1
|
Collinson RJ, Wilson L, Boey D, Ng ZY, Mirzai B, Chuah HS, Howman R, Grove CS, Malherbe JAJ, Leahy MF, Linden MD, Fuller KA, Erber WN, Guo BB. Transcription factor 3 is dysregulated in megakaryocytes in myelofibrosis. Platelets 2024; 35:2304173. [PMID: 38303515 DOI: 10.1080/09537104.2024.2304173] [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: 11/15/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024]
Abstract
Transcription factor 3 (TCF3) is a DNA transcription factor that modulates megakaryocyte development. Although abnormal TCF3 expression has been identified in a range of hematological malignancies, to date, it has not been investigated in myelofibrosis (MF). MF is a Philadelphia-negative myeloproliferative neoplasm (MPN) that can arise de novo or progress from essential thrombocythemia [ET] and polycythemia vera [PV] and where dysfunctional megakaryocytes have a role in driving the fibrotic progression. We aimed to examine whether TCF3 is dysregulated in megakaryocytes in MPN, and specifically in MF. We first assessed TCF3 protein expression in megakaryocytes using an immunohistochemical approach analyses and showed that TCF3 was reduced in MF compared with ET and PV. Further, the TCF3-negative megakaryocytes were primarily located near trabecular bone and had the typical "MF-like" morphology as described by the WHO. Genomic analysis of isolated megakaryocytes showed three mutations, all predicted to result in a loss of function, in patients with MF; none were seen in megakaryocytes isolated from ET or PV marrow samples. We then progressed to transcriptomic sequencing of platelets which showed loss of TCF3 in MF. These proteomic, genomic and transcriptomic analyses appear to indicate that TCF3 is downregulated in megakaryocytes in MF. This infers aberrations in megakaryopoiesis occur in this progressive phase of MPN. Further exploration of this pathway could provide insights into TCF3 and the evolution of fibrosis and potentially lead to new preventative therapeutic targets.
Collapse
Affiliation(s)
- Ryan J Collinson
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Lynne Wilson
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Darren Boey
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Zi Yun Ng
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
- Department of Haematology, Royal Perth Hospital, Perth, WA, Australia
| | - Bob Mirzai
- PathWest Laboratory Medicine, Nedlands, WA, Australia
| | - Hun S Chuah
- Department of Haematology, Royal Perth Hospital, Perth, WA, Australia
- PathWest Laboratory Medicine, Nedlands, WA, Australia
- Department of Haematology, Rockingham General Hospital, Rockingham, WA, Australia
| | - Rebecca Howman
- Department of Haematology, Sir Charles Gairdner Hospital Nedlands Australia
| | - Carolyn S Grove
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
- Department of Haematology, Royal Perth Hospital, Perth, WA, Australia
- Department of Haematology, Sir Charles Gairdner Hospital Nedlands Australia
| | | | - Michael F Leahy
- Department of Haematology, Royal Perth Hospital, Perth, WA, Australia
- PathWest Laboratory Medicine, Nedlands, WA, Australia
| | - Matthew D Linden
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Kathryn A Fuller
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Wendy N Erber
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
- PathWest Laboratory Medicine, Nedlands, WA, Australia
| | - Belinda B Guo
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
2
|
Gobbo F, Martelli F, Di Virgilio A, Demaria E, Sarli G, Migliaccio AR. The Variation in the Traits Ameliorated by Inhibitors of JAK1/2, TGF-β, P-Selectin, and CXCR1/CXCR2 in the Gata1low Model Suggests That Myelofibrosis Should Be Treated by These Drugs in Combination. Int J Mol Sci 2024; 25:7703. [PMID: 39062946 PMCID: PMC11277099 DOI: 10.3390/ijms25147703] [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: 05/09/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Studies conducted on animal models have identified several therapeutic targets for myelofibrosis, the most severe of the myeloproliferative neoplasms. Unfortunately, many of the drugs which were effective in pre-clinical settings had modest efficacy when tested in the clinic. This discrepancy suggests that treatment for this disease requires combination therapies. To rationalize possible combinations, the efficacy in the Gata1low model of drugs currently used for these patients (the JAK1/2 inhibitor Ruxolitinib) was compared with that of drugs targeting other abnormalities, such as p27kip1 (Aplidin), TGF-β (SB431542, inhibiting ALK5 downstream to transforming growth factor beta (TGF-β) signaling and TGF-β trap AVID200), P-selectin (RB40.34), and CXCL1 (Reparixin, inhibiting the CXCL1 receptors CXCR1/2). The comparison was carried out by expressing the endpoints, which had either already been published or had been retrospectively obtained for this study, as the fold change of the values in the corresponding vehicles. In this model, only Ruxolitinib was found to decrease spleen size, only Aplidin and SB431542/AVID200 increased platelet counts, and with the exception of AVID200, all the inhibitors reduced fibrosis and microvessel density. The greatest effects were exerted by Reparixin, which also reduced TGF-β content. None of the drugs reduced osteopetrosis. These results suggest that future therapies for myelofibrosis should consider combining JAK1/2 inhibitors with drugs targeting hematopoietic stem cells (p27Kip1) or the pro-inflammatory milieu (TGF-β or CXCL1).
Collapse
Affiliation(s)
- Francesca Gobbo
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University, 40126 Bologna, Italy; (F.G.); (G.S.)
| | - Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.M.); (A.D.V.)
| | - Antonio Di Virgilio
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.M.); (A.D.V.)
| | - Elena Demaria
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University, 40126 Bologna, Italy;
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University, 40126 Bologna, Italy; (F.G.); (G.S.)
| | - Anna Rita Migliaccio
- Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, 73100 Lecce, Italy
| |
Collapse
|
3
|
Dehghan S, Kheshtchin N, Hassannezhad S, Soleimani M. Cell death classification: A new insight based on molecular mechanisms. Exp Cell Res 2023; 433:113860. [PMID: 38013091 DOI: 10.1016/j.yexcr.2023.113860] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/17/2023] [Accepted: 11/18/2023] [Indexed: 11/29/2023]
Abstract
Cells tend to disintegrate themselves or are forced to undergo such destructive processes in critical circumstances. This complex cellular function necessitates various mechanisms and molecular pathways in order to be executed. The very nature of cell death is essentially important and vital for maintaining homeostasis, thus any type of disturbing occurrence might lead to different sorts of diseases and dysfunctions. Cell death has various modalities and yet, every now and then, a new type of this elegant procedure gets to be discovered. The diversity of cell death compels the need for a universal organizing system in order to facilitate further studies, therapeutic strategies and the invention of new methods of research. Considering all that, we attempted to review most of the known cell death mechanisms and sort them all into one arranging system that operates under a simple but subtle decision-making (If \ Else) order as a sorting algorithm, in which it decides to place and sort an input data (a type of cell death) into its proper set, then a subset and finally a group of cell death. By proposing this algorithm, the authors hope it may solve the problems regarding newer and/or undiscovered types of cell death and facilitate research and therapeutic applications of cell death.
