1
|
Snoeck HW. Direct megakaryopoiesis. Curr Opin Hematol 2025:00062752-990000000-00109. [PMID: 40197720 DOI: 10.1097/moh.0000000000000871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
PURPOSE OF REVIEW Megakaryocytes are large, polyploid cells that produce platelets and originate from hematopoietic stem cells (HSCs) in the bone marrow. While in the classical paradigm, megakaryocytes are generated in a stepwise fashion through increasingly committed progenitor stages, studies using in-vivo barcoding, transplantation, and in-vitro culture have suggested that, in addition, a more direct pathway existed. The relevance of this direct pathway and its functional and phenotypic characteristics were unclear, however. RECENT FINDINGS Recent publications using fate-mapping and single-cell transplantation now unequivocally demonstrate the existence of a direct megakaryocyte differentiation pathway, provide molecular characterization, and indicate distinct roles and regulation of both pathways. The direct pathway originates from a separate subset of 'top' HSCs, is enhanced by hematopoietic stress, inflammation and aging, bypasses multipotential progenitors, may be more active in myeloproliferative neoplasms, and generates phenotypically distinct megakaryocyte progenitors and more reactive platelets. SUMMARY Novel insights into the direct megakaryocyte differentiation pathway provide a deeper understanding of HSC biology, hematological recovery after myeloablation, and aging of the hematopoietic system, and suggest that this pathway may contribute to the increase in thrombotic incidents with age and in myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Hans-Willem Snoeck
- Columbia Center for Stem Cell Therapies/Columbia Center for Human Development, Department of Medicine
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons
- Division of Pulmonary Medicine, Allergy and Critical Care, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
2
|
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
|
3
|
Fallatah W, Dhinoja S, Al Qaryoute A, Mary J, Cheng V, Jagadeeswaran P. Identification of Nfel1a and Nfel3 as novel regulators for zebrafish thrombopoiesis. Blood Cells Mol Dis 2025; 110:102897. [PMID: 39413675 DOI: 10.1016/j.bcmd.2024.102897] [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] [Received: 05/02/2024] [Revised: 10/06/2024] [Accepted: 10/09/2024] [Indexed: 10/18/2024]
Abstract
In mammalian hematopoiesis, megakaryocytes mature and become polyploid in the bone marrow before releasing platelets into circulation. In contrast, fish produce thrombocytes in kidney marrow, where young thrombocytes undergo maturation in circulation, akin to platelets. Despite morphological differences, our single-cell sequencing revealed significant gene expression similarities between fish thrombocytes and mammalian megakaryocytes, including Nfe2, which is crucial in platelet production. In addition to nfe2 expression, we found four nfe2-related genes, nfe2I1a, nfe2I1b, nfe2I2a, and nfe2I3, were expressed in mature thrombocytes. However, only nfe2, nfe2l2a, and nfe2l3 are expressed in young thrombocytes. Thus, we hypothesized that Nfe2-related factors may also be involved in thrombocyte production. To address this, we knocked down the four novel nfe2-related genes by the piggyback method and found both young and mature thrombocytes reduced when nfe2, nfe2l1a, and nfe2l3 were knocked down. They also exhibited greater gill bleeding and prolonged time to occlusion (TTO) compared to controls. In summary, our study shows Nfe2I1a and Nfe2l3 as novel transcription factors that are positive regulators influencing adult zebrafish thrombopoiesis.
Collapse
Affiliation(s)
- Weam Fallatah
- Department of Biological Sciences, University of North Texas, Denton, TX 76203, United States of America
| | - Sanchi Dhinoja
- Department of Biological Sciences, University of North Texas, Denton, TX 76203, United States of America
| | - Ayah Al Qaryoute
- Department of Biological Sciences, University of North Texas, Denton, TX 76203, United States of America
| | - Jabila Mary
- Department of Biological Sciences, University of North Texas, Denton, TX 76203, United States of America
| | - Vallerie Cheng
- Department of Biological Sciences, University of North Texas, Denton, TX 76203, United States of America
| | - Pudur Jagadeeswaran
- Department of Biological Sciences, University of North Texas, Denton, TX 76203, United States of America.
| |
Collapse
|
4
|
Cortese M, Hagan T, Rouphael N, Wu SY, Xie X, Kazmin D, Wimmers F, Gupta S, van der Most R, Coccia M, Aranuchalam PS, Nakaya HI, Wang Y, Coyle E, Horiuchi S, Wu H, Bower M, Mehta A, Gunthel C, Bosinger SE, Kotliarov Y, Cheung F, Schwartzberg PL, Germain RN, Tsang J, Li S, Albrecht R, Ueno H, Subramaniam S, Mulligan MJ, Khurana S, Golding H, Pulendran B. System vaccinology analysis of predictors and mechanisms of antibody response durability to multiple vaccines in humans. Nat Immunol 2025; 26:116-130. [PMID: 39747435 DOI: 10.1038/s41590-024-02036-z] [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: 07/15/2024] [Accepted: 11/11/2024] [Indexed: 01/04/2025]
Abstract
We performed a systems vaccinology analysis to investigate immune responses in humans to an H5N1 influenza vaccine, with and without the AS03 adjuvant, to identify factors influencing antibody response magnitude and durability. Our findings revealed a platelet and adhesion-related blood transcriptional signature on day 7 that predicted the longevity of the antibody response, suggesting a potential role for platelets in modulating antibody response durability. As platelets originate from megakaryocytes, we explored the effect of thrombopoietin (TPO)-mediated megakaryocyte activation on antibody response longevity. We found that TPO administration enhanced the durability of vaccine-induced antibody responses. TPO-activated megakaryocytes also promoted survival of human bone-marrow plasma cells through integrin β1/β2-mediated cell-cell interactions, along with survival factors APRIL and the MIF-CD74 axis. Using machine learning, we developed a classifier based on this platelet-associated signature, which predicted antibody response longevity across six vaccines from seven independent trials, highlighting a conserved mechanism for vaccine durability.
Collapse
Affiliation(s)
- Mario Cortese
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Thomas Hagan
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Sheng-Yang Wu
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Xia Xie
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Dmitri Kazmin
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Florian Wimmers
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Shakti Gupta
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | | | | | - Prabhu S Aranuchalam
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | | | - Yating Wang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Elizabeth Coyle
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Shu Horiuchi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hanchih Wu
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary Bower
- Hope Clinic of the Emory Vaccine Center, Decatur, GA, USA
| | - Aneesh Mehta
- Hope Clinic of the Emory Vaccine Center, Decatur, GA, USA
| | | | - Steve E Bosinger
- Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA, USA
- Yerkes Genomics Core Laboratory, Yerkes National Primate Research Center, Atlanta, GA, USA
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuri Kotliarov
- NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD, USA
| | - Foo Cheung
- NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD, USA
| | - Pamela L Schwartzberg
- NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD, USA
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Ronald N Germain
- NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD, USA
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - John Tsang
- NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD, USA
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Shuzhao Li
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Randy Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hideki Ueno
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology, Kyoto University, Kyoto, Japan
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Mark J Mulligan
- Division of Infectious Diseases and Immunology, Department of Medicine and NYU Langone Vaccine Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
5
|
Furniss JA, Tarassova N, Poole AW. Platelet generation in vivo and in vitro. Blood 2024; 144:2283-2294. [PMID: 39357055 DOI: 10.1182/blood.2024024601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/08/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
ABSTRACT Platelets play crucial roles in hemostasis, thrombosis, and immunity, but our understanding of their complex biogenesis (thrombopoiesis) is currently incomplete. Deeper insight into the mechanisms of platelet biogenesis inside and outside the body is fundamental for managing hematological disorders and for the development of novel cell-based therapies. In this article, we address the current understanding of in vivo thrombopoiesis, including mechanisms of platelet generation from megakaryocytes (proplatelet formation, cytoplasmic fragmentation, and membrane budding) and their physiological location. Progress has been made in replicating these processes in vitro for potential therapeutic application, notably in platelet transfusion and bioengineering of platelets for novel targeted therapies. The current platelet-generating systems and their limitations, particularly yield, scalability, and functionality, are discussed. Finally, we highlight the current controversies and challenges in the field that need to be addressed to achieve a full understanding of these processes, in vivo and in vitro.
Collapse
Affiliation(s)
- Jonathan A Furniss
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Nathalie Tarassova
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Alastair W Poole
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
6
|
Chen C, Wang N, Zhang X, Fu Y, Zhong Z, Wu H, Wei Y, Duan Y. Highly efficient generation of mature megakaryocytes and functional platelets from human embryonic stem cells. Stem Cell Res Ther 2024; 15:454. [PMID: 39609933 PMCID: PMC11603724 DOI: 10.1186/s13287-024-04071-x] [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: 08/05/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Platelet transfusion therapy has made a great breakthrough in clinical practice, and the differentiation of human embryonic stem cells (hESCs) to produce functional platelets has become a new potential approach, however, efficient generation of functional platelets still faces great challenges. Here, we presented a novel approach to highly and efficiently generate mature megakaryocytes (MKs) and functional platelets from hESCs. METHODS In hypoxic conditions, we successfully replicated the maturation process of MKs and platelets in a controlled in vitro environment by introducing an optimal combination of cytokines at various stages of development. This method led to the generation of MKs and platelets derived from hESCs. Subsequently, mature MKs and functional platelets were further comprehensively investigated and characterized using a variety of methodologies, including flow cytometry analysis, RT-qPCR validation, Giemsa-Wright's staining, immunofluorescent staining, RNA transcriptome analysis, and DNA ploidy analysis. Additionally, the in vivo function of platelets was evaluated through the transplantation using thrombocytopenia model mice. RESULTS Under our 3D differentiation conditions with four sequential stages, hESCs could be efficiently induced into mature MKs, with 95% expressing CD41aCD42a or 90% expressing CD41aCD42b, and those MKs exhibited polyploid properties, produced filamentous proplatelet structures and further generated platelets. Furthermore, 95% of platelets showed CD42b+CD62p+ phenotype upon the stimulation with ADP and TRAP-6, while 50% of platelets exhibited the ability to bind PAC-1, indicating that hESC-derived platelets possessed the in vitro functionality. In mice models of thrombocytopenia, hESC-derived platelets effectively restored hemostasis in a manner comparable to human blood-derived platelets. Further investigation on the mechanism of this sequential differentiation revealed that cellular differentiation and molecular interactions during the generation of hESC-derived MKs and platelets recapitulated the developmental trajectory of the megakaryopoiesis and thrombopoiesis. CONCLUSIONS Thus, our results demonstrated that we successfully established a highly efficient differentiation of hESCs into mature MKs and functional platelets in vitro. The in vivo functionality of hESC-derived platelets closely resembles that of natural human platelets, thus offering a promising avenue for the development of functional platelets suitable for future clinical applications.
Collapse
Affiliation(s)
- Chuxin Chen
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Guangzhou, 510180, People's Republic of China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Ning Wang
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Guangzhou, 510180, People's Republic of China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Xueyan Zhang
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Guangzhou, 510180, People's Republic of China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Yingjie Fu
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Guangzhou, 510180, People's Republic of China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Zhiyong Zhong
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Haibin Wu
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Guangzhou, 510180, People's Republic of China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Yaming Wei
- Department of Blood Transfusion, the Second Affiliation Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Guangzhou, 510180, China.
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Guangzhou, 510180, People's Republic of China.
- Department of Blood Transfusion, the Second Affiliation Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Guangzhou, 510180, China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China.
- The Innovation Centre of Ministry of Education for Development and Diseases, the Second Affiliated Hospital of South China University of Technology, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
7
|
Ning Y, Zheng M, Zhang Y, Jiao Y, Wang J, Zhang S. RhoA-ROCK2 signaling possesses complex pathophysiological functions in cancer progression and shows promising therapeutic potential. Cancer Cell Int 2024; 24:339. [PMID: 39402585 PMCID: PMC11475559 DOI: 10.1186/s12935-024-03519-7] [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] [Received: 01/12/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024] Open
Abstract
The Rho GTPase signaling pathway is responsible for cell-specific processes, including actin cytoskeleton organization, cell motility, cell division, and the transcription of specific genes. The implications of RhoA and the downstream effector ROCK2 in cancer epithelial-mesenchymal transition, migration, invasion, and therapy resistance associated with stem cells highlight the potential of targeting RhoA/ROCK2 signaling in therapy. Tumor relapse can occur due to cancer cells that do not fully respond to adjuvant chemoradiotherapy, targeted therapy, or immunotherapy. Rho signaling-mediated mitotic defects and cytokinesis failure lead to asymmetric cell division, allowing cells to form polyploids to escape cytotoxicity and promote tumor recurrence and metastasis. In this review, we elucidate the significance of RhoA/ROCK2 in the mechanisms of cancer progression and summarize their inhibitors that may improve treatment strategies.
