | - Mary R. Cahill
- Department of Haematology and CancerResearch@UCC, Cork University Hospital, University College Cork, T12 XF62 Cork, Ireland;
| | - Paul W. Young
- School of Biochemistry & Cell Biology, University College Cork, T12 XF62 Cork, Ireland;
| |
Collapse
|
23
|
Scheller I, Beck S, Göb V, Gross C, Neagoe RAI, Aurbach K, Bender M, Stegner D, Nagy Z, Nieswandt B. Thymosin β4 is essential for thrombus formation by controlling the G-actin/F-actin equilibrium in platelets. Haematologica 2021; 107:2846-2858. [PMID: 34348450 PMCID: PMC9713564 DOI: 10.3324/haematol.2021.278537] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Coordinated rearrangements of the actin cytoskeleton are pivotal for platelet biogenesis from megakaryocytes but also orchestrate key functions of peripheral platelets in hemostasis and thrombosis, such as granule release, the formation of filopodia and lamellipodia, or clot retraction. Along with profilin (Pfn) 1, thymosin β4 (encoded by Tmsb4x) is one of the two main G-actin-sequestering proteins within cells of higher eukaryotes, and its intracellular concentration is particularly high in cells that rapidly respond to external signals by increased motility, such as platelets. Here, we analyzed constitutive Tmsb4x knockout (KO) mice to investigate the functional role of the protein in platelet production and function. Thymosin β4 deficiency resulted in a macrothrombocytopenia with only mildly increased platelet volume and an unaltered platelet life span. Megakaryocyte numbers in the bone marrow and spleen were unaltered, however, Tmsb4x KO megakaryocytes showed defective proplatelet formation in vitro and in vivo. Thymosin β4-deficient platelets displayed markedly decreased G-actin levels and concomitantly increased F-actin levels resulting in accelerated spreading on fibrinogen and clot retraction. Moreover, Tmsb4x KO platelets showed activation defects and an impaired immunoreceptor tyrosine-based activation motif (ITAM) signaling downstream of the activating collagen receptor glycoprotein VI. These defects translated into impaired aggregate formation under flow, protection from occlusive arterial thrombus formation in vivo and increased tail bleeding times. In summary, these findings point to a critical role of thymosin β4 for actin dynamics during platelet biogenesis, platelet activation downstream of glycoprotein VI and thrombus stability.
Collapse
Affiliation(s)
- Inga Scheller
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany,*IS and SB contributed equally as co-first authors
| | - Sarah Beck
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany,*IS and SB contributed equally as co-first authors
| | - Vanessa Göb
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany
| | - Carina Gross
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany
| | - Raluca A. I. Neagoe
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany,Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Katja Aurbach
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany
| | - Markus Bender
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany
| | - David Stegner
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany
| | - Zoltan Nagy
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, and Rudolf Virchow Center for Integrative and Translational BioImaging, University of Würzburg, Würzburg, Germany,B. Nieswandt
| |
Collapse
|
24
|
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
|
25
|
Mitochondrial dynamics and reactive oxygen species initiate thrombopoiesis from mature megakaryocytes. Blood Adv 2021; 5:1706-1718. [PMID: 33720340 DOI: 10.1182/bloodadvances.2020002847] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
Blood platelets are essential for controlling hemostasis. They are released by megakaryocytes (MKs) located in the bone marrow, upon extension of cytoplasmic protrusions into the lumen of bone marrow sinusoids. Their number increases in postpulmonary capillaries, suggesting a role for oxygen gradient in thrombopoiesis (ie, platelet biogenesis). In this study, we show that initiation of thrombopoiesis from human mature MKs was enhanced under hyperoxia or during pro-oxidant treatments, whereas antioxidants dampened it. Quenching mitochondrial reactive oxygen species (mtROS) with MitoTEMPO decreased thrombopoiesis, whereas genetically enhancing mtROS by deacetylation-null sirtuin-3 expression increased it. Blocking cytosolic ROS production by NOX inhibitors had no impact. Classification according to the cell roundness index delineated 3 stages of thrombopoiesis in mature MKs. Early-stage round MKs exhibited the highest index, which correlated with low mtROS levels, a mitochondrial tubular network, and the mitochondrial recruitment of the fission activator Drp1. Intermediate MKs at the onset of thrombopoiesis showed high mtROS levels and small, well-delineated mitochondria. Terminal MKs showed the lowest roundness index and long proplatelet extensions. Inhibiting Drp1-dependent mitochondrial fission of mature MKs by Mdivi-1 favored a tubular mitochondrial network and lowered both mtROS levels and intermediate MKs proportion, whereas enhancing Drp1 activity genetically had opposite effects. Reciprocally, quenching mtROS limited mitochondrial fission in round MKs. These data demonstrate a functional coupling between ROS and mitochondrial fission in MKs, which is crucial for the onset of thrombopoiesis. They provide new molecular cues that control initiation of platelet biogenesis and may help elucidate some unexplained thrombocytopenia.
