Elsevier

Life Sciences

Volume 301, 15 July 2022, 120602
Life Sciences

Review article
Megakaryocytes in pulmonary diseases

https://doi.org/10.1016/j.lfs.2022.120602Get rights and content

Abstract

Megakaryocytes (MKs) are typical cellular components in the circulating blood flowing from the heart into the lungs. Physiologically, MKs function as an important regulator of platelet production and immunoregulation. However, dysfunction in MKs is considered a trigger in various diseases. It has been described that the lung is an important site of platelet biogenesis from extramedullary MKs, which may play an essential role in various pulmonary diseases. With detailed studies, there are different degrees of numerical changes of MKs in coronavirus disease 2019 (COVID-19), acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD), lung cancer, pulmonary fibrosis (PF), and other pulmonary diseases. Also, MKs inhibit or promote the development of pulmonary diseases through various pathways. Here, we summarize the current knowledge of MKs in pulmonary diseases, highlighting the physiological functions and integrated molecular mechanisms. We aim to shine new light on not only the subsequent study of MKs but also the diagnosis and treatment of pulmonary diseases.

Introduction

Megakaryocytes (MKs) are precursor cells in the blood system responsible for producing platelets (PLT) [1]. Like other blood cells, MKs are derived from hematopoietic stem cell, which found mainly in bone marrow, producing all blood cells in the circulatory system [1]. According to the classical hematopoietic model, HSC develops into megakaryocyte progenitor (MkP) through the gradual differentiation process of long-term HSC, short-term HSC, multipotent progenitors (MPP) and bipo-tent megakaryocyte-erythrocyte progenitor (MEP) [1]. Recent studies have shown that HSC contain a subset of cells with biased megakaryocyte potential, which can rapidly differentiate into MKs to effectively replenish PLT during inflammatory stress without going through MPP and MEP stages [2]. MkP maintains self-renewal through active mitosis. When confronted with some as-yet unidentified triggering event, MkP stops mitosis and enters a progress called endomitosis, in which the nucleus and cells grow in size, DNA continues to replicate but neither the nucleus nor the cytoplasm divides, and in this way, a single giant nucleus is formed with between 4 and 16 times the DNA of normal diploid DNA (hence the name MKs) [3]. This is also a classic example of developmentally regulated polyploidization in mammals. After completion of endomitosis, the cytoplasm of MKs begins to expand rapidly, filling with platelet-specific particles and forming an elaborate and highly convoluted invaginal membrane system (IMS) in preparation for platelet formation and release [4]. The main job of MKs is to produce and replenish platelets to maintain the body's normal coagulation function. Toxins, alcohol intake, and vitamin B12 deficiency can inhibit endomitosis and maturation of MKs. Some drugs, such as Bortezomib, can reduce PLT shedding in existing mature MKs, leading to thrombocytopenia and increasing the risk of bleeding [3]. Apoptosis of MKs caused by antiplatelet antibodies and cytotoxic T cells is the main cause of immune thrombocytopenia (ITP) [3]. In addition to acting as precursors to PLT, MKs can promote angiogenesis, bone formation [5], [6]and even maintain homeostatic quiescence [7], [8], [9] (important for stemness of HSC) by releasing mediators such as vascular endothelial growth factor and transforming growth factor (TGF). Both MKs and PLT are believed to have inflammatory cell functions and play an active role in innate and acquired immunity [10]. Notably, MKs can play a pro-inflammatory role independently of PLT, such as the transfer of IL-1-rich microparticles released by MKs to synovial tissues that aggravate inflammatory arthritis [10].

MKs were discovered in the lung early in 1893 [11]. Since then, much more information about indirect signs of platelet biogenesis in the lung has become available, which manifests that MKs decrease with PLT release after blood passes through the pulmonary circulation [12], [13], [14], [15]. The process by which MKs release PLT in the lung has not been discovered until 2017 when it was first dynamically and directly observed through intravital microscopy [16]. Based on those empirical studies, Lung has been recognized as one of the important biogenic sites of platelet in addition to bone marrow [16], [17]. Furthermore, MKs inherent in the lung have more essential physiological functions besides the same role of platelet biogenesis as those MKs in the bone marrow. More recent evidence suggests that pulmonary inherent MKs have various immunoregulatory functions [18], and transcriptome sequencing outcomes indicate that they express more mRNA related to innate immunity, such as Toll-like receptors (TLRs), chemokines, and cytokines [19].