Collapse
Affiliation(s)
- Yidi Ning
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, P.R. China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, P.R. China
| | - Yue Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, P.R. China
| | - Yuqi Jiao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, P.R. China
| | - Jiangping Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, P.R. China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, P.R. China.
| |
Collapse
|
8
|
Milosevic TV, Vertenoeil G, Vainchenker W, Tulkens PM, Constantinescu SN, Van Bambeke F. Oxazolidinone antibiotics impair ex vivo megakaryocyte differentiation from hematopoietic progenitor cells and their maturation into platelets. Antimicrob Agents Chemother 2024; 68:e0053324. [PMID: 39297641 PMCID: PMC11460550 DOI: 10.1128/aac.00533-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/04/2024] [Indexed: 10/09/2024] Open
Abstract
Oxazolidinones (linezolid and tedizolid) adverse reactions include thrombocytopenia, the mechanism of which is still largely unknown. In cultured cells, oxazolidinones impair mitochondrial protein synthesis and oxidative metabolism. As mitochondrial activity is essential for megakaryocyte differentiation and maturation into platelets, we examined whether oxazolidinones impair these processes ex vivo and alter, in parallel, the activity of mitochondrial cytochrome c-oxidase (CYTOX; enzyme partly encoded by the mitochondrial genome) and cell morphology. Human CD34+ cells were isolated, incubated with cytokines (up to 14 days) and clinically relevant oxazolidinone concentrations or in control conditions, and used for (i) clonogenic assays [counting of megakaryocyte (CFU-Mk), granulocyte-monocyte (CFU-GM), burst-forming unit-erythroid (BFU-E) colonies]; (ii) the measure of the expression of megakaryocyte surface antigens (CD34 to CD41 and CD42); (iii) counting of proplatelets; (iv) the measurement of CYTOX activity; and (v) cell morphology (optic and electron microscopy). Oxazolidinones caused a significant decrease in BFU-E but not CFU-Mk or CFU-GM colonies. Yet, the megakaryocytic lineage was markedly affected, with a decreased differentiation of CD34+ into CD41+/CD42+ cells, an abolition of proplatelet formation and striking decrease in the numbers of large polylobulated nucleus megakaryocytes, with a complete loss of intracellular demarcation membrane system, disappearance of mitochondria, and suppression of CYTOX activity. These alterations were more marked in cells incubated with tedizolid than linezolid. These data suggest that oxazolidinones may induce thrombocytopenia by impairing megakaryocytic differentiation through mitochondrial dysfunction. Pharmacological interventions to prevent this toxicity might therefore be difficult as mitochondrial toxicity is most probably inherently linked to their antibacterial activity.
Collapse
Affiliation(s)
- Tamara V. Milosevic
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Gaëlle Vertenoeil
- Signal Transduction and Molecular Hematology Unit (SIGN), de Duve Institute, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Ludwig Institute for Cancer Research, Brussels, Belgium
| | - William Vainchenker
- UMR 1170, Institut National de la Santé et de la Recherche Médicale, Université de Paris-Sud & Institut Gustave Roussy, Villejuif, France
| | - Paul M. Tulkens
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Stefan N. Constantinescu
- Signal Transduction and Molecular Hematology Unit (SIGN), de Duve Institute, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Ludwig Institute for Cancer Research, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
- Nuffield Department of Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
9
|
Faiz M, Kalev‐Zylinska ML, Dunstan‐Harrison C, Singleton DC, Hay MP, Ledgerwood EC. Megakaryocyte maturation involves activation of the adaptive unfolded protein response. Genes Cells 2024; 29:889-901. [PMID: 39138929 PMCID: PMC11555628 DOI: 10.1111/gtc.13151] [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: 02/12/2024] [Revised: 07/21/2024] [Accepted: 07/26/2024] [Indexed: 08/15/2024]
Abstract
Endoplasmic reticulum stress triggers the unfolded protein response (UPR) to promote cell survival or apoptosis. Transient endoplasmic reticulum stress activation has been reported to trigger megakaryocyte production, and UPR activation has been reported as a feature of megakaryocytic cancers. However, the role of UPR signaling in megakaryocyte biology is not fully understood. We studied the involvement of UPR in human megakaryocytic differentiation using PMA (phorbol 12-myristate 13-acetate)-induced maturation of megakaryoblastic cell lines and thrombopoietin-induced differentiation of human peripheral blood-derived progenitors. Our results demonstrate that an adaptive UPR is a feature of megakaryocytic differentiation and that this response is not associated with ER stress-induced apoptosis. Differentiation did not alter the response to the canonical endoplasmic reticulum stressors DTT or thapsigargin. However, thapsigargin, but not DTT, inhibited differentiation, consistent with the involvement of Ca2+ signaling in megakaryocyte differentiation.
Collapse
Affiliation(s)
- Mifra Faiz
- Department of BiochemistrySchool of Biomedical Sciences, University of OtagoDunedinNew Zealand
| | - Maggie L. Kalev‐Zylinska
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine & PathologyFaculty of Medical and Health Sciences, The University of AucklandAucklandNew Zealand
| | | | - Dean C. Singleton
- Auckland Cancer Society Research CentreFaculty of Medical and Health Sciences, The University of AucklandAucklandNew Zealand
| | - Michael P. Hay
- Auckland Cancer Society Research CentreFaculty of Medical and Health Sciences, The University of AucklandAucklandNew Zealand
| | - Elizabeth C. Ledgerwood
- Department of BiochemistrySchool of Biomedical Sciences, University of OtagoDunedinNew Zealand
| |
Collapse
|
10
|
Edwards F, Fantozzi G, Simon AY, Morretton JP, Herbette A, Tijhuis AE, Wardenaar R, Foulane S, Gemble S, Spierings DC, Foijer F, Mariani O, Vincent-Salomon A, Roman-Roman S, Sastre-Garau X, Goundiam O, Basto R. Centrosome amplification primes ovarian cancer cells for apoptosis and potentiates the response to chemotherapy. PLoS Biol 2024; 22:e3002759. [PMID: 39236086 PMCID: PMC11441705 DOI: 10.1371/journal.pbio.3002759] [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: 11/23/2023] [Revised: 09/30/2024] [Accepted: 07/17/2024] [Indexed: 09/07/2024] Open
Abstract
Centrosome amplification is a feature of cancer cells associated with chromosome instability and invasiveness. Enhancing chromosome instability and subsequent cancer cell death via centrosome unclustering and multipolar divisions is an aimed-for therapeutic approach. Here, we show that centrosome amplification potentiates responses to conventional chemotherapy in addition to its effect on multipolar divisions and chromosome instability. We perform single-cell live imaging of chemotherapy responses in epithelial ovarian cancer cell lines and observe increased cell death when centrosome amplification is induced. By correlating cell fate with mitotic behaviors, we show that enhanced cell death can occur independently of chromosome instability. We identify that cells with centrosome amplification are primed for apoptosis. We show they are dependent on the apoptotic inhibitor BCL-XL and that this is not a consequence of mitotic stresses associated with centrosome amplification. Given the multiple mechanisms that promote chemotherapy responses in cells with centrosome amplification, we assess such a relationship in an epithelial ovarian cancer patient cohort. We show that high centrosome numbers associate with improved treatment responses and longer overall survival. Our work identifies apoptotic priming as a clinically relevant consequence of centrosome amplification, expanding our understanding of this pleiotropic cancer cell feature.
Collapse
Affiliation(s)
- Frances Edwards
- Biology of centrosomes and genetic instability, Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Giulia Fantozzi
- Biology of centrosomes and genetic instability, Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Anthony Y. Simon
- Biology of centrosomes and genetic instability, Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Jean-Philippe Morretton
- Biology of centrosomes and genetic instability, Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Aurelie Herbette
- Department of Translational Research, Institut Curie, PSL University, Paris, France
| | - Andrea E. Tijhuis
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rene Wardenaar
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Stacy Foulane
- Biology of centrosomes and genetic instability, Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Simon Gemble
- Biology of centrosomes and genetic instability, Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Diana C.J. Spierings
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | | | - Sergio Roman-Roman
- Department of Translational Research, Institut Curie, PSL University, Paris, France
| | | | - Oumou Goundiam
- Department of Translational Research, Institut Curie, PSL University, Paris, France
| | - Renata Basto
- Biology of centrosomes and genetic instability, Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| |
Collapse
|
11
|
Garyn CM, Bover O, Murray JW, Ma J, Salas-Briceno K, Ross SR, Snoeck HW. G2 arrest primes hematopoietic stem cells for megakaryopoiesis. Cell Rep 2024; 43:114388. [PMID: 38935497 PMCID: PMC11330628 DOI: 10.1016/j.celrep.2024.114388] [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: 10/12/2023] [Revised: 04/22/2024] [Accepted: 06/06/2024] [Indexed: 06/29/2024] Open
Abstract
In contrast to most hematopoietic lineages, megakaryocytes (MKs) can derive rapidly and directly from hematopoietic stem cells (HSCs). The underlying mechanism is unclear, however. Here, we show that DNA damage induces MK markers in HSCs and that G2 arrest, an integral part of the DNA damage response, suffices for MK priming followed by irreversible MK differentiation in HSCs, but not in progenitors. We also show that replication stress causes DNA damage in HSCs and is at least in part due to uracil misincorporation in vitro and in vivo. Consistent with this notion, thymidine attenuated DNA damage, improved HSC maintenance, and reduced the generation of CD41+ MK-committed HSCs. Replication stress and concomitant MK differentiation is therefore one of the barriers to HSC maintenance. DNA damage-induced MK priming may allow rapid generation of a lineage essential to immediate organismal survival, while also removing damaged cells from the HSC pool.
Collapse
Affiliation(s)
- Corey M Garyn
- Columbia Center for Human Development/Center for Stem Cell Therapies, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Oriol Bover
- Columbia Center for Human Development/Center for Stem Cell Therapies, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - John W Murray
- Columbia Center for Human Development/Center for Stem Cell Therapies, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jing Ma
- Department of Microbiology and Immunology, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| | - Karen Salas-Briceno
- Department of Microbiology and Immunology, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| | - Susan R Ross
- Department of Microbiology and Immunology, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| | - Hans-Willem Snoeck
- Columbia Center for Human Development/Center for Stem Cell Therapies, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA.
| |
Collapse
|
12
|
Wang Z, Liu Z, Zhou P, Niu X, Sun Z, He H, Zhu Z. The involvement of krüppel-like transcription factor 2 in megakaryocytic differentiation induction by phorbol 12-myrestrat 13-acetate. Biomark Res 2024; 12:65. [PMID: 39014479 PMCID: PMC11253501 DOI: 10.1186/s40364-024-00614-9] [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: 02/23/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Megakaryocytic differentiation is a complicated process regulated by a series of transcription factors in a context- and stage-dependent manner. Recent studies have suggested that krüppel-like transcription factor 2 (KLF2) is involved in the control of embryonic erythroid precursor cell differentiation and maturation. However, the function and mechanism of KLF2 in regulating megakaryocytic differentiation remain unclear. METHODS The expression patterns of krüppel-like transcription factors (KLFs) during megakaryocytic differentiation were identified from public databases. Phorbol 12-myristate 13-acetate (PMA) treatment of the myeloid-erythroid-leukemic cell lines K562 and HEL were used as cellular megakaryocytic differentiation models. A lentiviral transduction system was utilized to achieve the goal of amplifying or reducing KLF2. The expression of KLF2 was examined using real-time PCR and western blot. The impact of KLF2 on the megakaryocytic differentiation of K562 cells was examined by flow cytometry, Giemsa staining, Phalloidin staining and western blot. RNA-sequencing (RNA-seq) and chromatin immunoprecipitation-sequencing (ChIP-seq) technologies were used to identify the KLF2-regulated targets. RESULTS KLF2 is increased in the maturation process of megakaryocytes. KLF2 overexpression accelerated the PMA-induced megakaryocytic differentiation, as reflected by an increased percentage of CD41/CD61 cells, an increased number of polyploid cells, and an elevated expression of P21 and P27. KLF2 knockdown exhibited the opposite results, indicating that KLF2 knockdown suppressed the megakaryocytic differentiation. Further, combination of the RNA-seq and ChIP-seq results suggested that chimerin 1 (CHN1) and potassium voltage-gated channel subfamily Q member 5 (KCNQ5) may be target genes regulated of KLF2. Both CHN1 and KCNQ5 knockdown could block the megakaryocytic differentiation to some content. CONCLUSION This study implicated a regulatory role of KLF2 in megakaryocytic differentiation, which may suggest KLF2 as a target for illness with abnormal megakaryocytic differentiation.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Hematology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China.
- Henan University, Kaifeng, Henan, China.
- Zhengzhou University, Zhengzhou, Henan, China.
| | - Zhongwen Liu
- Department of Hematology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Henan University, Kaifeng, Henan, China
- Zhengzhou University, Zhengzhou, Henan, China
| | - Pan Zhou
- Department of Hematology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Xiaona Niu
- Department of Hematology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Henan University, Kaifeng, Henan, China
- Zhengzhou University, Zhengzhou, Henan, China
| | | | - Huan He
- Zhengzhou University, Zhengzhou, Henan, China
| | - Zunmin Zhu
- Department of Hematology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China.
- Henan University, Kaifeng, Henan, China.
- Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
13
|
Chen S, Looney MR. Understanding megakaryocyte phenotypes and the impact on platelet biogenesis. Transfusion 2024; 64:1372-1380. [PMID: 38923572 PMCID: PMC11251837 DOI: 10.1111/trf.17927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 06/28/2024]
Affiliation(s)
- Shiyu Chen
- Departments of Medicine and Laboratory Medicine, University
of California, San Francisco, San Francisco, CA, U.SA
| | - Mark R. Looney
- Departments of Medicine and Laboratory Medicine, University
of California, San Francisco, San Francisco, CA, U.SA
| |
Collapse
|
14
|
Donati M, Kazakov DV. Beyond typical histology of BAP1-inactivated melanocytoma. Pathol Res Pract 2024; 259:155162. [PMID: 38326181 DOI: 10.1016/j.prp.2024.155162] [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] [Received: 10/12/2023] [Revised: 01/05/2024] [Accepted: 01/20/2024] [Indexed: 02/09/2024]
Abstract
BAP1-inactivated melanocytoma (BIM) is a novel subgroup of melanocytic neoplasm listed in the 5th edition of WHO classification of skin tumor. BIM is characterized by two molecular alterations, including a mitogenic driver mutation (usually BRAF gene) and the loss of function of BAP1, a tumor suppressor gene located on chromosome 3p21, which encodes for BRCA1-associated protein (BAP1). The latter represents a nuclear-localized deubiquitinase involved in several cellular processes including cell cycle regulation, chromatin remodeling, DNA damage response, differentiation, senescence and cell death. BIMs are histologically characterized by a population of large epithelioid melanocytes with well-demarcated cytoplasmic borders and copious eosinophilic cytoplasm, demonstrating loss of BAP1 nuclear expression by immunohistochemistry. Recently, we have published a series of 50 cases, extending the morphological spectrum of the neoplasm and highlighting some new microscopic features. In the current article, we focus on some new histological features, attempting to explain and link them to certain mechanisms of tumor development, including senescence, endoreplication, endocycling, asymmetric cytokinesis, entosis and others. In light of the morphological and molecular findings observed in BIM, we postulated that this entity unmasks a fine mechanism of tumor in which both clonal/stochastic and hierarchical model can be unified.
Collapse
Affiliation(s)
- Michele Donati
- Department of Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Department of Pathology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21 - 00128 Roma, Italy.
| | - Dmitry V Kazakov
- IDP Dermatohistopathologie Institut, Pathologie Institut Enge, Zurich, Switzerland
| |
Collapse
|
15
|
Wang Y, Lv Y, Jiang X, Yu X, Wang D, Liu D, Liu X, Sun Y. Long non-coding RNA NORAD regulates megakaryocyte differentiation and proplatelet formation via the DUSP6/ERK signaling pathway. Biochem Biophys Res Commun 2024; 715:150004. [PMID: 38678784 DOI: 10.1016/j.bbrc.2024.150004] [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] [Received: 02/28/2024] [Revised: 04/14/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
Megakaryopoiesis and platelet production is a complex process that is underpotential regulation at multiple stages. Many long non-coding RNAs (lncRNAs) are distributed in hematopoietic stem cells and platelets. lncRNAs may play important roles as key epigenetic regulators in megakaryocyte differentiation and proplatelet formation. lncRNA NORAD can affect cell ploidy by sequestering PUMILIO proteins, although its direct effect on megakaryocyte differentiation and thrombopoiesis is still unknown. In this study, we demonstrate NORAD RNA is highly expressed in the cytoplasm during megakaryocyte differentiation. Interestingly, we identified for the first time that NORAD has a strong inhibitory effect on megakaryocyte differentiation and proplatelet formation from cultured megakaryocytes. DUSP6/ERK1/2 pathway is activated in response to NORAD knockdown during megakaryocytopoiesis, which is achieved by sequestering PUM2 proteins. Finally, compared with the wild-type control mice, NORAD knockout mice show a faster platelet recovery after severe thrombocytopenia induced by 6 Gy total body irradiation. These findings demonstrate lncRNA NORAD has a key role in regulating megakaryocyte differentiation and thrombopoiesis, which provides a promising molecular target for the treatment of platelet-related diseases such as severe thrombocytopenia.
Collapse
Affiliation(s)
- Yong Wang
- College of Pharmacy, Binzhou Medical University, China
| | - Yan Lv
- College of Life Science, Yantai University, China
| | - Xiaoli Jiang
- College of Pharmacy, Binzhou Medical University, China
| | - Xin Yu
- College of Pharmacy, Binzhou Medical University, China
| | - Delong Wang
- College of Pharmacy, Binzhou Medical University, China
| | - Desheng Liu
- College of Pharmacy, Binzhou Medical University, China
| | - Xiangyong Liu
- College of Pharmacy, Binzhou Medical University, China
| | - Yeying Sun
- College of Pharmacy, Binzhou Medical University, China.
| |
Collapse
|
16
|
Chen L, Liu J, Chen K, Su Y, Chen Y, Lei Y, Si J, Zhang J, Zhang Z, Zou W, Zhang X, Rondina MT, Wang QF, Li Y. SET domain containing 2 promotes megakaryocyte polyploidization and platelet generation through methylation of α-tubulin. J Thromb Haemost 2024; 22:1727-1741. [PMID: 38537781 DOI: 10.1016/j.jtha.2024.03.010] [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] [Received: 08/29/2023] [Revised: 02/23/2024] [Accepted: 03/12/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Megakaryocytes (MKs) are polyploid cells responsible for producing ∼1011 platelets daily in humans. Unraveling the mechanisms regulating megakaryopoiesis holds the promise for the production of clinical-grade platelets from stem cells, overcoming significant current limitations in platelet transfusion medicine. Previous work identified that loss of the epigenetic regulator SET domain containing 2 (SETD2) was associated with an increased platelet count in mice. However, the role of SETD2 in megakaryopoiesis remains unknown. OBJECTIVES Here, we examined how SETD2 regulated MK development and platelet production using complementary murine and human systems. METHODS We manipulated the expression of SETD2 in multiple in vitro and ex vivo models to assess the ploidy of MKs and the function of platelets. RESULTS The genetic ablation of Setd2 increased the number of high-ploidy bone marrow MKs. Peripheral platelet counts in Setd2 knockout mice were significantly increased ∼2-fold, and platelets exhibited normal size, morphology, and function. By knocking down and overexpressing SETD2 in ex vivo human cell systems, we demonstrated that SETD2 negatively regulated MK polyploidization by controlling methylation of α-tubulin, microtubule polymerization, and MK nuclear division. Small-molecule inactivation of SETD2 significantly increased the production of high-ploidy MKs and platelets from human-induced pluripotent stem cells and cord blood CD34+ cells. CONCLUSION These findings identify a previously unrecognized role for SETD2 in regulating megakaryopoiesis and highlight the potential of targeting SETD2 to increase platelet production from human cells for transfusion practices.
Collapse
Affiliation(s)
- Lei Chen
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jingkun Liu
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Kunying Chen
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yanxun Su
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yihe Chen
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ying Lei
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jia Si
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jie Zhang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zhaojun Zhang
- University of Chinese Academy of Sciences, Beijing, China; Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center of Bioinformation, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Weiguo Zou
- Shanghai Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China; State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Matthew T Rondina
- Departments of Internal Medicine and Pathology, Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA; Department of Internal Medicine and the Geriatric Research, Education, and Clinical Center, George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah, USA.
| | - Qian-Fei Wang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Yueying Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
17
|
Ghosh S, Choudhury D, Ghosh D, Mondal M, Singha D, Malakar P. Characterization of polyploidy in cancer: Current status and future perspectives. Int J Biol Macromol 2024; 268:131706. [PMID: 38643921 DOI: 10.1016/j.ijbiomac.2024.131706] [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] [Received: 11/22/2023] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Various cancers frequently exhibit polyploidy, observed in a condition where a cell possesses more than two sets of chromosomes, which is considered a hallmark of the disease. The state of polyploidy often leads to aneuploidy, where cells possess an abnormal number or structure of chromosomes. Recent studies suggest that oncogenes contribute to aneuploidy. This finding significantly underscores its impact on cancer. Cancer cells exposed to certain chemotherapeutic drugs tend to exhibit an increased incidence of polyploidy. This occurrence is strongly associated with several challenges in cancer treatment, including metastasis, resistance to chemotherapy and the recurrence of malignant tumors. Indeed, it poses a significant hurdle to achieve complete tumor eradication and effective cancer therapy. Recently, there has been a growing interest in the field of polyploidy related to cancer for developing effective anti-cancer therapies. Polyploid cancer cells confer both advantages and disadvantages to tumor pathogenicity. This review delineates the diverse characteristics of polyploid cells, elucidates the pivotal role of polyploidy in cancer, and explores the advantages and disadvantages it imparts to cancer cells, along with the current approaches tried in lab settings to target polyploid cells. Additionally, it considers experimental strategies aimed at addressing the outstanding questions within the realm of polyploidy in relation to cancer.
Collapse
Affiliation(s)
- Srijonee Ghosh
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute (RKMVERI), Kolkata, India
| | - Debopriya Choudhury
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute (RKMVERI), Kolkata, India
| | - Dhruba Ghosh
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute (RKMVERI), Kolkata, India
| | - Meghna Mondal
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute (RKMVERI), Kolkata, India
| | - Didhiti Singha
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute (RKMVERI), Kolkata, India
| | - Pushkar Malakar
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute (RKMVERI), Kolkata, India.
| |
Collapse
|
18
|
Feely C, Kaushal N, D’Avino PP, Martin J. Modifying platelets at their birth: anti-thrombotic therapy without haemorrhage. Front Pharmacol 2024; 15:1343896. [PMID: 38562457 PMCID: PMC10982340 DOI: 10.3389/fphar.2024.1343896] [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: 12/06/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024] Open
Abstract
Cardiovascular disease is a leading cause of death. The current approach to the prevention of arterial thrombosis in cardiovascular disease is dependent on the use of therapies which inhibit the activation of platelets. Predictably these are associated with an increased risk of haemorrhage which causes significant morbidity. The thrombotic potential of an activated platelet is modifiable; being determined before thrombopoiesis. Increased megakaryocyte ploidy is associated with larger and more active platelets carrying an increased risk of thrombosis. The reduction in the ploidy of megakaryocytes is therefore a novel area of therapeutic interest for reducing thrombosis. We propose a new therapeutic approach for the prevention and treatment of thrombosis by targeting the reduction in ploidy of megakaryocytes. We examine the role of a receptor mediated event causing megakaryocytes to increase ploidy, the potential for targeting the molecular mechanisms underpinning megakaryocyte endomitosis and the existence of two separate regulatory pathways to maintain haemostasis by altering the thrombotic potential of platelets as targets for novel therapeutic approaches producing haemostatically competent platelets which are not prothrombotic.
Collapse
Affiliation(s)
- Conor Feely
- Centre for Clinical Pharmacology, Institute of Health Informatics, University College London, London, United Kingdom
| | - Nitika Kaushal
- Centre for Clinical Pharmacology, Institute of Health Informatics, University College London, London, United Kingdom
| | - Pier Paolo D’Avino
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - John Martin
- Centre for Clinical Pharmacology, Institute of Health Informatics, University College London, London, United Kingdom
- Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
19
|
Lin GL, Chang HH, Lin WT, Liou YS, Lai YL, Hsieh MH, Chen PK, Liao CY, Tsai CC, Wang TF, Chu SC, Kau JH, Huang HH, Hsu HL, Sun DS. Dachshund Homolog 1: Unveiling Its Potential Role in Megakaryopoiesis and Bacillus anthracis Lethal Toxin-Induced Thrombocytopenia. Int J Mol Sci 2024; 25:3102. [PMID: 38542074 PMCID: PMC10970148 DOI: 10.3390/ijms25063102] [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: 01/26/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Lethal toxin (LT) is the critical virulence factor of Bacillus anthracis, the causative agent of anthrax. One common symptom observed in patients with anthrax is thrombocytopenia, which has also been observed in mice injected with LT. Our previous study demonstrated that LT induces thrombocytopenia by suppressing megakaryopoiesis, but the precise molecular mechanisms behind this phenomenon remain unknown. In this study, we utilized 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced megakaryocytic differentiation in human erythroleukemia (HEL) cells to identify genes involved in LT-induced megakaryocytic suppression. Through cDNA microarray analysis, we identified Dachshund homolog 1 (DACH1) as a gene that was upregulated upon TPA treatment but downregulated in the presence of TPA and LT, purified from the culture supernatants of B. anthracis. To investigate the function of DACH1 in megakaryocytic differentiation, we employed short hairpin RNA technology to knock down DACH1 expression in HEL cells and assessed its effect on differentiation. Our data revealed that the knockdown of DACH1 expression suppressed megakaryocytic differentiation, particularly in polyploidization. We demonstrated that one mechanism by which B. anthracis LT induces suppression of polyploidization in HEL cells is through the cleavage of MEK1/2. This cleavage results in the downregulation of the ERK signaling pathway, thereby suppressing DACH1 gene expression and inhibiting polyploidization. Additionally, we found that known megakaryopoiesis-related genes, such as FOSB, ZFP36L1, RUNX1, FLI1, AHR, and GFI1B genes may be positively regulated by DACH1. Furthermore, we observed an upregulation of DACH1 during in vitro differentiation of CD34-megakaryocytes and downregulation of DACH1 in patients with thrombocytopenia. In summary, our findings shed light on one of the molecular mechanisms behind LT-induced thrombocytopenia and unveil a previously unknown role for DACH1 in megakaryopoiesis.