Collapse
|
26
|
Actin/microtubule crosstalk during platelet biogenesis in mice is critically regulated by Twinfilin1 and Cofilin1. Blood Adv 2021; 4:2124-2134. [PMID: 32407474 DOI: 10.1182/bloodadvances.2019001303] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/13/2020] [Indexed: 01/24/2023] Open
Abstract
Rearrangements of the microtubule (MT) and actin cytoskeleton are pivotal for platelet biogenesis. Hence, defects in actin- or MT-regulatory proteins are associated with platelet disorders in humans and mice. Previous studies in mice revealed that loss of the actin-depolymerizing factor homology (ADF-H) protein Cofilin1 (Cof1) in megakaryocytes (MKs) results in a moderate macrothrombocytopenia but normal MK numbers, whereas deficiency in another ADF-H protein, Twinfilin1 (Twf1), does not affect platelet production or function. However, recent studies in yeast have indicated a critical synergism between Twf1 and Cof1 in the regulation of actin dynamics. We therefore investigated platelet biogenesis and function in mice lacking both Twf1 and Cof1 in the MK lineage. In contrast to single deficiency in either protein, Twf1/Cof1 double deficiency (DKO) resulted in a severe macrothrombocytopenia and dramatically increased MK numbers in bone marrow and spleen. DKO MKs exhibited defective proplatelet formation in vitro and in vivo as well as impaired spreading and altered assembly of podosome-like structures on collagen and fibrinogen in vitro. These defects were associated with aberrant F-actin accumulation and, remarkably, the formation of hyperstable MT, which appears to be caused by dysregulation of the actin- and MT-binding proteins mDia1 and adenomatous polyposis coli. Surprisingly, the mild functional defects described for Cof1-deficient platelets were only slightly aggravated in DKO platelets suggesting that both proteins are largely dispensable for platelet function in the peripheral blood. In summary, these findings reveal critical redundant functions of Cof1 and Twf1 in ensuring balanced actin/microtubule crosstalk during thrombopoiesis in mice and possibly humans.
Collapse
|
27
|
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: 2.5] [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
|
28
|
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: 4.8] [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
|
29
|
Platelets parameters in septic shock: clinical usefulness and prognostic value. Blood Coagul Fibrinolysis 2021; 31:421-425. [PMID: 33065574 DOI: 10.1097/mbc.0000000000000937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
: Septic shock is a common cause of admission in the ICUs. Despite tremendous improvement in the management modalities, mortality remains high. Early diagnosis and prompt resuscitation are required to improve prognosis. Therefore, identifying a biomarker that could reveal the sepsis at its earlier stage is of paramount importance. In this regards, platelet parameters, such as mean platelet volume, immature platelet fraction and platelet-derived microparticles have been investigated as possible sepsis biomarkers. In fact, haemostasis disturbances are one of the hallmark of septic shock where platelets play a pivotal role in orchestrating the inflammatory response of the host. Moreover, these parameters could have a prognostic value as the severity of the multiorgan dysfunction is correlated with the inflammatory reaction.
Collapse
|
30
|
Mini-review: Microtubule sliding in neurons. Neurosci Lett 2021; 753:135867. [PMID: 33812935 DOI: 10.1016/j.neulet.2021.135867] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/28/2022]
Abstract
Microtubule sliding is an underappreciated mechanism that contributes to the establishment, organization, preservation, and plasticity of neuronal microtubule arrays. Powered by molecular motor proteins and regulated in part by static crosslinker proteins, microtubule sliding is the movement of microtubules relative to other microtubules or to non-microtubule structures such as the actin cytoskeleton. In addition to other important functions, microtubule sliding significantly contributes to the establishment and maintenance of microtubule polarity patterns in different regions of the neuron. The purpose of this article is to review the state of knowledge on microtubule sliding in the neuron, with emphasis on its mechanistic underpinnings as well as its functional significance.