It has aroused more and more attention that MKs are involved in the progression of pulmonary diseases, such as pneumonia, including coronavirus disease 2019 (COVID-19), acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), and lung cancer. The first thing that raises our concern is that pulmonary diseases are often accompanied by quantitative or functional abnormalities of MKs and PLT. In pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), MKs not only involve in antiviral processes but also inhibit the advancement of COVID-19 by approaches like altering gene expression and participating in antigen presentation [20]. However, MKs can also play an adverse role against the diseases, like promoting the inflammatory response in ARDS and COPD [21], [22], [23]. In addition, MKs contain various fibrosis-related growth factors, which enables MKs to promote the development of fibrotic phenotype in PF through various pathways. More than that, MKs seem to have different roles in different stages of the disease. For instance, in lung cancer, MKs exert anti-tumor effects by inhibiting angiogenesis in the tumor microenvironment in the early stage, while exert tumor-promoting effects by releasing intracellular cytokines in the late stage. Notably, MKs play a crucial role in the development of pulmonary diseases. Targeting regulation of MKs holds promising prospects for the treatment of pulmonary diseases.

In this review, we first summarize the major physiological functions of MKs, especially the immunomodulatory effect. Then, we mainly focus on MKs' pathological function, including the molecular mechanisms in some comma pulmonary diseases. We also propose possible therapeutic ideas for those pulmonary diseases based on our current knowledge of MKs, hoping to shine new light on not only the subsequent study of MKs but also the diagnosis and treatment of pulmonary diseases.

Section snippets

MKs-Platelet axis

MKs, that release PLT in the pulmonary circulation generally come from extrapulmonary tissues such as bone marrow and spleen [24]. In the bone marrow, MKs are differentiated from HSC by a process dependent on the thrombopoietin (TPO) [25]. Under the influence of fibroblast growth factor-4 (FGF-4) and stromal-derived-factor-1 (SDF-1), which is also known as C-X-C chemokine ligand 12 (CXCL12), MKs reposition around the bone marrow sinuses and interact with the endothelial cells mediated by

MKs and pneumonia

MKs are involved in various pneumonia and play different roles. As reported in a case in 2011, a 56-year-old female patient with polycythemia vera (PV) harboring a JAK2V617F mutation showed a complication of organizing pneumonia (OP). Atypical MKs (CD41+, CD68, S-100) were diffusely distributed around granulation tissue, especially plugging alveolar blood capillaries, while a large number of PLT aggregated in granulation tissues. These findings suggested that in the JAK2 mutation or the

MKs and lung cancer

Lung cancer is the most common cause of cancer mortality around the world [100]. There-into non-small cell lung cancer (NSCLC) accounts for approximately 80% [101]. Although multiple treatments have been developed, the prognosis of patients with advanced NSCLC is generally poor, with an overall 5-year survival rate of 24% [102]. However, experiments on NSCLC report that the increased number and density of MKs in NSCLC tissues suggests a poor prognosis [103]. Pulmonary circulation is the source

Clinical targets

Some drugs targeting MKs or PLT have been applied to pulmonary diseases. For MKs-targeted drugs, stimulation of MKs production may inhibit lung cancer progression. In lung cancer, erythropenia may occur during chemotherapy. However, platelet transfusion may have defects such as allogeneic immunity, so specific stimulation of platelet production may be more reliable. Polyethylene glycol-conjugated recombinant human megakaryocyte growth and development factor is a recombinant molecule associated

Summary and prospect

MKs are platelet-producing cells derived from the differentiation of HSCs. In the bone marrow, MKs are derived from HSCs and develop into mononuclear, lobulated, and polyploid karyotype large cells depending on TPO. While in the lung, some of the MKs are derived from pulmonary HSCs or resident MKs, which are located in the lung interstitium. And the other intrapulmonary MKs come from extrapulmonary tissues such as bone marrow and spleen. Both types of MKs have the potential to produce PLT.