Collapse
Affiliation(s)
- Guan-Ling Lin
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan; (G.-L.L.); (H.-H.C.); (P.-K.C.)
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan; (W.-T.L.); (Y.-S.L.); (Y.-L.L.); (M.-H.H.)
| | - Hsin-Hou Chang
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan; (G.-L.L.); (H.-H.C.); (P.-K.C.)
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan; (W.-T.L.); (Y.-S.L.); (Y.-L.L.); (M.-H.H.)
| | - Wei-Ting Lin
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan; (W.-T.L.); (Y.-S.L.); (Y.-L.L.); (M.-H.H.)
| | - Yu-Shan Liou
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan; (W.-T.L.); (Y.-S.L.); (Y.-L.L.); (M.-H.H.)
| | - Yi-Ling Lai
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan; (W.-T.L.); (Y.-S.L.); (Y.-L.L.); (M.-H.H.)
| | - Min-Hua Hsieh
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan; (W.-T.L.); (Y.-S.L.); (Y.-L.L.); (M.-H.H.)
| | - Po-Kong Chen
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan; (G.-L.L.); (H.-H.C.); (P.-K.C.)
| | - Chi-Yuan Liao
- Department of Obstetrics and Gynecology, Mennonite Christian Hospital, Hualien 97004, Taiwan; (C.-Y.L.); (C.-C.T.)
| | - Chi-Chih Tsai
- Department of Obstetrics and Gynecology, Mennonite Christian Hospital, Hualien 97004, Taiwan; (C.-Y.L.); (C.-C.T.)
| | - Tso-Fu Wang
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan; (T.-F.W.); (S.-C.C.)
- Department of Medicine, College of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Buddhist Tzu Chi Stem Cells Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Sung-Chao Chu
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan; (T.-F.W.); (S.-C.C.)
- Department of Medicine, College of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Buddhist Tzu Chi Stem Cells Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Jyh-Hwa Kau
- Institute of Preventive Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (J.-H.K.); (H.-H.H.); (H.-L.H.)
| | - Hsin-Hsien Huang
- Institute of Preventive Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (J.-H.K.); (H.-H.H.); (H.-L.H.)
| | - Hui-Ling Hsu
- Institute of Preventive Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (J.-H.K.); (H.-H.H.); (H.-L.H.)
| | - Der-Shan Sun
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan; (G.-L.L.); (H.-H.C.); (P.-K.C.)
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan; (W.-T.L.); (Y.-S.L.); (Y.-L.L.); (M.-H.H.)
| |
Collapse
|
20
|
Shin E, Park C, Park T, Chung H, Hwang H, Bak SH, Chung KS, Yoon SR, Kim TD, Choi I, Lee CH, Jung H, Noh JY. Deficiency of thioredoxin-interacting protein results in age-related thrombocytopenia due to megakaryocyte oxidative stress. J Thromb Haemost 2024; 22:834-850. [PMID: 38072375 DOI: 10.1016/j.jtha.2023.11.020] [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] [Received: 03/14/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Platelets are generated from megakaryocytes (MKs), mainly located in the bone marrow (BM). Megakaryopoiesis can be affected by genetic disorders, metabolic diseases, and aging. The molecular mechanisms underlying platelet count regulation have not been fully elucidated. OBJECTIVES In the present study, we investigated the role of thioredoxin-interacting protein (TXNIP), a protein that regulates cellular metabolism in megakaryopoiesis, using a Txnip-/- mouse model. METHODS Wild-type (WT) and Txnip-/- mice (2-27-month-old) were studied. BM-derived MKs were analyzed to investigate the role of TXNIP in megakaryopoiesis with age. The global transcriptome of BM-derived CD41+ megakaryocyte precursors (MkPs) of WT and Txnip-/- mice were compared. The CD34+ hematopoietic stem cells isolated from human cord blood were differentiated into MKs. RESULTS Txnip-/- mice developed thrombocytopenia at 4 to 5 months that worsened with age. During ex vivo megakaryopoiesis, Txnip-/- MkPs remained small, with decreased levels of MK-specific markers. Critically, Txnip-/- MkPs exhibited reduced mitochondrial reactive oxygen species, which was related to AKT activity. Txnip-/- MkPs also showed elevated glycolysis alongside increased glucose uptake for ATP production. Total RNA sequencing revealed enrichment for oxidative stress- and apoptosis-related genes in differentially expressed genes between Txnip-/- and WT MkPs. The effects of TXNIP on MKs were recapitulated during the differentiation of human cord blood-derived CD34+ hematopoietic stem cells. CONCLUSION We provide evidence that the megakaryopoiesis pathway becomes exhausted with age in Txnip-/- mice with a decrease in terminal, mature MKs that response to thrombocytopenic challenge. Overall, this study demonstrates the role of TXNIP in megakaryopoiesis, regulating mitochondrial metabolism.
Collapse
Affiliation(s)
- Eunju Shin
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, Korea; College of Pharmacy, Chungnam National University, Yuseong-gu, Daejeon, Korea
| | - Charny Park
- Bioinformatics Team, Research Institute, National Cancer Center, Ilsandong-gu, Gyeonggi-do, Korea
| | - Taeho Park
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, Korea; Department of Functional Genomics, Korea University of Science and Technology, Yuseong-gu, Daejeon, Korea
| | - Hyunmin Chung
- College of Pharmacy, Chungnam National University, Yuseong-gu, Daejeon, Korea; Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, Korea
| | - Hyeyeong Hwang
- Bioinformatics Team, Research Institute, National Cancer Center, Ilsandong-gu, Gyeonggi-do, Korea
| | - Seong Ho Bak
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, Korea; Department of Functional Genomics, Korea University of Science and Technology, Yuseong-gu, Daejeon, Korea
| | - Kyung-Sook Chung
- Department of Functional Genomics, Korea University of Science and Technology, Yuseong-gu, Daejeon, Korea; Stem Cell Convergence Research Center and Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, Korea
| | - Suk Ran Yoon
- Department of Functional Genomics, Korea University of Science and Technology, Yuseong-gu, Daejeon, Korea; Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, Korea
| | - Tae-Don Kim
- Department of Functional Genomics, Korea University of Science and Technology, Yuseong-gu, Daejeon, Korea; Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, Korea
| | - Inpyo Choi
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, Korea
| | - Chang Hoon Lee
- R&D Center, SCBIO Co, Ltd, Munji-ro, Yuseong-gu, Daejeon, Korea; Therapeutics and Biotechnology Division, Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon, Korea
| | - Haiyoung Jung
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, Korea; Department of Functional Genomics, Korea University of Science and Technology, Yuseong-gu, Daejeon, Korea
| | - Ji-Yoon Noh
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, Korea; Department of Functional Genomics, Korea University of Science and Technology, Yuseong-gu, Daejeon, Korea.
| |
Collapse
|
21
|
Wang Y, Tamori Y. Polyploid Cancer Cell Models in Drosophila. Genes (Basel) 2024; 15:96. [PMID: 38254985 PMCID: PMC10815460 DOI: 10.3390/genes15010096] [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: 11/06/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Cells with an abnormal number of chromosomes have been found in more than 90% of solid tumors, and among these, polyploidy accounts for about 40%. Polyploidized cells most often have duplicate centrosomes as well as genomes, and thus their mitosis tends to promote merotelic spindle attachments and chromosomal instability, which produces a variety of aneuploid daughter cells. Polyploid cells have been found highly resistant to various stress and anticancer therapies, such as radiation and mitogenic inhibitors. In other words, common cancer therapies kill proliferative diploid cells, which make up the majority of cancer tissues, while polyploid cells, which lurk in smaller numbers, may survive. The surviving polyploid cells, prompted by acute environmental changes, begin to mitose with chromosomal instability, leading to an explosion of genetic heterogeneity and a concomitant cell competition and adaptive evolution. The result is a recurrence of the cancer during which the tenacious cells that survived treatment express malignant traits. Although the presence of polyploid cells in cancer tissues has been observed for more than 150 years, the function and exact role of these cells in cancer progression has remained elusive. For this reason, there is currently no effective therapeutic treatment directed against polyploid cells. This is due in part to the lack of suitable experimental models, but recently several models have become available to study polyploid cells in vivo. We propose that the experimental models in Drosophila, for which genetic techniques are highly developed, could be very useful in deciphering mechanisms of polyploidy and its role in cancer progression.
Collapse
Affiliation(s)
| | - Yoichiro Tamori
- Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| |
Collapse
|
22
|
Ranjit S, Wang Y, Zhu J, Cheepala SB, Schuetz EG, Cho WJ, Xu B, Robinson CG, Wu G, Naren AP, Schuetz JD. ABCC4 impacts megakaryopoiesis and protects megakaryocytes against 6-mercaptopurine induced cytotoxicity. Drug Resist Updat 2024; 72:101017. [PMID: 37988981 PMCID: PMC10874622 DOI: 10.1016/j.drup.2023.101017] [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: 08/16/2023] [Revised: 10/21/2023] [Accepted: 11/06/2023] [Indexed: 11/23/2023]
Abstract
The role of ABCC4, an ATP-binding cassette transporter, in the process of platelet formation, megakaryopoiesis, is unknown. Here, we show that ABCC4 is highly expressed in megakaryocytes (MKs). Mining of public genomic data (ATAC-seq and genome wide chromatin interactions, Hi-C) revealed that key megakaryopoiesis transcription factors (TFs) interacted with ABCC4 regulatory elements and likely accounted for high ABCC4 expression in MKs. Importantly these genomic interactions for ABCC4 ranked higher than for genes with known roles in megakaryopoiesis suggesting a role for ABCC4 in megakaryopoiesis. We then demonstrate that ABCC4 is required for optimal platelet formation as in vitro differentiation of fetal liver derived MKs from Abcc4-/- mice exhibited impaired proplatelet formation and polyploidization, features required for optimal megakaryopoiesis. Likewise, a human megakaryoblastic cell line, MEG-01 showed that acute ABCC4 inhibition markedly suppressed key processes in megakaryopoiesis and that these effects were related to reduced cAMP export and enhanced dissociation of a negative regulator of megakaryopoiesis, protein kinase A (PKA) from ABCC4. PKA activity concomitantly increased after ABCC4 inhibition which was coupled with significantly reduced GATA-1 expression, a TF needed for optimal megakaryopoiesis. Further, ABCC4 protected MKs from 6-mercaptopurine (6-MP) as Abcc4-/- mice show a profound reduction in MKs after 6-MP treatment. In total, our studies show that ABCC4 not only protects the MKs but is also required for maximal platelet production from MKs, suggesting modulation of ABCC4 function might be a potential therapeutic strategy to regulate platelet production.
Collapse
Affiliation(s)
- Sabina Ranjit
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Childres's Research Hospital, USA
| | - Yao Wang
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Childres's Research Hospital, USA
| | - Jingwen Zhu
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Childres's Research Hospital, USA; Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Satish B Cheepala
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Childres's Research Hospital, USA
| | - Erin G Schuetz
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Childres's Research Hospital, USA
| | - Woo Jung Cho
- Cell and Tissue Imaging Center, St Jude Children's Research Hospital, USA
| | - Beisi Xu
- Center for Applied Bioinformatics, St Jude Children's Research Hospital, USA
| | | | - Gang Wu
- Center for Applied Bioinformatics, St Jude Children's Research Hospital, USA
| | - Anjaparavanda P Naren
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - John D Schuetz
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Childres's Research Hospital, USA.
| |
Collapse
|
23
|
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
|
24
|
Yalcin BH, Macas J, Wiercinska E, Harter PN, Fawaz M, Schmachtel T, Ghiro I, Bieniek E, Kosanovic D, Thom S, Fruttiger M, Taketo MM, Schermuly RT, Rieger MA, Plate KH, Bonig H, Liebner S. Wnt/β-Catenin-Signaling Modulates Megakaryopoiesis at the Megakaryocyte-Erythrocyte Progenitor Stage in the Hematopoietic System. Cells 2023; 12:2765. [PMID: 38067194 PMCID: PMC10706863 DOI: 10.3390/cells12232765] [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: 09/11/2023] [Revised: 11/20/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
The bone marrow (BM) hematopoietic system (HS) gives rise to blood cells originating from hematopoietic stem cells (HSCs), including megakaryocytes (MKs) and red blood cells (erythrocytes; RBCs). Many steps of the cell-fate decision remain to be elucidated, being important for cancer treatment. To explore the role of Wnt/β-catenin for MK and RBC differentiation, we activated β-catenin signaling in platelet-derived growth factor b (Pdgfb)-expressing cells of the HS using a Cre-lox approach (Ctnnb1BM-GOF). FACS analysis revealed that Pdgfb is mainly expressed by megakaryocytic progenitors (MKPs), MKs and platelets. Recombination resulted in a lethal phenotype in mutants (Ctnnb1BM-GOFwt/fl, Ctnnb1BM-GOFfl/fl) 3 weeks after tamoxifen injection, showing an increase in MKs in the BM and spleen, but no pronounced anemia despite reduced erythrocyte counts. BM transplantation (BMT) of Ctnnb1BM-GOF BM into lethally irradiated wildtype recipients (BMT-Ctnnb1BM-GOF) confirmed the megakaryocytic, but not the lethal phenotype. CFU-MK assays in vitro with BM cells of Ctnnb1BM-GOF mice supported MK skewing at the expense of erythroid colonies. Molecularly, the runt-related transcription factor 1 (RUNX1) mRNA, known to suppress erythropoiesis, was upregulated in Ctnnb1BM-GOF BM cells. In conclusion, β-catenin activation plays a key role in cell-fate decision favoring MK development at the expense of erythroid production.