Collapse
|
31
|
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
|
32
|
Khan AO, Slater A, Maclachlan A, Nicolson PLR, Pike JA, Reyat JS, Yule J, Stapley R, Rayes J, Thomas SG, Morgan NV. Post-translational polymodification of β1-tubulin regulates motor protein localisation in platelet production and function. Haematologica 2020; 107:243-259. [PMID: 33327716 PMCID: PMC8719104 DOI: 10.3324/haematol.2020.270793] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Indexed: 11/17/2022] Open
Abstract
In specialized cells, the expression of specific tubulin isoforms and their subsequent post-translational modifications drive and coordinate unique morphologies and behaviors. The mechanisms by which b1-tubulin, the platelet and megakaryocyte (MK) lineage restricted tubulin isoform, drives platelet production and function remains poorly understood. We investigated the roles of two key post-translational tubulin polymodifications (polyglutamylation and polyglycylation) on these processes using a cohort of thrombocytopenic patients, human induced pluripotent stem cell derived MK, and healthy human donor platelets. We find distinct patterns of polymodification in MK and platelets, mediated by the antagonistic activities of the cell specific expression of tubulin tyrosine ligase like enzymes and cytosolic carboxypeptidase enzymes. The resulting microtubule patterning spatially regulates motor proteins to drive proplatelet formation in megakaryocytes, and the cytoskeletal reorganization required for thrombus formation. This work is the first to show a reversible system of polymodification by which different cell specific functions are achieved.
Collapse
Affiliation(s)
- Abdullah O Khan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK, B15 2TT.
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK, B15 2TT
| | - Annabel Maclachlan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK, B15 2TT
| | - Phillip L R Nicolson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK, B15 2TT
| | - Jeremy A Pike
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK, B15 2TT; Centre of Membrane and Protein and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands
| | - Jasmeet S Reyat
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK, B15 2TT
| | - Jack Yule
- Centre of Membrane and Protein and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands
| | - Rachel Stapley
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK, B15 2TT
| | - Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK, B15 2TT
| | - Steven G Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK, B15 2TT; Centre of Membrane and Protein and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands
| | - Neil V Morgan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK, B15 2TT.
| |
Collapse
|
33
|
Boscher J, Guinard I, Eckly A, Lanza F, Léon C. Blood platelet formation at a glance. J Cell Sci 2020; 133:133/20/jcs244731. [DOI: 10.1242/jcs.244731] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
ABSTRACT
The main function of blood platelets is to ensure hemostasis and prevent hemorrhages. The 1011 platelets needed daily are produced in a well-orchestrated process. However, this process is not yet fully understood and in vitro platelet production is still inefficient. Platelets are produced in the bone marrow by megakaryocytes, highly specialized precursor cells that extend cytoplasmic projections called proplatelets (PPTs) through the endothelial barrier of sinusoid vessels. In this Cell Science at a Glance article and the accompanying poster we discuss the mechanisms and pathways involved in megakaryopoiesis and platelet formation processes. We especially address the – still underestimated – role of the microenvironment of the bone marrow, and present recent findings on how PPT extension in vivo differs from that in vitro and entails different mechanisms. Finally, we recapitulate old but recently revisited evidence that – although bone marrow does produce megakaryocytes and PPTs – remodeling and the release of bona fide platelets, mainly occur in the downstream microcirculation.
Collapse
Affiliation(s)
- Julie Boscher
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - Ines Guinard
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - Anita Eckly
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - François Lanza
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - Catherine Léon
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| |
Collapse
|
34
|
PAK1 Regulates MEC-17 Acetyltransferase Activity and Microtubule Acetylation during Proplatelet Extension. Int J Mol Sci 2020; 21:ijms21207531. [PMID: 33066011 PMCID: PMC7589885 DOI: 10.3390/ijms21207531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Mature megakaryocytes extend long processes called proplatelets from which platelets are released in the blood stream. The Rho GTPases Cdc42 and Rac as well as their downstream target, p21-activated kinase 2 (PAK2), have been demonstrated to be important for platelet formation. Here we address the role, during platelet formation, of PAK1, another target of the Rho GTPases. PAK1 decorates the bundled microtubules (MTs) of megakaryocyte proplatelets. Using a validated cell model which recapitulates proplatelet formation, elongation and platelet release, we show that lack of PAK1 activity increases the number of proplatelets but restrains their elongation. Moreover, in the absence of PAK1 activity, cells have hyperacetylated MTs and lose their MT network integrity. Using inhibitors of the tubulin deacetylase HDAC6, we demonstrate that abnormally high levels of MT acetylation are not sufficient to increase the number of proplatelets but cause loss of MT integrity. Taken together with our previous demonstration that MT acetylation is required for proplatelet formation, our data reveal that MT acetylation levels need to be tightly regulated during proplatelet formation. We identify PAK1 as a direct regulator of the MT acetylation levels during this process as we found that PAK1 phosphorylates the MT acetyltransferase MEC-17 and inhibits its activity.