Abbreviations

    AURKA

    Aurora kinase A

    β4GalT1

    β-1,4-galactosyltransferase 1

    ARDS

    acute respiratory distress syndrome

    APC

    antigen-presenting cells

    COPD

    chronic obstructive pulmonary disease

    COVID-19

    coronavirus disease 2019

    CXCL12

    C-X-C chemokine ligand 12

    CXCR4

    C-X-C chemokine receptor type 4

    CCR7

    CC-chemokine receptor 7

    cPLA2

    cytoplasmic phospholipase 2

    DAD

    Diffuse alveolar injury

    DPEMH

    diffuse pulmonary extramedullary hematopoiesis

    DQ-Ova

    DQ-ovalbumin

    EMH

    extramedullary hematopoiesis

    FGF-4

    fibroblast growth factor-4

    HSCs

    hematopoietic

CRediT authorship contribution statement

Di-Yun Huang, Zhuo-Ran Ke, Guan-Ming Wang, and Yong Zhou wrote the original draft. Di-Yun Huang, Zhuo-Ran Ke, and Guan-Ming Wang prepared the figures. Hui-Hui Yang revised the figures. Cha-Xiang Guan and Tian-Liang Ma edited and critically reviewed the manuscript. All authors read and approved the final manuscript.

Declaration of competing interest

The authors declared no conflict of interest.

Acknowledgments

This work was supported by the Major Research Plan in the field of Social Development of the Hunan Province (2020SK3024) and the National University Student Innovation Program (S2020105330820).

References (119)

  • D. Zucker-Franklin et al.

    Platelet production in the pulmonary capillary bed: new ultrastructural evidence for an old concept [J]

    Am. J. Pathol.

    (2000)
  • J.N. Thon

    SDF-1 directs megakaryocyte relocation [J]

    Blood

    (2014)
  • J. Kailashiya

    Platelet-derived microparticles analysis: techniques, challenges and recommendations [J]

    Anal. Biochem.

    (2018)
  • L.M. Beaulieu et al.

    Regulatory effects of TLR2 on megakaryocytic cell function [J]

    Blood

    (2011)
  • D. Metcalf

    Hematopoietic cytokines [J]

    Blood

    (2008)
  • L. Guo et al.

    Platelet MHC class I mediates CD8+ T-cell suppression during sepsis [J]

    Blood

    (2021)
  • T.M. Vallance et al.

    Development and characterization of a novel, megakaryocyte NF-kappaB reporter cell line for investigating inflammatory responses [J]

    J. Thromb. Haemost.

    (2021)
  • Z. Wang et al.

    Hypermucoviscous Klebsiella pneumoniae infections induce platelet aggregation and apoptosis and inhibit maturation of megakaryocytes [J]

    Thromb. Res.

    (2018)
  • L.P. D'Atri et al.

    Expression and functionality of toll-like receptor 3 in the megakaryocytic lineage [J]

    J. Thromb. Haemost.

    (2015)
  • B.K. Manne et al.

    Platelet gene expression and function in patients with COVID-19 [J]

    Blood

    (2020)
  • G. Ortiz-Muñoz et al.

    Aspirin-triggered 15-epi-lipoxin A4 regulates neutrophil-platelet aggregation and attenuates acute lung injury in mice [J]

    Blood

    (2014)
  • R.V. Mandal et al.

    Megakaryocytes and platelet homeostasis in diffuse alveolar damage [J]

    Exp. Mol. Pathol.

    (2007)
  • G. Couldwell et al.

    Modulation of megakaryopoiesis and platelet production during inflammation [J]

    Thromb. Res.

    (2019)
  • M. Viecca et al.

    Enhanced platelet inhibition treatment improves hypoxemia in patients with severe Covid-19 and hypercoagulability. A case control, proof of concept study [J]

    Pharmacol. Res.

    (2020)
  • L. Richeldi et al.

    Idiopathic pulmonary fibrosis [J]

    Lancet

    (2017)
  • H. Castro-Malaspina et al.

    Human megakaryocyte stimulation of proliferation of bone marrow fibroblasts [J]

    Blood

    (1981)
  • J. Thachil

    The lung megakaryocytes and pulmonary fibrosis in systemic sclerosis [J]

    Med. Hypotheses

    (2009)
  • A. Schmitt et al.

    Pathologic interaction between megakaryocytes and polymorphonuclear leukocytes in myelofibrosis [J]

    Blood

    (2000)
  • N. Papadantonakis et al.

    Megakaryocyte pathology and bone marrow fibrosis: the lysyl oxidase connection [J]

    Blood

    (2012)
  • A. Eliades et al.