Collapse
Affiliation(s)
- Burak H. Yalcin
- Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany (J.M.); (I.G.); (K.H.P.)
| | - Jadranka Macas
- Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany (J.M.); (I.G.); (K.H.P.)
| | - Eliza Wiercinska
- Institute for Transfusion Medicine and Immunohaematology, and DRK-Blutspendedienst BaWüHe, Goethe University Frankfurt, 60528 Frankfurt am Main, Germany
| | - Patrick N. Harter
- Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany (J.M.); (I.G.); (K.H.P.)
| | - Malak Fawaz
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany (M.A.R.)
| | - Tessa Schmachtel
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany (M.A.R.)
| | - Ilaria Ghiro
- Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany (J.M.); (I.G.); (K.H.P.)
| | - Ewa Bieniek
- German Center for Lung Research (DZL), Department of Internal Medicine, Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (E.B.); (D.K.)
| | - Djuro Kosanovic
- German Center for Lung Research (DZL), Department of Internal Medicine, Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (E.B.); (D.K.)
| | - Sonja Thom
- Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany (J.M.); (I.G.); (K.H.P.)
| | | | - Makoto M. Taketo
- Kyoto University Hospital-iACT Graduate School of Medicine, Kyoto University, Kyoto 06-8501, Japan
| | - Ralph T. Schermuly
- German Center for Lung Research (DZL), Department of Internal Medicine, Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (E.B.); (D.K.)
| | - Michael A. Rieger
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany (M.A.R.)
- German Cancer Consortium (DKTK) at the German Cancer Research Center, 69120 Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Partner Site Frankfurt, 60590 Frankfurt am Main, Germany
| | - Karl H. Plate
- Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany (J.M.); (I.G.); (K.H.P.)
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohaematology, and DRK-Blutspendedienst BaWüHe, Goethe University Frankfurt, 60528 Frankfurt am Main, Germany
- Department of Medicine/Division of Hematology, University of Washington, Seattle, WA 98195, USA
| | - Stefan Liebner
- Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany (J.M.); (I.G.); (K.H.P.)
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Partner Site Frankfurt, 60590 Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
| |
Collapse
|
25
|
Vittoria MA, Quinton RJ, Ganem NJ. Whole-genome doubling in tissues and tumors. Trends Genet 2023; 39:954-967. [PMID: 37714734 PMCID: PMC10840902 DOI: 10.1016/j.tig.2023.08.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/17/2023]
Abstract
The overwhelming majority of proliferating somatic human cells are diploid, and this genomic state is typically maintained across successive cell divisions. However, failures in cell division can induce a whole-genome doubling (WGD) event, in which diploid cells transition to a tetraploid state. While some WGDs are developmentally programmed to produce nonproliferative tetraploid cells with specific cellular functions, unscheduled WGDs can be catastrophic: erroneously arising tetraploid cells are ill-equipped to cope with their doubled cellular and chromosomal content and quickly become genomically unstable and tumorigenic. Deciphering the genetics that underlie the genesis, physiology, and evolution of whole-genome doubled (WGD+) cells may therefore reveal therapeutic avenues to selectively eliminate pathological WGD+ cells.
Collapse
Affiliation(s)
- Marc A Vittoria
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Ryan J Quinton
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Neil J Ganem
- Department of Medicine, Division of Hematology and Oncology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA; Department of Pharmacology, Physiology, and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
26
|
Thanasegaran S, Daimon E, Shibukawa Y, Yamazaki N, Okamoto N. Modelling Takenouchi-Kosaki syndrome using disease-specific iPSCs. Stem Cell Res 2023; 73:103221. [PMID: 37918315 DOI: 10.1016/j.scr.2023.103221] [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] [Received: 05/01/2023] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
Takenouchi-Kosaki Syndrome (TKS) is a congenital multi-organ disorder caused by the de novo missense mutation c.191A > G p. Tyr64Cys (Y64C) in the CDC42 gene. We previously elucidated the functional abnormalities and thrombopoietic effects of Y64C using HEK293 and MEG01 cells. In the present study, we used iPSCs derived from TKS patients to model the disease and successfully recapitulated macrothrombocytopenia, a prominent TKS phenotype. The megakaryopoietic differentiation potential of TKS-iPSCs and platelet production capacity were examined using an efficient platelet production method redesigned from existing protocols. The results obtained showed that TKS-iPSCs produced fewer hematopoietic progenitor cells, exhibited defective megakaryopoiesis, and released platelets with an abnormally low count and giant morphology. We herein report the first analysis of TKS-iPSC-derived megakaryocytes and platelets, and currently utilize this model to perform drug evaluations for TKS. Therefore, our simple yet effective differentiation method, which mimics the disease in a dish, is a feasible strategy for studying hematopoiesis and related diseases.
Collapse
Affiliation(s)
- Suganya Thanasegaran
- Department of Molecular Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Etsuko Daimon
- Department of Molecular Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Yukinao Shibukawa
- Department of Molecular Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Natsuko Yamazaki
- Department of Molecular Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Nobuhiko Okamoto
- Department of Molecular Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan.
| |
Collapse
|
27
|
Liu H, Ishikawa-Ankerhold H, Winterhalter J, Lorenz M, Vladymyrov M, Massberg S, Schulz C, Orban M. Multiphoton In Vivo Microscopy of Embryonic Thrombopoiesis Reveals the Generation of Platelets through Budding. Cells 2023; 12:2411. [PMID: 37830625 PMCID: PMC10572188 DOI: 10.3390/cells12192411] [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] [Received: 07/26/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023] Open
Abstract
Platelets are generated by specialized cells called megakaryocytes (MKs). However, MK's origin and platelet release mode have remained incompletely understood. Here, we established direct visualization of embryonic thrombopoiesis in vivo by combining multiphoton intravital microscopy (MP-IVM) with a fluorescence switch reporter mouse model under control of the platelet factor 4 promoter (Pf4CreRosa26mTmG). Using this microscopy tool, we discovered that fetal liver MKs provide higher thrombopoietic activity than yolk sac MKs. Mechanistically, fetal platelets were released from MKs either by membrane buds or the formation of proplatelets, with the former constituting the key process. In E14.5 c-Myb-deficient embryos that lack definitive hematopoiesis, MK and platelet numbers were similar to wild-type embryos, indicating the independence of embryonic thrombopoiesis from definitive hematopoiesis at this stage of development. In summary, our novel MP-IVM protocol allows the characterization of thrombopoiesis with high spatio-temporal resolution in the mouse embryo and has identified membrane budding as the main mechanism of fetal platelet production.
Collapse
Affiliation(s)
- Huan Liu
- Department of Internal Medicine I, Ludwig Maximilians University, 81377 Munich, Germany; (H.L.); (H.I.-A.); (J.W.); (M.L.); (S.M.)
| | - Hellen Ishikawa-Ankerhold
- Department of Internal Medicine I, Ludwig Maximilians University, 81377 Munich, Germany; (H.L.); (H.I.-A.); (J.W.); (M.L.); (S.M.)
| | - Julia Winterhalter
- Department of Internal Medicine I, Ludwig Maximilians University, 81377 Munich, Germany; (H.L.); (H.I.-A.); (J.W.); (M.L.); (S.M.)
| | - Michael Lorenz
- Department of Internal Medicine I, Ludwig Maximilians University, 81377 Munich, Germany; (H.L.); (H.I.-A.); (J.W.); (M.L.); (S.M.)
| | - Mykhailo Vladymyrov
- Laboratory for High Energy Physics (LHEP), Albert Einstein Center for Fundamental Physics, University of Bern, 3012 Bern, Switzerland;
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
- Data Science Lab, Mathematical Institute, University of Bern, 3012 Bern, Switzerland
| | - Steffen Massberg
- Department of Internal Medicine I, Ludwig Maximilians University, 81377 Munich, Germany; (H.L.); (H.I.-A.); (J.W.); (M.L.); (S.M.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80802 Munich, Germany
| | - Christian Schulz
- Department of Internal Medicine I, Ludwig Maximilians University, 81377 Munich, Germany; (H.L.); (H.I.-A.); (J.W.); (M.L.); (S.M.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80802 Munich, Germany
| | - Mathias Orban
- Department of Internal Medicine I, Ludwig Maximilians University, 81377 Munich, Germany; (H.L.); (H.I.-A.); (J.W.); (M.L.); (S.M.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80802 Munich, Germany
| |
Collapse
|
28
|
Nagai K, Niihori T, Muto A, Hayashi Y, Abe T, Igarashi K, Aoki Y. Mecom mutation related to radioulnar synostosis with amegakaryocytic thrombocytopenia reduces HSPCs in mice. Blood Adv 2023; 7:5409-5420. [PMID: 37099686 PMCID: PMC10509669 DOI: 10.1182/bloodadvances.2022008462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 04/28/2023] Open
Abstract
Radioulnar synostosis with amegakaryocytic thrombocytopenia (RUSAT) is an inherited bone marrow failure syndrome characterized by the congenital fusion of the forearm bones. RUSAT is largely caused by missense mutations that are clustered in a specific region of the MDS1 and EVI1 complex locus (MECOM). EVI1, a transcript variant encoded by MECOM, is a zinc finger transcription factor involved in hematopoietic stem cell maintenance that induce leukemic transformation when overexpressed. Mice with exonic deletions in Mecom show reduced hematopoietic stem and progenitor cells (HSPCs). However, the pathogenic roles of RUSAT-associated MECOM mutations in vivo have not yet been elucidated. To investigate the impact of the RUSAT-associated MECOM mutation on the phenotype, we generated knockin mice harboring a point mutation (translated into EVI1 p.H752R and MDS1-EVI1 p.H942R), which corresponds to an EVI1 p.H751R and MDS1-EVI1 p.H939R mutation identified in a patient with RUSAT. Homozygous mutant mice died at embryonic day 10.5 to 11.5. Heterozygous mutant mice (Evi1KI/+ mice) grew normally without radioulnar synostosis. Male Evi1KI/+ mice, aged between 5 and 15 weeks, exhibited lower body weight, and those aged ≥16 weeks showed low platelet counts. Flow cytometric analysis of bone marrow cells revealed a decrease in HSPCs in Evi1KI/+ mice between 8 and 12 weeks. Moreover, Evi1KI/+ mice showed delayed leukocyte and platelet recovery after 5-fluorouracil-induced myelosuppression. These findings suggest that Evi1KI/+ mice recapitulate the bone marrow dysfunction in RUSAT, similar to that caused by loss-of-function Mecom alleles.
Collapse
Affiliation(s)
- Koki Nagai
- Department of Medical Genetics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuya Niihori
- Department of Medical Genetics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akihiko Muto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshikazu Hayashi
- Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Taiki Abe
- Department of Medical Genetics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoko Aoki
- Department of Medical Genetics, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
29
|
Sun Z, Wang B, Shen Y, Ma K, Wang T, Wang Y, Lin D. MXRA7 is involved in megakaryocyte differentiation and platelet production. BLOOD SCIENCE 2023; 5:160-169. [PMID: 37546710 PMCID: PMC10400050 DOI: 10.1097/bs9.0000000000000167] [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: 01/08/2023] [Accepted: 06/16/2023] [Indexed: 08/08/2023] Open
Abstract
Matrix remodeling is a critical process in hematopoiesis. The biology of MXRA7, as a matrix remodeling associated gene, has still not been reported in hematopoietic process. Public databases showed that MXRA7 expressed in hematopoietic stem cells, suggesting that it may be involved in hematopoiesis. We found that the amounts of megakaryocytes were lower in bone marrow and spleen from Mxra7-/- mice compared with that from wild-type mice. Knock-out of MXRA7 also reduced the amount of platelet in peripheral blood and affected the function of platelets. Knock-out of MXRA7 inhibited hematopoietic stem/progenitor cells differentiate to megakaryocytes possibly through down-regulating the expression of GATA-1 and FOG-1. Moreover, knockdown of MXRA7 in MEG-01 cells could inhibit the cell proliferation and cell apoptosis. Knockdown of MXRA7 inhibited the differentiation of MEG-01 cells and proplatelet formation through suppressing the ERK/MAPK signaling pathway and the expression of β-tubulin. In conclusion, the current study demonstrated the potential significance of MXRA7 in megakaryocyte differentiation and platelet production. The novel findings proposed a new target for the treatment of platelet-related diseases, and much more investigations are guaranteed to dissect the mechanisms of MXRA7 in megakaryocyte differentiation and platelet production.