Collapse
|
35
|
French SL, Vijey P, Karhohs KW, Wilkie AR, Horin LJ, Ray A, Posorske B, Carpenter AE, Machlus KR, Italiano JE. High-content, label-free analysis of proplatelet production from megakaryocytes. J Thromb Haemost 2020; 18:2701-2711. [PMID: 32662223 PMCID: PMC7988437 DOI: 10.1111/jth.15012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 07/03/2020] [Accepted: 07/09/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND The mechanisms that regulate platelet biogenesis remain unclear; factors that trigger megakaryocytes (MKs) to initiate platelet production are poorly understood. Platelet formation begins with proplatelets, which are cellular extensions originating from the MK cell body. OBJECTIVES Proplatelet formation is an asynchronous and dynamic process that poses unique challenges for researchers to accurately capture and analyze. We have designed an open-source, high-content, high-throughput, label-free analysis platform. METHODS Phase-contrast images of live, primary MKs are captured over a 24-hour period. Pixel-based machine-learning classification done by ilastik generates probability maps of key cellular features (circular MKs and branching proplatelets), which are processed by a customized CellProfiler pipeline to identify and filter structures of interest based on morphology. A subsequent reinforcement classification, by CellProfiler Analyst, improves the detection of cellular structures. RESULTS This workflow yields the percent of proplatelet production, area, count of proplatelets and MKs, and other statistics including skeletonization information for measuring proplatelet branching and length. We propose using a combination of these analyzed metrics, in particular the area measurements of MKs and proplatelets, when assessing in vitro proplatelet production. Accuracy was validated against manually counted images and an existing algorithm. We then used the new platform to test compounds known to cause thrombocytopenia, including bromodomain inhibitors, and uncovered previously unrecognized effects of drugs on proplatelet formation, thus demonstrating the utility of our analysis platform. CONCLUSION This advance in creating unbiased data analysis will increase the scale and scope of proplatelet production studies and potentially serve as a valuable resource for investigating molecular mechanisms of thrombocytopenia.
Collapse
Affiliation(s)
- Shauna L. French
- Division of Hematology, Brigham and Women’s Hospital; Boston, MA, USA 02115
- Department of Medicine, Harvard Medical School; Boston, MA, USA 02115
| | - Prakrith Vijey
- Division of Hematology, Brigham and Women’s Hospital; Boston, MA, USA 02115
| | - Kyle W. Karhohs
- Imaging Platform, Broad Institute of Harvard and MIT; Cambridge, MA, USA 02142
| | - Adrian R. Wilkie
- Division of Hematology, Brigham and Women’s Hospital; Boston, MA, USA 02115
- Department of Medicine, Harvard Medical School; Boston, MA, USA 02115
| | - Lillian J. Horin
- Department of Medicine, Harvard Medical School; Boston, MA, USA 02115
- Department of Systems Biology, Harvard Medical School; Boston, MA, USA 02115
| | - Anjana Ray
- Division of Hematology, Brigham and Women’s Hospital; Boston, MA, USA 02115
| | - Benjamin Posorske
- Division of Hematology, Brigham and Women’s Hospital; Boston, MA, USA 02115
| | - Anne E. Carpenter
- Imaging Platform, Broad Institute of Harvard and MIT; Cambridge, MA, USA 02142
| | - Kellie R. Machlus
- Division of Hematology, Brigham and Women’s Hospital; Boston, MA, USA 02115
- Department of Medicine, Harvard Medical School; Boston, MA, USA 02115
| | - Joseph E. Italiano
- Division of Hematology, Brigham and Women’s Hospital; Boston, MA, USA 02115
- Department of Medicine, Harvard Medical School; Boston, MA, USA 02115
- Vascular Biology Program, Department of Surgery; Boston Children’s Hospital; Boston, MA, USA 02115
| |
Collapse
|
36
|
Zuidscherwoude M, Haining EJ, Simms VA, Watson S, Grygielska B, Hardy AT, Bacon A, Watson SP, Thomas SG. Loss of mDia1 and Fhod1 impacts platelet formation but not platelet function. Platelets 2020; 32:1051-1062. [PMID: 32981398 PMCID: PMC8635707 DOI: 10.1080/09537104.2020.1822522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
An organized and dynamic cytoskeleton is required for platelet formation and function. Formins are a large family of actin regulatory proteins which are also able to regulate microtubule dynamics. There are four formin family members expressed in human and mouse megakaryocytes and platelets. We have previously shown that the actin polymerization activity of formin proteins is required for cytoskeletal dynamics and platelet spreading using a small molecule inhibitor. In the current study, we analyze transgenic mouse models deficient in two of these proteins, mDia1 and Fhod1, along with a model lacking both proteins. We demonstrate that double knockout mice display macrothrombocytopenia which is due to aberrant megakaryocyte function and a small decrease in platelet lifespan. Platelet function is unaffected by the loss of these proteins. This data indicates a critical role for formins in platelet and megakaryocyte function.