    Control of megakaryocyte expansion and bone marrow fibrosis by lysyl oxidase [J]

    J. Biol. Chem.

    (2011)
  • C.A. Koch et al.

    Nonhepatosplenic extramedullary hematopoiesis: associated diseases, pathology, clinical course, and treatment [J]

    Mayo Clin. Proc.

    (2003)
  • S. Asakura et al.

    Agnogenic myeloid metaplasia with extramedullary hematopoiesis and fibrosis in the lung. Report of two cases [J]

    Chest

    (1994)
  • F.R. Hirsch et al.

    Lung cancer: current therapies and new targeted treatments [J]

    Lancet

    (2017)
  • K.R. Machlus et al.

    The incredible journey: from megakaryocyte development to platelet formation [J]

    J. Cell Biol.

    (2013)
  • Y. Tang et al.

    Megakaryocytes promote bone formation through coupling osteogenesis with angiogenesis by secreting TGF-β1 [J]

    Theranostics

    (2020)
  • L. Jiang et al.

    SHP-1 regulates hematopoietic stem cell quiescence by coordinating TGF-β signaling [J]

    J. Exp. Med.

    (2018)
  • E. Boilard et al.

    17 - platelets and megakaryocytes

  • Y. Zhou et al.

    The platelet-producing function of lung] [J

    Sheng Li Xue Bao

    (2017)
  • N.T. Pedersen

    Occurrence of megakaryocytes in various vessels and their retention in the pulmonary capillaries in man [J]

    Scand. J. Haematol.

    (1978)
  • A. Kallinikos-Maniatis

    Megakaryocytes and platelets in central venous and arterial blood [J]

    Acta Haematol.

    (1969)
  • N.T. Pedersen

    The pulmonary vessels as a filter for circulating megakaryocytes in rats [J]

    Scand. J. Haematol.

    (1974)
  • R.F. Levine et al.

    Circulating megakaryocytes: delivery of large numbers of intact, mature megakaryocytes to the lungs [J]

    Eur. J. Haematol.

    (1993)
  • E. Lefrancais et al.

    The lung is a site of platelet biogenesis and a reservoir for haematopoietic progenitors [J]

    Nature

    (2017)
  • M. Banerjee et al.

    Platelets endocytose viral particles and are activated via TLR (toll-like receptor) signaling [J]

    Arterioscler. Thromb. Vasc. Biol.

    (2020)
  • J.P. Bernardes et al.

    Longitudinal multi-omics analyses identify responses of megakaryocytes, erythroid cells, and plasmablasts as hallmarks of severe COVID-19 [J]

    Immunity

    (2020)
  • A.V. Washington et al.

    Platelet biology of the rapidly failing lung [J]

    Br. J. Haematol.

    (2020)
  • J.B. Kral-Pointner et al.

    Platelet PI3K modulates innate leukocyte extravasation during acid-induced acute lung inflammation [J]

    Thromb. Haemost.

    (2019)
  • J. Grommes et al.

    Disruption of platelet-derived chemokine heteromers prevents neutrophil extravasation in acute lung injury [J]

    Am. J. Respir. Crit. Care Med.

    (2012)
  • E. Lefrançais et al.

    Platelet biogenesis in the lung circulation [J]

    Physiology (Bethesda)

    (2019)
  • K.R. Machlus et al.

    The incredible journey: from megakaryocyte development to platelet formation [J]

    J. Cell Biol.

    (2013)
  • Cited by (2)

    • Clinical, laboratory and immunohistochemical characterization of in situ pulmonary arterial thrombosis in fatal COVID-19

      2022, Thrombosis Research
      Citation Excerpt :

      The pathological examination of pulmonary vasculature in COVID-19 patients showed diffuse alveolar damage (DAD) with concurrent thrombi formation, even in the microcirculation [12]. Some autopsy studies reported the combined presence of megakaryocytes and thrombi in the lung of COVID-19 patients [22,23], however, the implication of megakaryocytes in COVID-19 pulmonary coagulopathy and even in other pulmonary diseases is still debated [24]. Although many biomarkers have been proposed to estimate the thrombotic risk in COVID-19, a recent consensus statement from the International COVID-19 thrombosis biomarkers Colloquium recommended only C-Reactive Protein and D-dimer to assess the risk of venous thromboembolic events, both biomarkers being correlated to the disease severity [25].

    1

    These authors contributed equally to this work.

    View full text