Collapse
Affiliation(s)
- Zhenjiang Sun
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Benfang Wang
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou 215006, China
- Department of Clinical Laboratory, The Affiliated Jiangyin Hospital of Southeast University, Jiangyin 214400, China
| | - Ying Shen
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Kunpeng Ma
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Ting Wang
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Yiqiang Wang
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Dandan Lin
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou 215006, China
| |
Collapse
|
30
|
Garyn CM, Bover O, Murray JW, Jing M, Salas-Briceno K, Ross SR, Snoeck HW. DNA damage primes hematopoietic stem cells for direct megakaryopoiesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.13.540665. [PMID: 37333356 PMCID: PMC10274687 DOI: 10.1101/2023.05.13.540665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Hematopoietic stem cells (HSCs) reside in the bone marrow (BM), can self-renew, and generate all cells of the hematopoietic system. 1 Most hematopoietic lineages arise through successive, increasingly lineage-committed progenitors. In contrast, megakaryocytes (MKs), hyperploid cells that generate platelets essential to hemostasis, can derive rapidly and directly from HSCs. 2 The underlying mechanism is unknown however. Here we show that DNA damage and subsequent arrest in the G2 phase of the cell cycle rapidly induce MK commitment specifically in HSCs, but not in progenitors, through an initially predominantly post-transcriptional mechanism. Cycling HSCs show extensive replication-induced DNA damage associated with uracil misincorporation in vivo and in vitro . Consistent with this notion, thymidine attenuated DNA damage, rescued HSC maintenance and reduced the generation of CD41 + MK-committed HSCs in vitro . Similarly, overexpression of the dUTP-scavenging enzyme, dUTPase, enhanced in vitro maintenance of HSCs. We conclude that a DNA damage response drives direct megakaryopoiesis and that replication stress-induced direct megakaryopoiesis, at least in part caused by uracil misincorporation, is a barrier to HSC maintenance in vitro . DNA damage-induced direct megakaryopoiesis may allow rapid generation of a lineage essential to immediate organismal survival, while simultaneously removing damaged HSCs and potentially avoiding malignant transformation of self-renewing stem cells.
Collapse
|
31
|
Schvarzstein M, Alam F, Toure M, Yanowitz JL. An Emerging Animal Model for Querying the Role of Whole Genome Duplication in Development, Evolution, and Disease. J Dev Biol 2023; 11:26. [PMID: 37367480 PMCID: PMC10299280 DOI: 10.3390/jdb11020026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/23/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Whole genome duplication (WGD) or polyploidization can occur at the cellular, tissue, and organismal levels. At the cellular level, tetraploidization has been proposed as a driver of aneuploidy and genome instability and correlates strongly with cancer progression, metastasis, and the development of drug resistance. WGD is also a key developmental strategy for regulating cell size, metabolism, and cellular function. In specific tissues, WGD is involved in normal development (e.g., organogenesis), tissue homeostasis, wound healing, and regeneration. At the organismal level, WGD propels evolutionary processes such as adaptation, speciation, and crop domestication. An essential strategy to further our understanding of the mechanisms promoting WGD and its effects is to compare isogenic strains that differ only in their ploidy. Caenorhabditis elegans (C. elegans) is emerging as an animal model for these comparisons, in part because relatively stable and fertile tetraploid strains can be produced rapidly from nearly any diploid strain. Here, we review the use of Caenorhabditis polyploids as tools to understand important developmental processes (e.g., sex determination, dosage compensation, and allometric relationships) and cellular processes (e.g., cell cycle regulation and chromosome dynamics during meiosis). We also discuss how the unique characteristics of the C. elegans WGD model will enable significant advances in our understanding of the mechanisms of polyploidization and its role in development and disease.
Collapse
Affiliation(s)
- Mara Schvarzstein
- Biology Department, Brooklyn College at the City University of New York, Brooklyn, NY 11210, USA
- Biology Department, The Graduate Center at the City University of New York, New York, NY 10016, USA
- Biochemistry Department, The Graduate Center at the City University of New York, New York, NY 10016, USA
| | - Fatema Alam
- Biology Department, Brooklyn College at the City University of New York, Brooklyn, NY 11210, USA
| | - Muhammad Toure
- Biology Department, Brooklyn College at the City University of New York, Brooklyn, NY 11210, USA
| | - Judith L. Yanowitz
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA;
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
32
|
Martin JF, D'Avino PP. A theory of rapid evolutionary change explaining the de novo appearance of megakaryocytes and platelets in mammals. J Cell Sci 2022; 135:285954. [PMID: 36515566 PMCID: PMC10112974 DOI: 10.1242/jcs.260286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Platelets are found only in mammals. Uniquely, they have a log Gaussian volume distribution and are produced from megakaryocytes, large cells that have polyploid nuclei. In this Hypothesis, we propose that a possible explanation for the origin of megakaryocytes and platelets is that, ∼220 million years ago, an inheritable change occurred in a mammalian ancestor that caused the haemostatic cell line of the animal to become polyploid. This inheritable change occurred specifically in the genetic programme of the cell lineage from which the haemostatic cell originated and led, because of increase in cell size, to its fragmentation into cytoplasmic particles (platelets) in the pulmonary circulatory system, as found in modern mammals. We hypothesize that these fragments originating from the new large haemostatic polyploid cells proved to be more efficient at stopping bleeding, and, therefore, the progeny of this ancestor prospered through natural selection. We also propose experimental strategies that could provide evidence to support this hypothesis.
Collapse
Affiliation(s)
- John F Martin
- Division of Medicine, University College London, 5 University Street, London WC1E 6JF, UK
| | - Paolo Pier D'Avino
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| |
Collapse
|
33
|
Qin J, Zhang J, Jiang J, Zhang B, Li J, Lin X, Wang S, Zhu M, Fan Z, Lv Y, He L, Chen L, Yue W, Li Y, Pei X. Direct chemical reprogramming of human cord blood erythroblasts to induced megakaryocytes that produce platelets. Cell Stem Cell 2022; 29:1229-1245.e7. [PMID: 35931032 DOI: 10.1016/j.stem.2022.07.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/08/2022] [Accepted: 07/13/2022] [Indexed: 11/19/2022]
Abstract
Reprogramming somatic cells into megakaryocytes (MKs) would provide a promising source of platelets. However, using a pharmacological approach to generate human MKs from somatic cells remains an unmet challenge. Here, we report that a combination of four small molecules (4M) successfully converted human cord blood erythroblasts (EBs) into induced MKs (iMKs). The iMKs could produce proplatelets and release functional platelets, functionally resembling natural MKs. Reprogramming trajectory analysis revealed an efficient cell fate conversion of EBs into iMKs by 4M via the intermediate state of bipotent precursors. 4M induced chromatin remodeling and drove the transition of transcription factor (TF) regulatory network from key erythroid TFs to essential TFs for megakaryopoiesis, including FLI1 and MEIS1. These results demonstrate that the chemical reprogramming of cord blood EBs into iMKs provides a simple and efficient approach to generate MKs and platelets for clinical applications.
Collapse
Affiliation(s)
- Jinhua Qin
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China
| | - Jian Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Jianan Jiang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bowen Zhang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China
| | - Jisheng Li
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiaosong Lin
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Sihan Wang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China
| | - Meiqi Zhu
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zeng Fan
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China
| | - Yang Lv
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China
| | - Lijuan He
- South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China; Institute of Health Service and Transfusion Medicine, Beijing 100850, China
| | - Lin Chen
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China
| | - Wen Yue
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China
| | - Yanhua Li
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China.
| | - Xuetao Pei
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China.
| |
Collapse
|
34
|
Duchmann M, Wagner-Ballon O, Boyer T, Cheok M, Fournier E, Guerin E, Fenwarth L, Badaoui B, Freynet N, Benayoun E, Lusina D, Garcia I, Gardin C, Fenaux P, Pautas C, Quesnel B, Turlure P, Terré C, Thomas X, Lambert J, Renneville A, Preudhomme C, Dombret H, Itzykson R, Cluzeau T. Machine learning identifies the independent role of dysplasia in the prediction of response to chemotherapy in AML. Leukemia 2022; 36:656-663. [PMID: 34615986 DOI: 10.1038/s41375-021-01435-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/17/2022]
Abstract
The independent prognostic impact of specific dysplastic features in acute myeloid leukemia (AML) remains controversial and may vary between genomic subtypes. We apply a machine learning framework to dissect the relative contribution of centrally reviewed dysplastic features and oncogenetics in 190 patients with de novo AML treated in ALFA clinical trials. One hundred and thirty-five (71%) patients achieved complete response after the first induction course (CR). Dysgranulopoiesis, dyserythropoiesis and dysmegakaryopoiesis were assessable in 84%, 83% and 63% patients, respectively. Multi-lineage dysplasia was present in 27% of assessable patients. Micromegakaryocytes (q = 0.01), hypolobulated megakaryocytes (q = 0.08) and hyposegmented granulocytes (q = 0.08) were associated with higher ELN-2017 risk. Using a supervised learning algorithm, the relative importance of morphological variables (34%) for the prediction of CR was higher than demographic (5%), clinical (2%), cytogenetic (25%), molecular (29%), and treatment (5%) variables. Though dysplasias had limited predictive impact on survival, a multivariate logistic regression identified the presence of hypolobulated megakaryocytes (p = 0.014) and micromegakaryocytes (p = 0.035) as predicting lower CR rates, independently of monosomy 7 (p = 0.013), TP53 (p = 0.004), and NPM1 mutations (p = 0.025). Assessment of these specific dysmegakarypoiesis traits, for which we identify a transcriptomic signature, may thus guide treatment allocation in AML.
Collapse
Affiliation(s)
- Matthieu Duchmann
- Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, France.,Université de Paris, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, 75010, Paris, France
| | - Orianne Wagner-Ballon
- Département d'Hématologie et Immunologie biologiques, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Créteil, France.,INSERM U955 IMRB, UPEC, Créteil, France
| | - Thomas Boyer
- Service d'Hématologie Biologique, CHU Lille, Lille, France.,Service d'Hématologie Biologique, CHU Amiens-Picardie, Amiens, France
| | | | - Elise Fournier
- Service d'Hématologie Biologique, CHU Lille, Lille, France
| | - Estelle Guerin
- Service d'Hématologie biologique, Hôpital Dupuytren, Limoges, France.,UMR CNRS 7276/INSERM 1262, CHU Limoges, Limoges, France
| | - Laurène Fenwarth
- Université Lille, CNRS, INSERM, CHU Lille, IRCL, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000, Lille, France
| | - Bouchra Badaoui
- Département d'Hématologie et Immunologie biologiques, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Créteil, France
| | - Nicolas Freynet
- Département d'Hématologie et Immunologie biologiques, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Créteil, France
| | - Emmanuel Benayoun
- Département d'Hématologie et Immunologie biologiques, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Créteil, France
| | - Daniel Lusina
- Laboratoire d'Hématologie, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris, Université Sorbonne Paris Cité, Bobigny, France
| | - Isabel Garcia
- Laboratoire d'Hématologie, Hôpital André Mignot, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Claude Gardin
- Département d'Hématologie Clinique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris, Bobigny, France
| | - Pierre Fenaux
- Département d'Hématologie Clinique, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, France
| | - Cécile Pautas
- Département d'Hématologie clinique, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Créteil, France
| | - Bruno Quesnel
- CHU Lille, Service des Maladies du Sang, 59000, Lille, France
| | - Pascal Turlure
- Département d'Hématologie Clinique, CHU Limoges, Limoges, France
| | - Christine Terré
- Laboratoire de Cytogénétique, Hôpital André Mignot, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Xavier Thomas
- Département d'Hématologie Clinique, Hospices Civils de Lyon, Hôpital Lyon-Sud, Pierre Bénite, France
| | - Juliette Lambert
- Département d'Hématologie Clinique, Hôpital André Mignot, Centre Hospitalier de Versailles, Le Chesnay, France
| | | | - Claude Preudhomme
- Université Lille, CNRS, INSERM, CHU Lille, IRCL, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000, Lille, France
| | - Hervé Dombret
- Département d'Hématologie Clinique, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, EA-3518, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Raphael Itzykson
- Université de Paris, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, 75010, Paris, France. .,Département d'Hématologie Clinique, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, France.
| | - Thomas Cluzeau
- Département d'Hématologie, Université Côte d'Azur, CHU de Nice, Nice, France.
| |
Collapse
|
35
|
Erratum: Ultrastructural alterations of megakaryocytes in thrombocytopenia: A review of 43 cases. BLOOD SCIENCE 2022; 3:107-112. [PMID: 35402843 PMCID: PMC8975046 DOI: 10.1097/bs9.0000000000000093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/29/2021] [Indexed: 11/25/2022] Open
Abstract
Thrombocytopenia is a frequent occurrence in a variety of hematopoietic diseases; however, the details of the mechanism leading to low platelet count remain elusive. Megakaryocytes are a series of progenitor cells responsible for the production of platelets. Alterations in megakaryocytes in the bone marrow are a causative factor resulting in thrombocytopenia in varied diseases. Based on ultrastructural analysis of incidentally encountered megakaryocytes in 43 patients with blood diseases marked by low platelet counts, electron micrographs demonstrated that aberrant megakaryocytes predominated in idiopathic thrombocytopenic purpura, aplastic anemia, and myelodysplastic syndrome; autophagy, apoptosis, and cellular damage in megakaryocytes were a prominent feature in aplastic anemia. On the other hand, poorly differentiated megakaryocytes predominated in acute megakaryoblastic leukemia (AMKL) although damaged megakaryocytes were seen in non-AMKL acute leukemia. This paper documents the ultrastructural alterations of megakaryocytes associated with thrombocytopenia and reveals distinctive features for particular blood diseases. A comment is made on future avenues of research emphasizing membrane fusion proteins.