Collapse
Affiliation(s)
- Sepehr Dehghan
- Department of Medical Basic Sciences, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Nasim Kheshtchin
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Maryam Soleimani
- Department of Medical Basic Sciences, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Druzhkova I, Ignatova N, Shirmanova M. Cell-in-Cell Structures in Gastrointestinal Tumors: Biological Relevance and Clinical Applications. J Pers Med 2023; 13:1149. [DOI: https:/doi.org/10.3390/jpm13071149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2023] Open
Abstract
This review summarizes information about cell-in-cell (CIC) structures with a focus on gastrointestinal tumors. The phenomenon when one cell lives in another one has attracted an attention of researchers over the past decades. We briefly discuss types of CIC structures and mechanisms of its formation, as well as the biological basis and consequences of the cell-engulfing process. Numerous clinico-histopathological studies demonstrate the significance of these structures as prognostic factors, mainly correlated with negative prognosis. The presence of CIC structures has been identified in all gastrointestinal tumors. However, the majority of studies concern pancreatic cancer. In this field, in addition to the assessment of the prognostic markers, the attempts to manipulate the ability of cells to form CISs have been done in order to stimulate the death of the inner cell. Number of CIC structures also correlates with genetic features for some gastrointestinal tu-mors. The role of CIC structures in the responses of tumors to therapies, both chemotherapy and immunotherapy, seems to be the most poorly studied. However, there is some evidence of involvement of CIC structures in treatment failure. Here, we summarized the current literature on CIC structures in cancer with a focus on gastrointestinal tumors and specified future perspectives for investigation.
Collapse
Affiliation(s)
- Irina Druzhkova
- Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | - Nadezhda Ignatova
- Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | - Marina Shirmanova
- Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| |
Collapse
|
5
|
Druzhkova I, Ignatova N, Shirmanova M. Cell-in-Cell Structures in Gastrointestinal Tumors: Biological Relevance and Clinical Applications. J Pers Med 2023; 13:1149. [PMID: 37511762 PMCID: PMC10381133 DOI: 10.3390/jpm13071149] [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: 06/21/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
This review summarizes information about cell-in-cell (CIC) structures with a focus on gastrointestinal tumors. The phenomenon when one cell lives in another one has attracted an attention of researchers over the past decades. We briefly discuss types of CIC structures and mechanisms of its formation, as well as the biological basis and consequences of the cell-engulfing process. Numerous clinico-histopathological studies demonstrate the significance of these structures as prognostic factors, mainly correlated with negative prognosis. The presence of CIC structures has been identified in all gastrointestinal tumors. However, the majority of studies concern pancreatic cancer. In this field, in addition to the assessment of the prognostic markers, the attempts to manipulate the ability of cells to form CISs have been done in order to stimulate the death of the inner cell. Number of CIC structures also correlates with genetic features for some gastrointestinal tu-mors. The role of CIC structures in the responses of tumors to therapies, both chemotherapy and immunotherapy, seems to be the most poorly studied. However, there is some evidence of involvement of CIC structures in treatment failure. Here, we summarized the current literature on CIC structures in cancer with a focus on gastrointestinal tumors and specified future perspectives for investigation.
Collapse
Affiliation(s)
- Irina Druzhkova
- Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (N.I.); (M.S.)
| | | | | |
Collapse
|
6
|
Arciprete F, Verachi P, Martelli F, Valeri M, Balliu M, Guglielmelli P, Vannucchi AM, Migliaccio AR, Zingariello M. Inhibition of CXCR1/2 reduces the emperipolesis between neutrophils and megakaryocytes in the Gata1 low model of myelofibrosis. Exp Hematol 2023; 121:30-37. [PMID: 36863479 PMCID: PMC11780361 DOI: 10.1016/j.exphem.2023.02.003] [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: 02/08/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023]
Abstract
Emperipolesis between neutrophils and megakaryocytes was first identified by transmission electron microscopy. Although rare under steady-state conditions, its frequency greatly increases in myelofibrosis, the most severe of myeloproliferative neoplasms, in which it is believed to contribute to increasing the transforming growth factor (TGF)-β microenvironmental bioavailability responsible for fibrosis. To date, the challenge of performing studies by transmission electron microscopy has hampered the study of factors that drive the pathological emperipolesis observed in myelofibrosis. We established a user-friendly confocal microscopy method that detects emperipolesis by staining with CD42b, specifically expressed on megakaryocytes, coupled with antibodies that recognize the neutrophils (Ly6b or neutrophil elastase antibody). With such an approach, we first confirmed that the bone marrow from patients with myelofibrosis and from Gata1low mice, a model of myelofibrosis, contains great numbers of neutrophils and megakaryocytes in emperipolesis. Both in patients and Gata1low mice, the emperipolesed megakaryocytes were surrounded by high numbers of neutrophils, suggesting that neutrophil chemotaxis precedes the actual emperipolesis event. Because neutrophil chemotaxis is driven by CXCL1, the murine equivalent of human interleukin 8 that is expressed at high levels by malignant megakaryocytes, we tested the hypothesis that neutrophil/megakaryocyte emperipolesis could be reduced by reparixin, an inhibitor of CXCR1/CXCR2. Indeed, the treatment greatly reduced both neutrophil chemotaxis and their emperipolesis with the megakaryocytes in treated mice. Because treatment with reparixin was previously reported to reduce both TGF-β content and marrow fibrosis, these results identify neutrophil/megakaryocyte emperipolesis as the cellular interaction that links interleukin 8 to TGF-β abnormalities in the pathobiology of marrow fibrosis.
Collapse
Affiliation(s)
- Francesca Arciprete
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy
| | - Paola Verachi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Fabrizio Martelli
- National Center for Preclinical and Clinical Research and Evaluation of Pharmaceutical Drugs, Istituto Superiore di Sanità, Rome, Italy
| | - Mauro Valeri
- Center for Animal Experimentation and Well-Being, Istituto Superiore di Sanità, Rome, Italy
| | - Manjola Balliu
- Center Research and Innovation of Myeloproliferative Neoplasm, University Hospital Careggi, University of Florence, Florence, Italy
| | - Paola Guglielmelli
- Center Research and Innovation of Myeloproliferative Neoplasm, University Hospital Careggi, University of Florence, Florence, Italy
| | - Alessandro Maria Vannucchi
- Center Research and Innovation of Myeloproliferative Neoplasm, University Hospital Careggi, University of Florence, Florence, Italy
| | - Anna Rita Migliaccio
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy; Altius Institute for Biomedical Sciences, Seattle, WA
| | - Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy.
| |
Collapse
|
7
|
Abstract
The Philadelphia-negative myeloproliferative neoplasms (MPNs)-essential thrombocythemia (ET), polycythemia vera (PV) and myelofibrosis (MF), are characterized by a propensity for thrombotic events and variable risks for transformation to MF (for ET and PV) and acute leukemia. Leukocytosis, which serves a minor criterion for the diagnosis of MF, is present in a significant portion of patients with MPNs. The relation and impact of leukocytosis on disease course and outcomes of patients with MPNs has been studied in multiple, large retrospective and prospective studies. Despite this, the association of leukocytosis and thrombosis, fibrosis and leukemic transformation remains unclear. This article details the published investigations regarding the impact of leukocytosis in MPNs and explores the changing role of leukocytosis in disease prognostication as increasing emphasis is placed on molecular and genetic studies.