Collapse
Affiliation(s)
- Malou Zuidscherwoude
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Elizabeth J. Haining
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Victoria A. Simms
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Stephanie Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Beata Grygielska
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Alex T. Hardy
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Andrea Bacon
- Genome Editing Facility, Technology Hub, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Stephen P. Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Steven G. Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| |
Collapse
|
37
|
|
38
|
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: 1.2] [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
|
39
|
Abstract
Recent advances in super-resolution (sub-diffraction limited) microscopy have yielded remarkable insights into the nanoscale architecture and behavior of cells. In addition to the capacity to provide sub 100 nm resolution, these technologies offer unique quantitative opportunities with particular relevance to platelet and megakaryocyte biology. In this review, we provide a short introduction to modern super-resolution microscopy, its applications in the field of platelet and megakaryocyte biology, and emerging quantitative approaches which will allow for unprecedented insights into the biology of these unique cell types.
Collapse
Affiliation(s)
- Abdullah O Khan
- Institute of Cardiovascular Sciences, College of Medical and Dental Science, University of Birmingham , Birmingham, UK
| | - Jeremy A Pike
- Institute of Cardiovascular Sciences, College of Medical and Dental Science, University of Birmingham , Birmingham, UK.,Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham , UK
| |
Collapse
|
40
|
Abstract
There is increasing awareness that platelets play a significant role in creating a hypercoagulable environment that mediates tumor progression, beyond their classical hemostatic function. Platelets have heterogenic responses to agonists, and differential release and uptake of bioactive molecules may be manipulated via reciprocal cross-talk with cells of the tumor microenvironment. Platelets thus promote tumor progression by enhancing tumor growth, promoting the development of tumor-associated vasculature and encouraging invasion. In the metastatic process, platelets form the shield that protects tumor cells from high-velocity forces and immunosurveillance, while ensuring the establishment of the pre-metastatic niche. This review presents the complexity of these concepts, considering platelets as biomarkers for diagnosis, prognosis and potentially as therapeutic targets in cancer.
Collapse
Affiliation(s)
- Tanya N Augustine
- School of Anatomical Sciences, Faculty of the Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| |
Collapse
|
41
|
Abstract
Platelets - blood cells continuously produced from megakaryocytes mainly in the bone marrow - are implicated not only in haemostasis and arterial thrombosis, but also in other physiological and pathophysiological processes. This Review describes current evidence for the heterogeneity in platelet structure, age, and activation properties, with consequences for a diversity of platelet functions. Signalling processes of platelet populations involved in thrombus formation with ongoing coagulation are well understood. Genetic approaches have provided information on multiple genes related to normal haemostasis, such as those encoding receptors and signalling or secretory proteins, that determine platelet count and/or responsiveness. As highly responsive and secretory cells, platelets can alter the environment through the release of growth factors, chemokines, coagulant factors, RNA species, and extracellular vesicles. Conversely, platelets will also adapt to their environment. In disease states, platelets can be positively primed to reach a pre-activated condition. At the inflamed vessel wall, platelets interact with leukocytes and the coagulation system, interactions mediating thromboinflammation. With current antiplatelet therapies invariably causing bleeding as an undesired adverse effect, novel therapies can be more beneficial if directed against specific platelet responses, populations, interactions, or priming conditions. On the basis of these novel concepts and processes, we discuss several initiatives to target platelets therapeutically.
Collapse
|
42
|
Luo XL, Jiang JY, Huang Z, Chen LX. Autophagic regulation of platelet biology. J Cell Physiol 2019; 234:14483-14488. [PMID: 30714132 DOI: 10.1002/jcp.28243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/25/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Platelets, developed from megakaryocytes, are characterized by anucleate and short-life span hemocyte in mammal vessel. Platelets are very important in the cardiovascular system. Studies indicate the occurrence of autophagy platelets and megakaryocytes. Moreover, abnormal autophagy decreases the number of platelets and suppresses platelet aggregation. In addition, mitophagy, as a kind of selective autophagy, could inhibit platelet aggregation under oxidative stress or hypoxic, whereas promote platelet aggregation after reperfusion. Finally, autophagy regulates hemorrhagic and thrombosis diseases by influencing the number and function of platelets. In this paper, the role of autophagy in platelets and megakaryocytes, as well as coupled with the promotive or inhibitory role of hemorrhagic and thrombosis diseases are elucidated. Therefore, autophagy may be a potentially therapeutic target in modulating the platelet-related diseases.