Collapse
|
36
|
Charney E. The "Golden Age" of Behavior Genetics? PERSPECTIVES ON PSYCHOLOGICAL SCIENCE 2022; 17:1188-1210. [PMID: 35180032 DOI: 10.1177/17456916211041602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The search for genetic risk factors underlying the presumed heritability of all human behavior has unfolded in two phases. The first phase, characterized by candidate-gene-association (CGA) studies, has fallen out of favor in the behavior-genetics community, so much so that it has been referred to as a "cautionary tale." The second and current iteration is characterized by genome-wide association studies (GWASs), single-nucleotide polymorphism (SNP) heritability estimates, and polygenic risk scores. This research is guided by the resurrection of, or reemphasis on, Fisher's "infinite infinitesimal allele" model of the heritability of complex phenotypes, first proposed over 100 years ago. Despite seemingly significant differences between the two iterations, they are united in viewing the discovery of risk alleles underlying heritability as a matter of finding differences in allele frequencies. Many of the infirmities that beset CGA studies persist in the era of GWASs, accompanied by a host of new difficulties due to the human genome's underlying complexities and the limitations of Fisher's model in the postgenomics era.
Collapse
Affiliation(s)
- Evan Charney
- The Samuel DuBois Cook Center on Social Equity, Duke University
| |
Collapse
|
37
|
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
|
38
|
Saumell S, Fernández-Serrano M, Mesa A, López-Cadenas F, Arenillas L, Alfonso A, Montoro MJ, Molero A, Leoz P, Riego V, Gallur L, Salamero O, Navarrete M, Tazón-Vega B, Ortega M, Reig Ò, Roué G, Calvo X, Prosper F, Díez-Campelo M, Valcárcel D. Prognostic impact of micromegakaryocytes in primary myelodysplastic syndromes. Leuk Lymphoma 2021; 63:1227-1235. [PMID: 34969346 DOI: 10.1080/10428194.2021.2018581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Micromegakaryocytes (microMKs) are considered a myelodysplastic feature of myeloid neoplasms in adults, with an adverse prognosis connotation. However, this notion in MDS has not been well proved. In our cohort of 287 MDS, patients with microMKs showed lower overall survival (OS) (HR, 2.12; 95% CI, 1.47-3.06; p = 0.000036) and higher risk of acute myeloid leukemia (AML) evolution (HR, 4.8; 95% CI, 2.9-11.01; p = 0.00021). Results were validated with an independent cohort. In multivariate analysis, the presence of microMKs maintained its independent association with OS (HR, 1.54, 95% CI, 1.13-2.1, p = 0.0059) and AML transformation (HR, 2.28, 95% CI, 1.2-4.4, p = 0.014). Moreover, by adding 1 point to the IPSS-R score in patients with microMKs, we improved the IPSS-R accuracy. Interestingly, adding that 1-point, 29% of intermediate IPSS-R risk group patients were upgraded to the high-risk group. In summary, we confirmed that the presence of microMKs implies worse outcomes in MDS and suggested a modification improving IPSS-R.
Collapse
Affiliation(s)
- Sílvia Saumell
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Barcelona, Spain
| | - Miranda Fernández-Serrano
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Alba Mesa
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Barcelona, Spain
| | - Félix López-Cadenas
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Leonor Arenillas
- Laboratory of Cytology, Department of Pathology, GRETNHE, IMIM Hospital del Mar Research Institute, Barcelona, Spain
| | - Ana Alfonso
- Clínica Universidad de Navarra, Universidad de Navarra, Pamplona, Spain
| | - Maria Julia Montoro
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Barcelona, Spain
| | - Antonieta Molero
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Barcelona, Spain
| | - Pilar Leoz
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Victoria Riego
- Clínica Universidad de Navarra, Universidad de Navarra, Pamplona, Spain
| | - Laura Gallur
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Barcelona, Spain
| | - Olga Salamero
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Barcelona, Spain
| | - Mayda Navarrete
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Barcelona, Spain
| | - Bárbara Tazón-Vega
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Barcelona, Spain
| | - Margarita Ortega
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Barcelona, Spain
| | - Òscar Reig
- Medical Oncology Department, Hospital Clínic, Translational Genomics and Targeted Therapeutics in Solid Tumors Group (IDIBAPS), Barcelona, Spain
| | - Gaël Roué
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Barcelona, Spain.,Lymphoma Translational Group, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Xavier Calvo
- Laboratory of Cytology, Department of Pathology, GRETNHE, IMIM Hospital del Mar Research Institute, Barcelona, Spain
| | - Felipe Prosper
- Clínica Universidad de Navarra, Universidad de Navarra, Pamplona, Spain
| | - María Díez-Campelo
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - David Valcárcel
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
39
|
Su CH, Liao WJ, Ke WC, Yang RB, Tarn WY. The Y14-p53 regulatory circuit in megakaryocyte differentiation and thrombocytopenia. iScience 2021; 24:103368. [PMID: 34816104 PMCID: PMC8593568 DOI: 10.1016/j.isci.2021.103368] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 09/27/2021] [Accepted: 10/26/2021] [Indexed: 11/19/2022] Open
Abstract
Thrombocytopenia-absent radius (TAR) syndrome is caused by RBM8A insufficiency. We generated megakaryocyte-specific Rbm8a knockout (Rbm8aKOMK) mice that exhibited marked thrombocytopenia, internal hemorrhage, and splenomegaly, providing evidence that genetic deficiency of Rbm8a causes a disorder of platelet production. Rbm8aKOMK mice accumulated low-ploidy immature megakaryocytes in the bone marrow and exhibited defective platelet activation and aggregation. Accordingly, depletion of Y14 (RBM8A) in human erythroleukemia (HEL) cells compromised phorbol-ester-induced polyploidization. Notably, Y14/RBM8A deficiency induced both p53 and p21 in megakaryocytes and HEL cells. Treatment with a p53 inhibitor restored ex vivo differentiation of Rbm8aKOMK megakaryocytes and unexpectedly activated Y14 expression in HEL cells. Trp53 knockout partially restored megakaryocyte differentiation by reversing cell-cycle arrest and increased platelet counts of Rbm8aKOMK, indicating that excess p53 in part accounts for thrombocytopenia in TAR syndrome. This study provides evidence for the role of the Y14-p53 circuit in platelet production and a potential therapeutic strategy.
Collapse
Affiliation(s)
- Chun-Hao Su
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Section 2, Nangang, Taipei 11529, Taiwan
| | - Wei-Ju Liao
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Section 2, Nangang, Taipei 11529, Taiwan
| | - Wei-Chi Ke
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Section 2, Nangang, Taipei 11529, Taiwan
| | - Ruey-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Section 2, Nangang, Taipei 11529, Taiwan
| | - Woan-Yuh Tarn
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Section 2, Nangang, Taipei 11529, Taiwan
- Corresponding author
| |
Collapse
|
40
|
Sladky VC, Eichin F, Reiberger T, Villunger A. Polyploidy control in hepatic health and disease. J Hepatol 2021; 75:1177-1191. [PMID: 34228992 DOI: 10.1016/j.jhep.2021.06.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/25/2021] [Accepted: 06/15/2021] [Indexed: 12/24/2022]
Abstract
A balanced increase in DNA content (ploidy) is observed in some human cell types, including bone-resorbing osteoclasts, platelet-producing megakaryocytes, cardiomyocytes or hepatocytes. The impact of increased hepatocyte ploidy on normal physiology and diverse liver pathologies is still poorly understood. Recent findings suggest swift genetic adaptation to hepatotoxic stress and the protection from malignant transformation as beneficial effects. Herein, we discuss the molecular mechanisms regulating hepatocyte polyploidisation and its implication for different liver diseases and hepatocellular carcinoma. We report on centrosomes' role in limiting polyploidy by activating the p53 signalling network (via the PIDDosome multiprotein complex) and we discuss the role of this pathway in liver disease. Increased hepatocyte ploidy is a hallmark of hepatic inflammation and may play a protective role against liver cancer. Our evolving understanding of hepatocyte ploidy is discussed from the perspective of its potential clinical application for risk stratification, prognosis, and novel therapeutic strategies in liver disease and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Valentina C Sladky
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Felix Eichin
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), 1090 Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Andreas Villunger
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), 1090 Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria.
| |
Collapse
|
41
|
Diab D, Pinon A, Ouk C, Hage-Sleiman R, Diab-Assaf M, Liagre B, Leger DY. Involvement of autophagy in diosgenin‑induced megakaryocyte differentiation in human erythroleukemia cells. Mol Med Rep 2021; 24:746. [PMID: 34458927 PMCID: PMC8436216 DOI: 10.3892/mmr.2021.12386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/21/2021] [Indexed: 11/06/2022] Open
Abstract
Natural agents have been used to restart the process of differentiation that is inhibited during leukemic transformation of hematopoietic stem or progenitor cells. Autophagy is a housekeeping pathway that maintains cell homeostasis against stress by recycling macromolecules and organelles and plays an important role in cell differentiation. In the present study, an experimental model was established to investigate the involvement of autophagy in the megakaryocyte differentiation of human erythroleukemia (HEL) cells induced by diosgenin [also known as (25R)‑Spirosten‑5‑en‑3b‑ol]. It was demonstrated that Atg7 expression was upregulated from day 1 of diosgenin‑induced differentiation and was accompanied by a significant elevation in the conversion of light chain 3 A/B (LC3‑A/B)‑I to LC3‑A/B‑II. Autophagy was modulated before or after the induction of megakaryocyte differentiation using 3‑methyladenine (3‑MA, autophagy inhibitor) and metformin (Met, autophagy initiation activator). 3‑MA induced a significant accumulation of the LC3 A/B‑II form at day 8 of differentiation. It was revealed that 3‑MA had a significant repressive effect on the nuclear (polyploidization) and membrane glycoprotein V [(GpV) expression] maturation. On the other hand, autophagy activation increased GpV genomic expression, but did not change the nuclear maturation profile after HEL cells treatment with Met. It was concluded that autophagy inhibition had a more prominent effect on the diosgenin‑differentiated cells than autophagy activation.
Collapse
Affiliation(s)
- Dima Diab
- PEIRENE Laboratory EA 7500, Faculty of Pharmacy, University of Limoges, 87025 Limoges, France
| | - Aline Pinon
- PEIRENE Laboratory EA 7500, Faculty of Pharmacy, University of Limoges, 87025 Limoges, France
| | - Catherine Ouk
- BISCEm Flow Cytometry/Microscopy Unit, University of Limoges, 87025 Limoges, France
| | - Rouba Hage-Sleiman
- Department of Biology, Faculty of Sciences, Lebanese University, Hadath El Jebbeh, Beyrouth 21219, Lebanon
| | - Mona Diab-Assaf
- Doctoral School of Sciences and Technology, Lebanese University, Hadath El Jebbeh, Beyrouth 21219, Lebanon
| | - Bertrand Liagre
- PEIRENE Laboratory EA 7500, Faculty of Pharmacy, University of Limoges, 87025 Limoges, France
| | - David Yannick Leger
- PEIRENE Laboratory EA 7500, Faculty of Pharmacy, University of Limoges, 87025 Limoges, France
| |
Collapse
|
42
|
Abstract
Thrombocytopoiesis is a complex process beginning at the level of hematopoietic stem cells, which ultimately generate megakaryocytes, large marrow cells with a distinctive morphology, and then, through a process of terminal maturation, megakaryocytes shed thousands of platelets into the circulation. This process is controlled by intrinsic and extrinsic factors. Emerging data indicate that an important intrinsic control on the late stages of thrombopoiesis is exerted by integrins, a family of transmembrane receptors composed of one α and one β subunit. One β subunit expressed by megakaryocytes is the β1 integrin, the role of which in the regulation of platelet formation is beginning to be clarified. Here, we review recent data indicating that activation of β1 integrin by outside-in and inside-out signaling regulates the interaction of megakaryocytes with the endosteal niche, which triggers their maturation, while its inactivation by galactosylation determines the migration of these cells to the perivascular niche, where they complete their terminal maturation and release platelets in the bloodstream. Furthermore, β1 integrin mediates the activation of transforming growth factor β (TGF-β), a protein produced by megakaryocytes that may act in an autocrine fashion to halt their maturation and affect the composition of their surrounding extracellular matrix. These findings suggest that β1 integrin could be a therapeutic target for inherited and acquired disorders of platelet production.