Collapse
Affiliation(s)
- Alexander Coltoff
- Department of Hematology/Oncology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
8
|
Verachi P, Gobbo F, Martelli F, Falchi M, di Virgilio A, Sarli G, Wilke C, Bruederle A, Prahallad A, Arciprete F, Zingariello M, Migliaccio AR. Preclinical studies on the use of a P-selectin-blocking monoclonal antibody to halt progression of myelofibrosis in the Gata1 low mouse model. Exp Hematol 2023; 117:43-61. [PMID: 36191885 PMCID: PMC10450205 DOI: 10.1016/j.exphem.2022.09.004] [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: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 01/10/2023]
Abstract
The bone marrow (BM) and spleen from patients with myelofibrosis (MF), as well as those from the Gata1low mouse model of the disease contain increased number of abnormal megakaryocytes. These cells express high levels of the adhesion receptor P-selectin on their surface, which triggers a pathologic neutrophil emperipolesis, leading to increased bioavailability of transforming growth factor-β (TGF-β) in the microenvironment and disease progression. With age, Gata1low mice develop a phenotype similar to that of patients with MF, which is the most severe of the Philadelphia-negative myeloproliferative neoplasms. We previously demonstrated that Gata1low mice lacking the P-selectin gene do not develop MF. In the current study, we tested the hypothesis that pharmacologic inhibition of P-selectin may normalize the phenotype of Gata1low mice that have already developed MF. To test this hypothesis, we have investigated the phenotype expressed by aged Gata1low mice treated with the antimouse monoclonal antibody RB40.34, alone and also in combination with ruxolitinib. The results indicated that RB40.34 in combination with ruxolitinib normalizes the phenotype of Gata1low mice with limited toxicity by reducing fibrosis and the content of TGF-β and CXCL1 (two drivers of fibrosis in this model) in the BM and spleen and by restoring hematopoiesis in the BM and the architecture of the spleen. In conclusion, we provide preclinical evidence that treatment with an antibody against P-selectin in combination with ruxolitinib may be more effective than ruxolitinib alone to treat MF in patients.
Collapse
Affiliation(s)
- Paola Verachi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Francesca Gobbo
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy; Department of Veterinary Medical Sciences, University of Bologna, Italy
| | - Fabrizio Martelli
- National Center for Preclinical and Clinical Research and Evaluation of Pharmaceutical Drugs, Istituto Superiore di Sanità, Rome, Italy
| | - Mario Falchi
- National Center for HIV/AIDS Research, Istituto Superiore di Sanità, Rome, Italy
| | - Antonio di Virgilio
- Center for Animal Experimentation and Well-being, Istituto Superiore di Santà, Rome, Italy
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, Italy
| | | | | | | | - Francesca Arciprete
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy
| | - Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy
| | - Anna Rita Migliaccio
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy; Altius Institute for Biomedical Sciences, Seattle, WA, USA.
| |
Collapse
|
9
|
A Novel Morphological Parameter Predicting Fibrotic Evolution in Myeloproliferative Neoplasms: New Evidence and Molecular Insights. Int J Mol Sci 2022; 23:ijms23147872. [PMID: 35887218 PMCID: PMC9322985 DOI: 10.3390/ijms23147872] [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: 05/20/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 02/05/2023] Open
Abstract
Philadelphia-negative chronic myeloproliferative neoplasms (MPNs) represent a group of hematological disorders that are traditionally considered as indistinct slow progressing conditions; still, a subset of cases shows a rapid evolution towards myelofibrotic bone marrow failure. Specific abnormalities in the megakaryocyte lineage seem to play a central role in this evolution, especially in the bone marrow fibrosis but also in the induction of myeloproliferation. In this review, we analyze the current knowledge of prognostic factors of MPNs related to their evolution to myelofibrotic bone marrow failure. Moreover, we focused the role of the megakaryocytic lineage in the various stages of MPNs, with updated examples of MPNs in vitro and in vivo models and new therapeutic implications.
Collapse
|
10
|
Varricchio L, Hoffman R. Megakaryocytes Are Regulators of the Tumor Microenvironment and Malignant Hematopoietic Progenitor Cells in Myelofibrosis. Front Oncol 2022; 12:906698. [PMID: 35646681 PMCID: PMC9130548 DOI: 10.3389/fonc.2022.906698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/15/2022] [Indexed: 12/15/2022] Open
Abstract
Megakaryocytes (MKs) are multifunctional hematopoietic cells that produce platelets, serve as components of bone marrow (BM) niches that support the development of hematopoietic stem and progenitor cell (HSPC) and provide inflammatory signals. MKs can dynamically change their activities during homeostasis and following stress, thereby regulating hematopoietic stem cell (HSC) function. Myelofibrosis (MF) is a progressive chronic myeloproliferative neoplasm (MPN) characterized by hyperactivation of JAK/STAT signaling and MK hyperplasia, which is associated with an aberrant inflammatory signature. Since JAK1/2 inhibitor alone is incapable of depleting the malignant HSC clones or reversing BM fibrosis, the identification of mechanisms that cooperate with MF JAK/STAT signaling to promote disease progression might help in developing combination therapies to modify disease outcomes. Chronic inflammation and MK hyperplasia result in an abnormal release of TGFβ1, which plays a critical role in the pathobiology of MF by contributing to the development of BM fibrosis. Dysregulated TGFβ signaling can also alter the hematopoietic microenvironment supporting the predominance of MF-HSCs and enhance the quiescence of the reservoir of wild-type HSCs. Upregulation of TGFβ1 levels is a relatively late event in MF, while during the early pre-fibrotic stage of MF the alarmin S100A8/S100A9 heterocomplex promotes pro-inflammatory responses and sustains the progression of MF-HSCs. In this review, we will discuss the recent advances in our understanding of the roles of abnormal megakaryopoiesis, and the altered microenvironment in MF progression and the development of novel combined targeted therapies to disrupt the aberrant interplay between MKs, the BM microenvironment and malignant HSCs which would potentially limit the expansion of MF-HSC clones.
Collapse
Affiliation(s)
- Lilian Varricchio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
11
|
Verachi P, Gobbo F, Martelli F, Martinelli A, Sarli G, Dunbar A, Levine RL, Hoffman R, Massucci MT, Brandolini L, Giorgio C, Allegretti M, Migliaccio AR. The CXCR1/CXCR2 Inhibitor Reparixin Alters the Development of Myelofibrosis in the Gata1 low Mice. Front Oncol 2022; 12:853484. [PMID: 35392239 PMCID: PMC8982152 DOI: 10.3389/fonc.2022.853484] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/16/2022] [Indexed: 11/13/2022] Open
Abstract
A major role for human (h)CXCL8 (interleukin-8) in the pathobiology of myelofibrosis (MF) has been suggested by observations indicating that MF megakaryocytes express increased levels of hCXCL8 and that plasma levels of this cytokine in MF patients are predictive of poor patient outcomes. Here, we demonstrate that, in addition to high levels of TGF-β, the megakaryocytes from the bone marrow of the Gata1 low mouse model of myelofibrosis express high levels of murine (m)CXCL1, the murine equivalent of hCXCL8, and its receptors CXCR1 and CXCR2. Treatment with the CXCR1/R2 inhibitor, Reparixin in aged-matched Gata1 low mice demonstrated reductions in bone marrow and splenic fibrosis. Of note, the levels of fibrosis detected using two independent methods (Gomori and reticulin staining) were inversely correlated with plasma levels of Reparixin. Immunostaining of marrow sections indicated that the bone marrow from the Reparixin-treated group expressed lower levels of TGF-β1 than those expressed by the bone marrow from vehicle-treated mice while the levels of mCXCL1, and expression of CXCR1 and CXCR2, were similar to that of vehicle-treated mice. Moreover, immunofluorescence analyses performed on bone marrow sections from Gata1 low mice indicated that treatment with Reparixin induced expression of GATA1 while reducing expression of collagen III in megakaryocytes. These data suggest that in Gata1low mice, Reparixin reduces fibrosis by reducing TGF-β1 and collagen III expression while increasing GATA1 in megakaryocytes. Our results provide a preclinical rationale for further evaluation of this drug alone and in combination with current JAK inhibitor therapy for the treatment of patients with myelofibrosis.