Collapse
Affiliation(s)
- Xu-Ling Luo
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Jin-Yong Jiang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Zhen Huang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Lin-Xi Chen
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
43
|
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: 4.3] [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
|
44
|
Gaertner F, Massberg S. Patrolling the vascular borders: platelets in immunity to infection and cancer. Nat Rev Immunol 2019; 19:747-760. [DOI: 10.1038/s41577-019-0202-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2019] [Indexed: 12/13/2022]
|
45
|
Lo RW, Li L, Leung R, Pluthero FG, Kahr WHA. NBEAL2 (Neurobeachin-Like 2) Is Required for Retention of Cargo Proteins by α-Granules During Their Production by Megakaryocytes. Arterioscler Thromb Vasc Biol 2019; 38:2435-2447. [PMID: 30354215 DOI: 10.1161/atvbaha.118.311270] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective- Human and mouse megakaryocytes lacking NBEAL2 (neurobeachin-like 2) produce platelets where α-granules lack protein cargo. This cargo is mostly megakaryocyte-synthesized, but some proteins, including FGN (fibrinogen), are endocytosed. In this study, we examined the trafficking of both types of cargo within primary megakaryocytes cultured from normal and NBEAL2-null mice, to determine the role of NBEAL2 in α-granule maturation. We also examined the interaction of NBEAL2 with the granule-associated protein P-selectin in human megakaryocytes and platelets. Approach and Results- Fluorescence microscopy was used to compare uptake of labeled FGN by normal and NBEAL2-null mouse megakaryocytes, which was similar in both. NBEAL2-null cells, however, showed decreased FGN retention, and studies with biotinylated protein showed rapid loss rather than increased degradation. Intracellular tracking via fluorescence microscopy revealed that in normal megakaryocytes, endocytosed FGN sequentially associated with compartments expressing RAB5 (Ras-related protein in brain 5), RAB7 (Ras-related protein in brain 7), and P-selectin, where it was retained. A similar initial pattern was observed in NBEAL2-null megakaryocytes, but then FGN passed from the P-selectin compartment to RAB11 (Ras-related protein in brain 11)-associated endosomes before release. Megakaryocyte-synthesized VWF (Von Willebrand factor) was observed to follow the same route out of NBEAL2-null cells. Immunofluorescence microscopy revealed intracellular colocalization of NBEAL2 with P-selectin in human megakaryocytes, proplatelets, and platelets. Native NBEAL2 and P-selectin were coimmunoprecipitated from platelets and megakaryocytes. Conclusions- NBEAL2 is not required for FGN uptake by megakaryocytes. NBEAL2 is required for the retention of both endocytosed and megakaryocyte-synthesized proteins by maturing α-granules, and possibly by platelet-borne granules. This function may involve interaction of NBEAL2 with P-selectin.
Collapse
Affiliation(s)
- Richard W Lo
- From the Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada (R.W.L., L.L., R.L., F.G.P., W.H.A.K.).,Department of Biochemistry, University of Toronto, ON, Canada (R.W.L., W.H.A.K.)
| | - Ling Li
- From the Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada (R.W.L., L.L., R.L., F.G.P., W.H.A.K.)
| | - Richard Leung
- From the Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada (R.W.L., L.L., R.L., F.G.P., W.H.A.K.)
| | - Fred G Pluthero
- From the Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada (R.W.L., L.L., R.L., F.G.P., W.H.A.K.)
| | - Walter H A Kahr
- From the Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada (R.W.L., L.L., R.L., F.G.P., W.H.A.K.).,Department of Biochemistry, University of Toronto, ON, Canada (R.W.L., W.H.A.K.).,Division of Haematology/Oncology, Department of Paediatrics, University of Toronto and The Hospital for Sick Children, ON, Canada (W.H.A.K.)