Collapse
Affiliation(s)
- Maria Mazzarini
- Biomedical and Neuromotor Sciences, Alma Mater University Bologna, Italy
| | - Paola Verachi
- Biomedical and Neuromotor Sciences, Alma Mater University Bologna, Italy
| | - Fabrizio Martelli
- National Center for Preclinical and Clinical Research and Evaluation of Pharmaceutical Drugs, Rome, Italy
| | - Anna Rita Migliaccio
- University Campus Biomedico, Rome, Italy
- Myeloproliferative Neoplasm-Research Consortium, New York, NY, USA
| |
Collapse
|
43
|
Liu C, Huang B, Wang H, Zhou J. The heterogeneity of megakaryocytes and platelets and implications for ex vivo platelet generation. Stem Cells Transl Med 2021; 10:1614-1620. [PMID: 34536061 PMCID: PMC8641090 DOI: 10.1002/sctm.21-0264] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/20/2021] [Accepted: 08/29/2021] [Indexed: 12/23/2022] Open
Abstract
Platelets, the chief effector of hemostasis, are small anucleate blood cells generated from megakaryocytes (MKs), and the defects in platelet production or function lead to a variety of bleeding complications. Emerging evidence indicates that MKs and platelets are much more diverse than previously appreciated and involved in many physiological and pathological processes besides hemostasis, such as innate and adaptive immune responses, angiogenesis, and tumor metastasis, while the ontogenic variations in MK and platelet function have also become a focus in the field. However, whether MKs and platelets fulfill these distinct functions by utilizing distinct subpopulations remains poorly understood. New studies aimed at deciphering the MK transcriptome at the single‐cell level have provided some key insights into the functional heterogeneity of MKs. In this review, we will discuss some of the recent discoveries of functional and developmental heterogeneity of MKs and its potential link to the heterogeneity of platelets. We will also discuss the implications of these findings while focusing on the ex vivo generation of platelets from human pluripotent stem cells. The improved understanding of the heterogeneity underlying human MK development and platelet production should open new avenues for future platelet regeneration and clinical treatment of related diseases.
Collapse
Affiliation(s)
- Cuicui Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, People's Republic of China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, Tianjin, People's Republic of China
| | - Baiming Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, People's Republic of China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, Tianjin, People's Republic of China
| | - Hongtao Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, People's Republic of China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, Tianjin, People's Republic of China
| | - Jiaxi Zhou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, People's Republic of China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, Tianjin, People's Republic of China
| |
Collapse
|
44
|
Noh JY. Megakaryopoiesis and Platelet Biology: Roles of Transcription Factors and Emerging Clinical Implications. Int J Mol Sci 2021; 22:ijms22179615. [PMID: 34502524 PMCID: PMC8431765 DOI: 10.3390/ijms22179615] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 12/13/2022] Open
Abstract
Platelets play a critical role in hemostasis and thrombus formation. Platelets are small, anucleate, and short-lived blood cells that are produced by the large, polyploid, and hematopoietic stem cell (HSC)-derived megakaryocytes in bone marrow. Approximately 3000 platelets are released from one megakaryocyte, and thus, it is important to understand the physiologically relevant mechanism of development of mature megakaryocytes. Many genes, including several key transcription factors, have been shown to be crucial for platelet biogenesis. Mutations in these genes can perturb megakaryopoiesis or thrombopoiesis, resulting in thrombocytopenia. Metabolic changes owing to inflammation, ageing, or diseases such as cancer, in which platelets play crucial roles in disease development, can also affect platelet biogenesis. In this review, I describe the characteristics of platelets and megakaryocytes in terms of their differentiation processes. The role of several critical transcription factors have been discussed to better understand the changes in platelet biogenesis that occur during disease or ageing.
Collapse
Affiliation(s)
- Ji-Yoon Noh
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| |
Collapse
|
45
|
Saultier P, Cabantous S, Puceat M, Peiretti F, Bigot T, Saut N, Bordet JC, Canault M, van Agthoven J, Loosveld M, Payet-Bornet D, Potier D, Falaise C, Bernot D, Morange PE, Alessi MC, Poggi M. GATA1 pathogenic variants disrupt MYH10 silencing during megakaryopoiesis. J Thromb Haemost 2021; 19:2287-2301. [PMID: 34060193 DOI: 10.1111/jth.15412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/24/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND GATA1 is an essential transcription factor for both polyploidization and megakaryocyte (MK) differentiation. The polyploidization defect observed in GATA1 variant carriers is not well understood. OBJECTIVE To extensively phenotype two pedigrees displaying different variants in the GATA1 gene and determine if GATA1 controls MYH10 expression levels, a key modulator of MK polyploidization. METHOD A total of 146 unrelated propositi with constitutional thrombocytopenia were screened on a multigene panel. We described the genotype-phenotype correlation in GATA1 variant carriers and investigated the effect of these novel variants on MYH10 transcription using luciferase constructs. RESULTS The clinical profile associated with the p.L268M variant localized in the C terminal zinc finger was unusual in that the patient displayed bleeding and severe platelet aggregation defects without early-onset thrombocytopenia. p.N206I localized in the N terminal zinc finger was associated, on the other hand, with severe thrombocytopenia (15G/L) in early life. High MYH10 levels were evidenced in platelets of GATA1 variant carriers. Analysis of MKs anti-GATA1 chromatin immunoprecipitation-sequencing data revealed two GATA1 binding sites, located in the 3' untranslated region and in intron 8 of the MYH10 gene. Luciferase reporter assays showed their respective role in the regulation of MYH10 gene expression. Both GATA1 variants significantly alter intron 8 driven MYH10 transcription. CONCLUSION The discovery of an association between MYH10 and GATA1 is a novel one. Overall, this study suggests that impaired MYH10 silencing via an intronic regulatory element is the most likely cause of GATA1-related polyploidization defect.
Collapse
Affiliation(s)
- Paul Saultier
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
- Department of Pediatric Hematology, Immunology and Oncology, APHM, La Timone Children's Hospital, Marseille, France
| | | | | | | | - Timothée Bigot
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
| | - Noémie Saut
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
- APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | | | | | - Johannes van Agthoven
- Structural Biology Program, Division of Nephrology/Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Marie Loosveld
- APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
- Aix-Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | | | | - Céline Falaise
- Department of Pediatric Hematology, Immunology and Oncology, APHM, La Timone Children's Hospital, Marseille, France
- APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | - Denis Bernot
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
| | - Pierre-Emmanuel Morange
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
- APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | - Marie-Christine Alessi
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
- APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | - Marjorie Poggi
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
| |
Collapse
|
46
|
Kirillova A, Han L, Liu H, Kühn B. Polyploid cardiomyocytes: implications for heart regeneration. Development 2021; 148:271050. [PMID: 34897388 DOI: 10.1242/dev.199401] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Terminally differentiated cells are generally thought to have arrived at their final form and function. Many terminally differentiated cell types are polyploid, i.e. they have multiple copies of the normally diploid genome. Mammalian heart muscle cells, termed cardiomyocytes, are one such example of polyploid cells. Terminally differentiated cardiomyocytes are bi- or multi-nucleated, or have polyploid nuclei. Recent mechanistic studies of polyploid cardiomyocytes indicate that they can limit cellular proliferation and, hence, heart regeneration. In this short Spotlight, we present the mechanisms generating bi- and multi-nucleated cardiomyocytes, and the mechanisms generating polyploid nuclei. Our aim is to develop hypotheses about how these mechanisms might relate to cardiomyocyte proliferation and cardiac regeneration. We also discuss how these new findings could be applied to advance cardiac regeneration research, and how they relate to studies of other polyploid cells, such as cancer cells.
Collapse
Affiliation(s)
- Anna Kirillova
- Medical Scientist Training Program, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA 15219, USA
| | - Lu Han
- Division of Cardiology, UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA
| | - Honghai Liu
- Division of Cardiology, UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA
| | - Bernhard Kühn
- Division of Cardiology, UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,McGowan Institute of Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| |
Collapse
|
47
|
Sugimoto K, Toume K. Amphibian thrombocyte-derived extracellular vesicles, including microRNAs, induce angiogenesis-related genes in endothelial cells. Genes Cells 2021; 26:757-771. [PMID: 34224189 DOI: 10.1111/gtc.12882] [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: 01/31/2021] [Revised: 06/22/2021] [Accepted: 06/30/2021] [Indexed: 11/29/2022]
Abstract
Thrombocytes circulate in the blood of nonmammalian vertebrates and are involved in hemostasis; however, many detailed characteristics of thrombocytes remain unclear. Recently, we established an amphibian thrombocyte cell line. Here, we report the finding that thrombocytes produce integrin alpha IIb (CD41)-positive extracellular vesicles (EVs), which include microRNAs (miRs). Flow cytometric analysis showed the expression of CD41+ and phosphatidylserine on the surface of EVs. Nanotracking analysis showed that these CD41+ EVs were approximately 100 nm in diameter. As CD41+ EVs were also observed from African clawed frogs, the production of CD41+ EVs might be common to amphibians. Microarray analysis showed that the CD41+ EVs contain many kinds of miRs. These CD41+ EVs were phagocytosed by endothelial cells and macrophages. qPCR analysis showed that many angiogenesis-related genes were up-regulated in CD41+ EV-treated endothelial cells. Over-expression of some miRs in the CD41+ EVs increased the proliferation of endothelial cells. These results indicated that thrombocytes produced CD41+ EVs, including miRs, that were received by endothelial cells to induce the expression of angiogenesis-related genes. These results indicated that the CD41+ EVs produced from thrombocytes act as signaling molecules to repair damaged blood vessels.
Collapse
Affiliation(s)
- Kenkichi Sugimoto
- Department of Cell Science, Faculty of the Graduate School of Science and Technology, Niigata University, Niigata, Japan
| | - Kayano Toume
- Department of Cell Science, Faculty of the Graduate School of Science and Technology, Niigata University, Niigata, Japan
| |
Collapse
|
48
|
Hamad MA, Schanze N, Schommer N, Nührenberg T, Duerschmied D. Reticulated Platelets-Which Functions Have Been Established by In Vivo and In Vitro Data? Cells 2021; 10:cells10051172. [PMID: 34065800 PMCID: PMC8150321 DOI: 10.3390/cells10051172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/29/2021] [Accepted: 05/10/2021] [Indexed: 12/18/2022] Open
Abstract
Reticulated platelets (RP) are the youngest platelet fraction released into the circulation. These immature platelets have increased RNA content, a larger cell volume, more dense granules, higher levels of surface activation markers and are thought to be more reactive compared to their mature counterparts. RP have been associated with cardiovascular disease, diabetes and increased mortality. Yet only a few animal studies investigating RP have been conducted so far and further investigations are warranted. Established methods to count RP are flow cytometry (staining with thiazole orange or SYTO13) or fully automated hematology analyzers (immature platelet fraction, IPF). IPF has been established as a diagnostic parameter in thrombocytopenia, cardiovascular disease and, in particular, the response to antiplatelet therapy. This review seeks to provide an overview of the key features of RP as well as preanalytical and analytical aspects that need to be considered when working with this platelet population.
Collapse
Affiliation(s)
- Muataz Ali Hamad
- Department of Cardiology and Angiology I, Heart Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany; (N.S.); (N.S.); (D.D.)
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg im Breisgau, Germany
- Correspondence: ; Tel.: +49-761-270-70470
| | - Nancy Schanze
- Department of Cardiology and Angiology I, Heart Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany; (N.S.); (N.S.); (D.D.)
| | - Nicolas Schommer
- Department of Cardiology and Angiology I, Heart Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany; (N.S.); (N.S.); (D.D.)
| | - Thomas Nührenberg
- Department of Cardiology and Angiology II, Heart Center, Faculty of Medicine, University of Freiburg, 79189 Bad Krozingen, Germany;
| | - Daniel Duerschmied
- Department of Cardiology and Angiology I, Heart Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany; (N.S.); (N.S.); (D.D.)
| |
Collapse
|
49
|
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
|
50
|
Zhang J, Qiao Q, Xu H, Zhou R, Liu X. Human cell polyploidization: The good and the evil. Semin Cancer Biol 2021; 81:54-63. [PMID: 33839294 DOI: 10.1016/j.semcancer.2021.04.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023]
Abstract
Therapeutic resistance represents a major cause of death for most lethal cancers. However, the underlying mechanisms of such resistance have remained unclear. The polyploid cells are due to an increase in DNA content, commonly associated with cell enlargement. In human, they play a variety of roles in physiology and pathologic conditions and perform the specialized functions during development, inflammation, and cancer. Recent work shows that cancer cells can be induced into polyploid giant cancer cells (PGCCs) that leads to reprogramming of surviving cancer cells to acquire resistance. In this article, we will review the polyploidy involved in development and inflammation, and the process of PGCCs formation and propagation that benefits to cell survival. We will discuss the potential opportunities in fighting resistant cancers. The increased knowledge of PGCCs will offer a completely new paradigm to explore the therapeutic intervention for lethal cancers.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Qing Qiao
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Hong Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Ru Zhou
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xinzhe Liu
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|