Collapse
Affiliation(s)
- Paola Verachi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University, Bologna, Italy
| | - Francesca Gobbo
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University, Bologna, Italy
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Martinelli
- Center for Animal Experimentation and Well-Being, Istituto Superiore di Santà, Rome, Italy
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Andrew Dunbar
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ross L. Levine
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ronald Hoffman
- Division of Hematology/Oncology, Tisch Cancer Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | | | | | | | - Anna Rita Migliaccio
- Center for Integrated Biomedical Research, Campus Bio-medico, Rome, Italy
- Altius Institute for Biomedical Sciences, Seattle, WA, United States
| |
Collapse
|
12
|
Khatib-Massalha E, Méndez-Ferrer S. Megakaryocyte Diversity in Ontogeny, Functions and Cell-Cell Interactions. Front Oncol 2022; 12:840044. [PMID: 35186768 PMCID: PMC8854253 DOI: 10.3389/fonc.2022.840044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Hematopoietic stem cells (HSCs) rely on local interactions in the bone marrow (BM) microenvironment with stromal cells and other hematopoietic cells that facilitate their survival and proliferation, and also regulate their functions. HSCs and multipotent progenitor cells differentiate into lineage-specific progenitors that generate all blood and immune cells. Megakaryocytes (Mks) are hematopoietic cells responsible for producing blood platelets, which are essential for normal hemostasis and blood coagulation. Although the most prominent function of Mks is platelet production (thrombopoiesis), other increasingly recognized functions include HSC maintenance and host immune response. However, whether and how these diverse programs are executed by different Mk subpopulations remains poorly understood. This Perspective summarizes our current understanding of diversity in ontogeny, functions and cell-cell interactions. Cumulative evidence suggests that BM microenvironment dysfunction, partly caused by mutated Mks, can induce or alter the progression of a variety of hematologic malignancies, including myeloproliferative neoplasms (MPNs) and other disorders associated with tissue scarring (fibrosis). Therefore, as an example of the heterogeneous functions of Mks in malignant hematopoiesis, we will discuss the role of Mks in the onset and progression of BM fibrosis. In this regard, abnormal interactions between of Mks and other immune cells might directly contribute to fibrotic diseases. Overall, further understanding of megakaryopoiesis and how Mks interact with HSCs and immune cells has potential clinical implications for stem cell transplantation and other therapies for hematologic malignancies, as well as for treatments to stimulate platelet production and prevent thrombocytopenia.
Collapse
Affiliation(s)
- Eman Khatib-Massalha
- Wellcome-Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Simón Méndez-Ferrer
- Wellcome-Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Instituto de Biomedicina de Sevilla-IBiS, Hospitales Universitarios Virgen del Rocío y Macarena/Spanish National Research Council (CSIC)/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
13
|
Zingariello M, Verachi P, Gobbo F, Martelli F, Falchi M, Mazzarini M, Valeri M, Sarli G, Marinaccio C, Melo-Cardenas J, Crispino JD, Migliaccio AR. Resident Self-Tissue of Proinflammatory Cytokines Rather than Their Systemic Levels Correlates with Development of Myelofibrosis in Gata1low Mice. Biomolecules 2022; 12:biom12020234. [PMID: 35204735 PMCID: PMC8961549 DOI: 10.3390/biom12020234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 02/05/2023] Open
Abstract
Serum levels of inflammatory cytokines are currently investigated as prognosis markers in myelofibrosis, the most severe Philadelphia-negative myeloproliferative neoplasm. We tested this hypothesis in the Gata1low model of myelofibrosis. Gata1low mice, and age-matched wild-type littermates, were analyzed before and after disease onset. We assessed cytokine serum levels by Luminex-bead-assay and ELISA, frequency and cytokine content of stromal cells by flow cytometry, and immunohistochemistry and bone marrow (BM) localization of GFP-tagged hematopoietic stem cells (HSC) by confocal microscopy. Differences in serum levels of 32 inflammatory-cytokines between prefibrotic and fibrotic Gata1low mice and their wild-type littermates were modest. However, BM from fibrotic Gata1low mice contained higher levels of lipocalin-2, CXCL1, and TGF-β1 than wild-type BM. Although frequencies of endothelial cells, mesenchymal cells, osteoblasts, and megakaryocytes were higher than normal in Gata1low BM, the cells which expressed these cytokines the most were malignant megakaryocytes. This increased bioavailability of proinflammatory cytokines was associated with altered HSC localization: Gata1low HSC were localized in the femur diaphysis in areas surrounded by microvessels, neo-bones, and megakaryocytes, while wild-type HSC were localized in the femur epiphysis around adipocytes. In conclusion, bioavailability of inflammatory cytokines in BM, rather than blood levels, possibly by reshaping the HSC niche, correlates with myelofibrosis in Gata1low mice.
Collapse
Affiliation(s)
| | - Paola Verachi
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
| | - Francesca Gobbo
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Mario Falchi
- National Center HIV/AIDS Research, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Mazzarini
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
| | - Mauro Valeri
- Center for Animal Experimentation and Well-Being, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy;
| | | | - Johanna Melo-Cardenas
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.M.-C.); (J.D.C.)
| | - John D. Crispino
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.M.-C.); (J.D.C.)
| | - Anna Rita Migliaccio
- Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
- Center for Integrated Biomedical Research, Campus Bio-Medico, 00128 Rome, Italy
- Correspondence:
| |
Collapse
|
14
|
Neutrophil transit time and localization within the megakaryocyte define morphologically distinct forms of emperipolesis. Blood Adv 2021; 6:2081-2091. [PMID: 34872109 PMCID: PMC9006297 DOI: 10.1182/bloodadvances.2021005097] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/10/2021] [Indexed: 11/30/2022] Open
Abstract
Emperipolesis (neutrophil transit through megakaryocytes) occurs in fast and slow forms that differ morphologically. Intramegakaryocytic neutrophils reside in emperisomes and in cytoplasm near the demarcation membrane system, endoplasmic reticulum, and nucleus.
Neutrophils transit through megakaryocytes in a process termed emperipolesis, but it is unknown whether this interaction is a single type of cell-in-cell interaction or a set of distinct processes. Using a murine in vitro model, we characterized emperipolesis by live-cell spinning disk microscopy and electron microscopy. Approximately half of neutrophils exited the megakaryocyte rapidly, typically in 10 minutes or less, displaying ameboid morphology as they passed through the host cell (fast emperipolesis). The remaining neutrophils assumed a sessile morphology, most remaining within the megakaryocyte for at least 60 minutes (slow emperipolesis). These neutrophils typically localized near the megakaryocyte nucleus. By ultrastructural assessment, all internalized neutrophils remained morphologically intact. Most neutrophils resided within emperisomes, but some could be visualized exiting the emperisome to enter the cell cytoplasm. Neutrophils in the cytoplasm assumed close contact with the platelet-forming demarcation membrane system or the perinuclear endoplasmic reticulum. These findings reveal that megakaryocyte emperipolesis reflects at least 2 distinct processes differing in transit time and morphology, fast and slow emperipolesis, suggesting divergent physiologic functions.