| |
Collapse
|
46
|
Noetzli LJ, French SL, Machlus KR. New Insights Into the Differentiation of Megakaryocytes From Hematopoietic Progenitors. Arterioscler Thromb Vasc Biol 2019; 39:1288-1300. [PMID: 31043076 PMCID: PMC6594866 DOI: 10.1161/atvbaha.119.312129] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/22/2019] [Indexed: 02/07/2023]
Abstract
Megakaryocytes are hematopoietic cells, which are responsible for the production of blood platelets. The traditional view of megakaryopoiesis describes the cellular journey from hematopoietic stem cells, through a hierarchical series of progenitor cells, ultimately to a mature megakaryocyte. Once mature, the megakaryocyte then undergoes a terminal maturation process involving multiple rounds of endomitosis and cytoplasmic restructuring to allow platelet formation. However, recent studies have begun to redefine this hierarchy and shed new light on alternative routes by which hematopoietic stem cells are differentiated into megakaryocytes. In particular, the origin of megakaryocytes, including the existence and hierarchy of megakaryocyte progenitors, has been redefined, as new studies are suggesting that hematopoietic stem cells originate as megakaryocyte-primed and can bypass traditional lineage checkpoints. Overall, it is becoming evident that megakaryopoiesis does not only occur as a stepwise process, but is dynamic and adaptive to biological needs. In this review, we will reexamine the canonical dogmas of megakaryopoiesis and provide an updated framework for interpreting the roles of traditional pathways in the context of new megakaryocyte biology. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Leila J Noetzli
- Division of Hematology, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Shauna L French
- Division of Hematology, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Kellie R Machlus
- Division of Hematology, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
47
|
Almazni I, Stapley R, Morgan NV. Inherited Thrombocytopenia: Update on Genes and Genetic Variants Which may be Associated With Bleeding. Front Cardiovasc Med 2019; 6:80. [PMID: 31275945 PMCID: PMC6593073 DOI: 10.3389/fcvm.2019.00080] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 05/31/2019] [Indexed: 01/10/2023] Open
Abstract
Inherited thrombocytopenia (IT) is comprised of a group of hereditary disorders characterized by a reduced platelet count as the main feature, and often with abnormal platelet function, which can subsequently lead to impaired haemostasis. Inherited thrombocytopenia results from genetic mutations in genes implicated in megakaryocyte differentiation and/or platelet formation and clearance. The identification of the underlying causative gene of IT is challenging given the high degree of heterogeneity, but important due to the presence of various clinical presentations and prognosis, where some defects can lead to hematological malignancies. Traditional platelet function tests, clinical manifestations, and hematological parameters allow for an initial diagnosis. However, employing Next-Generation Sequencing (NGS), such as Whole Genome and Whole Exome Sequencing (WES) can be an efficient method for discovering causal genetic variants in both known and novel genes not previously implicated in IT. To date, 40 genes and their mutations have been implicated to cause many different forms of inherited thrombocytopenia. Nevertheless, despite this advancement in the diagnosis of IT, the molecular mechanism underlying IT in some patients remains unexplained. In this review, we will discuss the genetics of thrombocytopenia summarizing the recent advancement in investigation and diagnosis of IT using phenotypic approaches, high-throughput sequencing, targeted gene panels, and bioinformatics tools.
Collapse
Affiliation(s)
- Ibrahim Almazni
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Rachel Stapley
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Neil V Morgan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
48
|
Bächer C, Gekle S. Computational modeling of active deformable membranes embedded in three-dimensional flows. Phys Rev E 2019; 99:062418. [PMID: 31330647 DOI: 10.1103/physreve.99.062418] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Indexed: 06/10/2023]
Abstract
Active gel theory has recently been very successful in describing biologically active materials such as actin filaments or moving bacteria in temporally fixed and simple geometries such as cubes or spheres. Here we develop a computational algorithm to compute the dynamic evolution of an arbitrarily shaped, deformable thin membrane of active material embedded in a three-dimensional flowing liquid. For this, our algorithm combines active gel theory with the classical theory of thin elastic shells. To compute the actual forces resulting from active stresses, we apply a parabolic fitting procedure to the triangulated membrane surface. Active forces are then dynamically coupled via an immersed-boundary method to the surrounding fluid whose dynamics can be solved by any standard, e.g., Lattice-Boltzmann, flow solver. We validate our algorithm using the Green's functions of Berthoumieux et al. [New J. Phys. 16, 065005 (2014)10.1088/1367-2630/16/6/065005] for an active cylindrical membrane subjected (i) to a locally increased active stress and (ii) to a homogeneous active stress. For the latter scenario, we predict in addition a nonaxisymmetric instability. We highlight the versatility of our method by analyzing the flow field inside an actively deforming cell embedded in external shear flow. Further applications may be cytoplasmic streaming or active membranes in blood flows.