Collapse
|
15
|
Bone marrow microenvironment of MPN cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021. [PMID: 34756245 DOI: 10.1016/bs.ircmb.2021.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
In this chapter, we will discuss the current knowledge concerning the alterations of the cellular components in the bone marrow niche in Myeloproliferative Neoplasms (MPNs), highlighting the central role of the megakaryocytes in MPN progression, and the extracellular matrix components characterizing the fibrotic bone marrow.
Collapse
|
16
|
Martelli F, Verachi P, Zingariello M, Mazzarini M, Vannucchi AM, Lonetti A, Bacci B, Sarli G, Migliaccio AR. hGATA1 Under the Control of a μLCR/β-Globin Promoter Rescues the Erythroid but Not the Megakaryocytic Phenotype Induced by the Gata1 low Mutation in Mice. Front Genet 2021; 12:720552. [PMID: 34707640 PMCID: PMC8542976 DOI: 10.3389/fgene.2021.720552] [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: 06/04/2021] [Accepted: 09/24/2021] [Indexed: 11/13/2022] Open
Abstract
The phenotype of mice carrying the Gata1low mutation that decreases expression of Gata1 in erythroid cells and megakaryocytes, includes anemia, thrombocytopenia, hematopoietic failure in bone marrow and development of extramedullary hematopoiesis in spleen. With age, these mice develop myelofibrosis, a disease sustained by alterations in stem/progenitor cells and megakaryocytes. This study analyzed the capacity of hGATA1 driven by a μLCR/β-globin promoter to rescue the phenotype induced by the Gata1low mutation in mice. Double hGATA1/Gata1low/0 mice were viable at birth with hematocrits greater than those of their Gata1low/0 littermates but platelet counts remained lower than normal. hGATA1 mRNA was expressed by progenitor and erythroid cells from double mutant mice but not by megakaryocytes analyzed in parallel. The erythroid cells from hGATA1/Gata1low/0 mice expressed greater levels of GATA1 protein and of α- and β-globin mRNA than cells from Gata1low/0 littermates and a reduced number of them was in apoptosis. By contrast, hGATA1/Gata1low/0 megakaryocytes expressed barely detectable levels of GATA1 and their expression of acetylcholinesterase, Von Willebrand factor and platelet factor 4 as well as their morphology remained altered. In comparison with Gata1+/0 littermates, Gata1low/0 mice contained significantly lower total and progenitor cell numbers in bone marrow while the number of these cells in spleen was greater than normal. The presence of hGATA1 greatly increased the total cell number in the bone marrow of Gata1low/0 mice and, although did not affect the total cell number of the spleen which remained greater than normal, it reduced the frequency of progenitor cells in this organ. The ability of hGATA1 to rescue the hematopoietic functions of the bone marrow of the double mutants was confirmed by the observation that these mice survive well splenectomy and did not develop myelofibrosis with age. These results indicate that hGATA1 under the control of µLCR/β-globin promoter is expressed in adult progenitors and erythroid cells but not in megakaryocytes rescuing the erythroid but not the megakaryocyte defect induced by the Gata1low/0 mutation.
Collapse
Affiliation(s)
- Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Paola Verachi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Maria Mazzarini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandro M Vannucchi
- Department of Clinical and Experimental Medicine, Center of Research and Innovation of Myeloproliferative neoplasms (CRIMM), AOU Careggi, University of Florence, Florence, Italy
| | - Annalisa Lonetti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Barbara Bacci
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Anna Rita Migliaccio
- Myeloproliferative Neoplasm Research Consortium, New York, NY, United States.,Department of Medicine and Surgery, University Campus Bio-Medico, Rome, Italy
| |
Collapse
|
17
|
Mukund K, Nayak P, Ashokkumar C, Rao S, Almeda J, Betancourt-Garcia MM, Sindhi R, Subramaniam S. Immune Response in Severe and Non-Severe Coronavirus Disease 2019 (COVID-19) Infection: A Mechanistic Landscape. Front Immunol 2021; 12:738073. [PMID: 34721400 PMCID: PMC8548832 DOI: 10.3389/fimmu.2021.738073] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/08/2021] [Indexed: 12/18/2022] Open
Abstract
The mechanisms underlying the immune remodeling and severity response in coronavirus disease 2019 (COVID-19) are yet to be fully elucidated. Our comprehensive integrative analyses of single-cell RNA sequencing (scRNAseq) data from four published studies, in patients with mild/moderate and severe infections, indicate a robust expansion and mobilization of the innate immune response and highlight mechanisms by which low-density neutrophils and megakaryocytes play a crucial role in the cross talk between lymphoid and myeloid lineages. We also document a marked reduction of several lymphoid cell types, particularly natural killer cells, mucosal-associated invariant T (MAIT) cells, and gamma-delta T (γδT) cells, and a robust expansion and extensive heterogeneity within plasmablasts, especially in severe COVID-19 patients. We confirm the changes in cellular abundances for certain immune cell types within a new patient cohort. While the cellular heterogeneity in COVID-19 extends across cells in both lineages, we consistently observe certain subsets respond more potently to interferon type I (IFN-I) and display increased cellular abundances across the spectrum of severity, as compared with healthy subjects. However, we identify these expanded subsets to have a more muted response to IFN-I within severe disease compared to non-severe disease. Our analyses further highlight an increased aggregation potential of the myeloid subsets, particularly monocytes, in COVID-19. Finally, we provide detailed mechanistic insights into the interaction between lymphoid and myeloid lineages, which contributes to the multisystemic phenotype of COVID-19, distinguishing severe from non-severe responses.
Collapse
Affiliation(s)
- Kavitha Mukund
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| | - Priya Nayak
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| | - Chethan Ashokkumar
- Plexision Inc., Pittsburgh, PA, United States
- Hillman Center for Pediatric Transplantation, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sohail Rao
- DHR Health and DHR Health Institute for Research and Development, Edinburg, TX, United States
| | - Jose Almeda
- DHR Health and DHR Health Institute for Research and Development, Edinburg, TX, United States
| | | | - Rakesh Sindhi
- Plexision Inc., Pittsburgh, PA, United States
- Hillman Center for Pediatric Transplantation, University of Pittsburgh, Pittsburgh, PA, United States
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
18
|
Kiem D, Wagner S, Magnes T, Egle A, Greil R, Melchardt T. The Role of Neutrophilic Granulocytes in Philadelphia Chromosome Negative Myeloproliferative Neoplasms. Int J Mol Sci 2021; 22:ijms22179555. [PMID: 34502471 PMCID: PMC8431305 DOI: 10.3390/ijms22179555] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 11/25/2022] Open
Abstract
Philadelphia chromosome negative myeloproliferative neoplasms (MPN) are composed of polycythemia vera (PV), essential thrombocytosis (ET), and primary myelofibrosis (PMF). The clinical picture is determined by constitutional symptoms and complications, including arterial and venous thromboembolic or hemorrhagic events. MPNs are characterized by mutations in JAK2, MPL, or CALR, with additional mutations leading to an expansion of myeloid cell lineages and, in PMF, to marrow fibrosis and cytopenias. Chronic inflammation impacting the initiation and expansion of disease in a major way has been described. Neutrophilic granulocytes play a major role in the pathogenesis of thromboembolic events via the secretion of inflammatory markers, as well as via interaction with thrombocytes and the endothelium. In this review, we discuss the molecular biology underlying myeloproliferative neoplasms and point out the central role of leukocytosis and, specifically, neutrophilic granulocytes in this group of disorders.
Collapse
Affiliation(s)
- Dominik Kiem
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.K.); (S.W.); (T.M.); (A.E.); (R.G.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Sandro Wagner
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.K.); (S.W.); (T.M.); (A.E.); (R.G.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Teresa Magnes
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.K.); (S.W.); (T.M.); (A.E.); (R.G.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Alexander Egle
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.K.); (S.W.); (T.M.); (A.E.); (R.G.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
- Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), 5020 Salzburg, Austria
| | - Richard Greil
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.K.); (S.W.); (T.M.); (A.E.); (R.G.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
- Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), 5020 Salzburg, Austria
| | - Thomas Melchardt
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.K.); (S.W.); (T.M.); (A.E.); (R.G.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
- Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), 5020 Salzburg, Austria
- Correspondence: ; Tel.: +43-57255-25801
| |
Collapse
|
19
|
Suljević D, Hamzić A, Islamagić E, Fejzić E, Alijagić A. Haematopoietic thrombocyte precursors in rat femoral and sternal bone marrow. BULGARIAN JOURNAL OF VETERINARY MEDICINE 2021. [DOI: 10.15547/bjvm.2019-0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This research presents the first findings on thrombopoiesis for Wistar rats. Haemopoietic cells from the femur and the sternum were analysed by light microscopy in combination with infrared and near-ultraviolet light for fine cytoplasmic structure analysis. Five main types of thrombocyte precursor cells were identified in the bone marrow samples: megakaryoblast, promegakaryocyte and megakaryocyte (basophilic, acidophilic and thrombocytogenic). More intensive thrombopoiesis and morphologically differentiated cells were found in sternum samples.
Collapse
|
20
|
Zingariello M, Rosti V, Vannucchi AM, Guglielmelli P, Mazzarini M, Barosi G, Genova ML, Migliaccio AR. Shared and Distinctive Ultrastructural Abnormalities Expressed by Megakaryocytes in Bone Marrow and Spleen From Patients With Myelofibrosis. Front Oncol 2020; 10:584541. [PMID: 33312951 PMCID: PMC7701330 DOI: 10.3389/fonc.2020.584541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/19/2020] [Indexed: 12/21/2022] Open
Abstract
Numerous studies have documented ultrastructural abnormalities in malignant megakaryocytes from bone marrow (BM) of myelofibrosis patients but the morphology of these cells in spleen, an important extramedullary site in this disease, was not investigated as yet. By transmission-electron microscopy, we compared the ultrastructural features of megakaryocytes from BM and spleen of myelofibrosis patients and healthy controls. The number of megakaryocytes was markedly increased in both BM and spleen. However, while most of BM megakaryocytes are immature, those from spleen appear mature with well-developed demarcation membrane systems (DMS) and platelet territories and are surrounded by platelets. In BM megakaryocytes, paucity of DMS is associated with plasma (thick with protrusions) and nuclear (dilated with large pores) membrane abnormalities and presence of numerous glycosomes, suggesting a skewed metabolism toward insoluble polyglucosan accumulation. By contrast, the membranes of the megakaryocytes from the spleen were normal but these cells show mitochondria with reduced crests, suggesting deficient aerobic energy-metabolism. These distinctive morphological features suggest that malignant megakaryocytes from BM and spleen express distinctive metabolic impairments that may play different roles in the pathogenesis of myelofibrosis.
Collapse
Affiliation(s)
- Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry Biotechnology and Advanced Diagnosis, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alessandro M Vannucchi
- CRIMM; Center Research and Innovation of Myeloproliferative Neoplasms, AOUC, University of Florence, Florence, Italy
| | - Paola Guglielmelli
- CRIMM; Center Research and Innovation of Myeloproliferative Neoplasms, AOUC, University of Florence, Florence, Italy
| | - Maria Mazzarini
- Biomedical and Neuromotor Sciences, Alma Mater University Bologna, Bologna, Italy
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry Biotechnology and Advanced Diagnosis, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Maria Luisa Genova
- Biomedical and Neuromotor Sciences, Alma Mater University Bologna, Bologna, Italy
| | - Anna Rita Migliaccio
- Biomedical and Neuromotor Sciences, Alma Mater University Bologna, Bologna, Italy.,Myeloproliferative Neoplasm-Research Consortium, New York, NY, United States
| |
Collapse
|
21
|
Schino M, Fiorentino V, Rossi E, Betti S, Di Cecca M, Ranucci V, Chiusolo P, Martini M, De Stefano V, Larocca LM. Bone marrow megakaryocytic activation predicts fibrotic evolution of Philadelphia-negative myeloproliferative neoplasms. Haematologica 2020; 106:3162-3169. [PMID: 33543865 PMCID: PMC8634198 DOI: 10.3324/haematol.2020.264143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Indexed: 11/13/2022] Open
Abstract
Philadelphia-negative chronic myeloproliferative neoplasms (MPN) have been traditionally considered as indistinctly slowly progressing conditions; recent evidence proves that a subset of cases have a rapid evolution, so that MPN prognosis needs to be personalized. We identified a new morphological parameter, defined as megakaryocytic activation (M-ACT) based on the coexistence of megakaryocytic emperipolesis, megakaryocytes (MK) cluster formation and evidence of arrangement of collagen fibers around the perimeter of MK. We retrospectively analyzed the bone marrow biopsy of two MPN cohorts of patients with polycythemia (PV) (n=64) and non-PV patients (including essential thrombocythemia, and early/prefibrotic primary myelofibrosis [PMF]) (n=222). M-ACT showed a significant correlation with splenomegaly, white blood cell count, and lactate dehydrogenase serum levels in both groups, with JAK2 V617F allele burden in PV patients, and with CALR mutations, and platelet count in non-PV patients. Progression-free survival, defined as PV-to-secondary MF progression and non-PV-to-overt PMF, was worse in both PV and early/prefibrotic PMF patients with M-ACT in comparison to those without M-ACT (P<0.0001). Interestingly, M-ACT was not found in the subgroup of essential thrombocythemia patients. In conclusion, M-ACT can be helpful in the differential diagnosis of MPN and can represent a new morphologic parameter with a predictive value for progression of MPN.