Collapse
Affiliation(s)
- Christian Bächer
- Biofluid Simulation and Modeling, Theoretische Physik VI, Universität Bayreuth, Universitätsstrasse 30, Bayreuth, Germany
| | - Stephan Gekle
- Biofluid Simulation and Modeling, Theoretische Physik VI, Universität Bayreuth, Universitätsstrasse 30, Bayreuth, Germany
| |
Collapse
|
49
|
Schurr Y, Spindler M, Kurz H, Bender M. The cytoskeletal crosslinking protein MACF1 is dispensable for thrombus formation and hemostasis. Sci Rep 2019; 9:7726. [PMID: 31118482 PMCID: PMC6531446 DOI: 10.1038/s41598-019-44183-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/27/2019] [Indexed: 12/15/2022] Open
Abstract
Coordinated reorganization of cytoskeletal structures is critical for key aspects of platelet physiology. While several studies have addressed the role of microtubules and filamentous actin in platelet production and function, the significance of their crosstalk in these processes has been poorly investigated. The microtubule-actin cross-linking factor 1 (MACF1; synonym: Actin cross-linking factor 7, ACF7) is a member of the spectraplakin family, and one of the few proteins expressed in platelets, which possess actin and microtubule binding domains thereby facilitating actin-microtubule interaction and regulation. We used megakaryocyte- and platelet-specific Macf1 knockout (Macf1fl/fl, Pf4-Cre) mice to study the role of MACF1 in platelet production and function. MACF1 deficient mice displayed comparable platelet counts to control mice. Analysis of the platelet cytoskeletal ultrastructure revealed a normal marginal band and actin network. Platelet spreading on fibrinogen was slightly delayed but platelet activation and clot traction was unaffected. Ex vivo thrombus formation and mouse tail bleeding responses were similar between control and mutant mice. These results suggest that MACF1 is dispensable for thrombopoiesis, platelet activation, thrombus formation and the hemostatic function in mice.
Collapse
Affiliation(s)
- Yvonne Schurr
- Institute of Experimental Biomedicine - Chair I, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - Markus Spindler
- Institute of Experimental Biomedicine - Chair I, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - Hendrikje Kurz
- Institute of Experimental Biomedicine - Chair I, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - Markus Bender
- Institute of Experimental Biomedicine - Chair I, University Hospital and Rudolf Virchow Center, Würzburg, Germany.
| |
Collapse
|
50
|
Matsumura T, Nakamura-Ishizu A, Takaoka K, Maki H, Muddineni SSNA, Wang CQ, Suzushima H, Kawakita M, Asou N, Matsuoka M, Kurokawa M, Osato M, Suda T. TUBB1 dysfunction in inherited thrombocytopenia causes genome instability. Br J Haematol 2019; 185:888-902. [PMID: 30854628 DOI: 10.1111/bjh.15835] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/18/2018] [Indexed: 12/20/2022]
Abstract
Inherited thrombocytopenia is a genetically heterogeneous disease characterized by varying degrees of thrombocytopenia and risk of haematological malignancy, and the genetic cause of many cases remains unknown. We performed whole-exome sequencing of a family with thrombocytopenia and myeloid malignancy and identified a novel TUBB1 variant, T149P. Screening of other thrombocytopenia pedigrees identified another TUBB1 variant, R251H. TUBB1 encodes the tubulin β-1 chain, a major component of microtubules abundant in megakaryocytes. Variant TUBB1 disrupted the normal assembly of microtubules and impaired proplatelet formation in vitro. In addition, DNA damage response was severely attenuated by loss of TUBB1. We found that the nuclear accumulation of p53 (also termed TP53) and the expression of pro-apoptotic genes triggered by genotoxic stress were blocked in TUBB1-deficient cells and, accordingly, apoptosis after DNA damage was diminished by knockdown of TUBB1. Thus, we have demonstrated that microtubule dysfunction confers resistance to apoptosis, even in DNA damage-accumulated cells, which explains genome instability in the affected individuals. These studies will lead us to a better understanding of how microtubule dysfunction can contribute to the accumulation of DNA damage, genetic instability and leukaemogenesis.
Collapse
Affiliation(s)
- Takayoshi Matsumura
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Ayako Nakamura-Ishizu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,International Research Centre for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kensuke Takaoka
- Department of Haematology and Oncology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Hiroaki Maki
- Department of Haematology and Oncology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Siva S N A Muddineni
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Chelsia Q Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | | | | | - Norio Asou
- International Medical Centre, Saitama Medical University, Saitama, Japan
| | - Masao Matsuoka
- Department of Haematology, Rheumatology, and Infectious Diseases, Kumamoto University School of Medicine, Kumamoto, Japan
| | - Mineo Kurokawa
- Department of Haematology and Oncology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Motomi Osato
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,International Research Centre for Medical Sciences, Kumamoto University, Kumamoto, Japan.,Centre for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, Japan
| | - Toshio Suda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,International Research Centre for Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|