Collapse
Affiliation(s)
- Mattia Schino
- Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome.
| | - Vincenzo Fiorentino
- Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome.
| | - Elena Rossi
- Department of Radiological and Hematological Sciences, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome.
| | - Silvia Betti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome.
| | - Monica Di Cecca
- Department of Radiological and Hematological Sciences, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome.
| | - Valentina Ranucci
- Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome.
| | - Patrizia Chiusolo
- Department of Radiological and Hematological Sciences, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome.
| | - Maurizio Martini
- Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome.
| | - Valerio De Stefano
- Department of Radiological and Hematological Sciences, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome.
| | - Luigi Maria Larocca
- Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome.
| |
Collapse
|
22
|
Bar-Natan M, Hoffman R. New insights into the causes of thrombotic events in patients with myeloproliferative neoplasms raise the possibility of novel therapeutic approaches. Haematologica 2019; 104:3-6. [PMID: 30598493 DOI: 10.3324/haematol.2018.205989] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Michal Bar-Natan
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
23
|
Wang X, Li Y, Li J, Li L, Zhu H, Chen H, Kong R, Wang G, Wang Y, Hu J, Sun B. Cell-in-Cell Phenomenon and Its Relationship With Tumor Microenvironment and Tumor Progression: A Review. Front Cell Dev Biol 2019; 7:311. [PMID: 31850347 PMCID: PMC6901391 DOI: 10.3389/fcell.2019.00311] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/14/2019] [Indexed: 12/17/2022] Open
Abstract
The term cell-in-cell, morphologically, refers to the presence of one cell within another. This phenomenon can occur in tumors but also among non-tumor cells. The cell-in-cell phenomenon was first observed 100 years ago, and it has since been found in a variety of tumor types. Recently, increasing attention has been paid to this phenomenon and the underlying mechanism has gradually been elucidated. There are three main related process: cannibalism, emperipolesis, and entosis. These processes are affected by many factors, including the tumor microenvironment, mitosis, and genetic factors. There is considerable evidence to suggest that the cell-in-cell phenomenon is associated with the prognosis of cancers, and it promotes tumor progression in most situations. Notably, in pancreatic cancer, the cell-in-cell phenomenon is associated with reduced metastasis, which is the opposite of what happens in other tumor types. Thus, it can also inhibit tumor progression. Studies show that cell-in-cell structure formation is affected by the tumor microenvironment, and that it may lead to changes in cellular characteristics. In this review, we summarize the different cell-in-cell processes and discuss their role in tumor progression and how they are regulated by different mechanisms.
Collapse
Affiliation(s)
- Xinlong Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yilong Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiating Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hong Zhu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hua Chen
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Kong
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yongwei Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jisheng Hu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
24
|
Abstract
Histology of bone marrow routinely identifies megakaryocytes that enclose neutrophils and other hematopoietic cells, a phenomenon termed emperipolesis. Preserved across mammalian species and enhanced with systemic inflammation and platelet demand, the nature and significance of emperipolesis remain largely unexplored. Recent advances demonstrate that emperipolesis is in fact a distinct form of cell-in-cell interaction. Following integrin-mediated attachment, megakaryocytes and neutrophils both actively drive entry via cytoskeletal rearrangement. Neutrophils enter a vacuole termed the emperisome which then releases them directly into the megakaryocyte cytoplasm. From this surprising location, neutrophils fuse with the demarcation membrane system to pass membrane to circulating platelets, enhancing the efficiency of thrombocytogenesis. Neutrophils then egress intact, carrying megakaryocyte membrane and potentially other cell components along with them. In this review, we summarize what is known about this intriguing cell-in-cell interaction and discuss potential roles for emperipolesis in megakaryocyte, platelet and neutrophil biology.
Collapse
Affiliation(s)
- Pierre Cunin
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School , Boston, MA, USA
| | - Peter A Nigrovic
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School , Boston, MA, USA.,Department of Medicine, Division of Immunology, Boston Children's Hospital, Harvard Medical School , Boston, MA, USA
| |
Collapse
|
25
|
Zingariello M, Martelli F, Verachi P, Bardelli C, Gobbo F, Mazzarini M, Migliaccio AR. Novel targets to cure primary myelofibrosis from studies on Gata1 low mice. IUBMB Life 2019; 72:131-141. [PMID: 31749302 DOI: 10.1002/iub.2198] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/24/2019] [Indexed: 01/06/2023]
Abstract
In 2002, we discovered that mice carrying the hypomorphic Gata1low mutation that reduces expression of the transcription factor GATA1 in megakaryocytes (Gata1low mice) develop myelofibrosis, a phenotype that recapitulates the features of primary myelofibrosis (PMF), the most severe of the Philadelphia-negative myeloproliferative neoplasms (MPNs). At that time, this discovery had a great impact on the field because mutations driving the development of PMF had yet to be discovered. Later studies identified that PMF, as the others MPNs, is associated with mutations activating the thrombopoietin/JAK2 axis raising great hope that JAK inhibitors may be effective to treat the disease. Unfortunately, ruxolitinib, the JAK1/2 inhibitor approved by FDA and EMEA for PMF, ameliorates symptoms but does not improve the natural course of the disease, and the cure of PMF is still an unmet clinical need. Although GATA1 is not mutated in PMF, reduced GATA1 content in megakaryocytes as a consequence of ribosomal deficiency is a hallmark of myelofibrosis (both in humans and mouse models) and, in fact, a driving event in the disease. Conversely, mice carrying the hypomorphic Gata1low mutation express an activated TPO/JAK2 pathway and partially respond to JAK inhibitors in a fashion similar to PMF patients (reduction of spleen size but limited improvement of the natural history of the disease). These observations cross-validated Gata1low mice as a bona fide animal model for PMF and prompted the use of this model to identify abnormalities that might be targeted to cure the disease. We will summarize here data generated in Gata1low mice indicating that the TGF-β/P-selectin axis is abnormal in PMF and represents a novel target for its treatment.
Collapse
Affiliation(s)
- Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | | | - Paola Verachi
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Claudio Bardelli
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Francesca Gobbo
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Maria Mazzarini
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Anna Rita Migliaccio
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy.,Myeloproliferative Neoplasms Research Consortium, New York, New York
| |
Collapse
|
26
|
Cruz NM, Gergis U, Silver RT. Myelofibrosis: best practices, controversies and 2019 update. Expert Rev Hematol 2019; 13:71-84. [PMID: 31709843 DOI: 10.1080/17474086.2020.1691519] [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: 10/25/2022]
Abstract
Introduction: Recent advances in the prognostic scheme and treatment of primary and secondary myelofibrosis (MF) have resulted in an overwhelming amount of clinical information to assimilate. The authors believe a comprehensive review that summarizes the most recent published literature, could serve as guidelines for the practicing hematologist.Areas covered: The authors provide a summary of landmark articles regarding epidemiology, symptoms, and pathogenesis of disease. The authors conducted a systematic literature review to answer questions regarding differences between primary myelofibrosis (PMF) and secondary myelofibrosis (SMF), appropriate use and selection of the current risk-stratification models, early versus late treatment of MF and current practices in allogeneic hematopoietic stem cell transplantation (allo-HCT) for MF. The authors conclude the article with their clinical opinion based on their experience and literature review. The purpose of this article is to identify current practices, address any variation, identify and investigate conflicting results and produce statements to guide decision-making.Expert opinion: In this section, the authors advocate for and provide examples of a standardized way of incorporating future discoveries in the pathogenesis and risk-stratification models of MF. They also discuss the importance of using only one risk-stratification model for PMF and one for SMF and their reasoning for early instead of late treatment of MF.
Collapse
Affiliation(s)
- Nicole M Cruz
- Division of Hematology & Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Usama Gergis
- Division of Hematology & Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Richard T Silver
- Division of Hematology & Medical Oncology, Weill Cornell Medicine, New York, NY, USA.,Richard T. Silver, M.D. Myeloproliferative Neoplasms (MPN) Center, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
27
|
Monocytic Myeloid Derived Suppressor Cells in Hematological Malignancies. Int J Mol Sci 2019; 20:ijms20215459. [PMID: 31683978 PMCID: PMC6862591 DOI: 10.3390/ijms20215459] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/27/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
In the era of novel agents and immunotherapies in solid and liquid tumors, there is an emerging need to understand the cross-talk between the neoplastic cells, the host immune system, and the microenvironment to mitigate proliferation, survival, migration and resistance to drugs. In the microenvironment of hematological tumors there are cells belonging to the normal bone marrow, extracellular matrix proteins, adhesion molecules, cytokines, and growth factors produced by both stromal cells and neoplastic cells themselves. In this context, myeloid suppressor cells are an emerging sub-population of regulatory myeloid cells at different stages of differentiation involved in cancer progression and chronic inflammation. In this review, monocytic myeloid derived suppressor cells and their potential clinical implications are discussed to give a comprehensive vision of their contribution to lymphoproliferative and myeloid disorders.
Collapse
|
28
|
Cunin P, Bouslama R, Machlus KR, Martínez-Bonet M, Lee PY, Wactor A, Nelson-Maney N, Morris A, Guo L, Weyrich A, Sola-Visner M, Boilard E, Italiano JE, Nigrovic PA. Megakaryocyte emperipolesis mediates membrane transfer from intracytoplasmic neutrophils to platelets. eLife 2019; 8:e44031. [PMID: 31042146 PMCID: PMC6494422 DOI: 10.7554/elife.44031] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/12/2019] [Indexed: 01/06/2023] Open
Abstract
Bone marrow megakaryocytes engulf neutrophils in a phenomenon termed emperipolesis. We show here that emperipolesis is a dynamic process mediated actively by both lineages, in part through the β2-integrin/ICAM-1/ezrin pathway. Tethered neutrophils enter in membrane-bound vesicles before penetrating into the megakaryocyte cytoplasm. Intracytoplasmic neutrophils develop membrane contiguity with the demarcation membrane system, thereby transferring membrane to the megakaryocyte and to daughter platelets. This phenomenon occurs in otherwise unmanipulated murine marrow in vivo, resulting in circulating platelets that bear membrane from non-megakaryocytic hematopoietic donors. Transit through megakaryocytes can be completed as rapidly as minutes, after which neutrophils egress intact. Emperipolesis is amplified in models of murine inflammation associated with platelet overproduction, contributing to platelet production in vitro and in vivo. These findings identify emperipolesis as a new cell-in-cell interaction that enables neutrophils and potentially other cells passing through the megakaryocyte cytoplasm to modulate the production and membrane content of platelets.
Collapse
Affiliation(s)
- Pierre Cunin
- Department of Medicine, Division of Rheumatology, Immunology and AllergyBrigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Rim Bouslama
- Department of Medicine, Division of Rheumatology, Immunology and AllergyBrigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Kellie R Machlus
- Department of Medicine, Hematology DivisionBrigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Marta Martínez-Bonet
- Department of Medicine, Division of Rheumatology, Immunology and AllergyBrigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Pui Y Lee
- Department of Medicine, Division of Rheumatology, Immunology and AllergyBrigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
- Department of Medicine, Division of ImmunologyBoston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Alexandra Wactor
- Department of Medicine, Division of Rheumatology, Immunology and AllergyBrigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Nathan Nelson-Maney
- Department of Medicine, Division of Rheumatology, Immunology and AllergyBrigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Allyn Morris
- Department of Medicine, Division of Rheumatology, Immunology and AllergyBrigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Li Guo
- Program in Molecular Medicine and Department of Internal MedicineUniversity of UtahSalt Lake CityUnited States
| | - Andrew Weyrich
- Program in Molecular Medicine and Department of Internal MedicineUniversity of UtahSalt Lake CityUnited States
| | - Martha Sola-Visner
- Department of NeonatologyBoston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Eric Boilard
- Centre de Recherche en Rhumatologie et ImmunologieCentre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université LavalQuébecCanada
| | - Joseph E Italiano
- Department of Medicine, Hematology DivisionBrigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
- Vascular Biology Program, Department of SurgeryBoston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Peter A Nigrovic
- Department of Medicine, Division of Rheumatology, Immunology and AllergyBrigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
- Department of Medicine, Division of ImmunologyBoston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
29
|
Emperipolesis: Sternal and Femoral Microenvironment Induces Megakaryiocyte Emperipolesis in the Wistar Strain. MACEDONIAN VETERINARY REVIEW 2019. [DOI: 10.2478/macvetrev-2019-0012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Emperipolesis is considered a physiological phenomena often present in various pathophysiological conditions, but its etiology is still unknown. In this study, we analyzed the number of megakaryocytes and the percentage of emperipoletic cells in the sternal and femoral bone marrow of Wistar rats. Five types in the thrombopoiesis lineage (megakaryoblasts, promegakaryocytes and megakaryocytes - acidophilic, basophilic and thrombocytogenic) were determined. Except for basophilic megakaryocytes, significant differences were found for number of thrombopoietic cells in the sternal and femoral bone marrow. A larger number of thrombocytogenic megakaryocytes were present in the sternal bone marrow. Emperipoletic cells were significantly present in the femoral compared to the sternal bone marrow. Emperipolesis was typical for lymphocytes and neutrophils individually, while emperipolesis with two or more cells within thrombopoietic cell was also present (1-7 %) and significant differences between the sternal and femoral bone marrow were detected. Emperipolesis was found in all analysed rats and it most commonly occured within mature megakaryocytes and rarely megakaryoblasts, while it was not recorded in the promegakaryocytes. The high incidence of megakaryocytes with emperopolesis in rats could be a consequence of “normal” cell retention in the cytoplasm of megakaryocytes while passing blood cells to circulation or related to haematopoietic response due to high incidence of inbreeding.
Collapse
|
30
|
Cunin P, Nigrovic PA. Megakaryocytes as immune cells. J Leukoc Biol 2019; 105:1111-1121. [PMID: 30645026 DOI: 10.1002/jlb.mr0718-261rr] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 12/19/2022] Open
Abstract
Platelets play well-recognized roles in inflammation, but their cell of origin-the megakaryocyte-is not typically considered an immune lineage. Megakaryocytes are large polyploid cells most commonly identified in bone marrow. Egress via sinusoids enables migration to the pulmonary capillary bed, where elaboration of platelets can continue. Beyond receptors involved in hemostasis and thrombosis, megakaryocytes express receptors that confer immune sensing capacity, including TLRs and Fc-γ receptors. They control the proliferation of hematopoietic cells, facilitate neutrophil egress from marrow, possess the capacity to cross-present antigen, and can promote systemic inflammation through microparticles rich in IL-1. Megakaryocytes internalize other hematopoietic lineages, especially neutrophils, in an intriguing cell-in-cell interaction termed emperipolesis. Together, these observations implicate megakaryocytes as direct participants in inflammation and immunity.
Collapse
Affiliation(s)
- Pierre Cunin
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter A Nigrovic
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Medicine, Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
31
|
|
32
|
Megakaryocyte Contribution to Bone Marrow Fibrosis: many Arrows in the Quiver. Mediterr J Hematol Infect Dis 2018; 10:e2018068. [PMID: 30416700 PMCID: PMC6223581 DOI: 10.4084/mjhid.2018.068] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/23/2018] [Indexed: 01/14/2023] Open
Abstract
In Primary Myelofibrosis (PMF), megakaryocyte dysplasia/hyperplasia determines the release of inflammatory cytokines that, in turn, stimulate stromal cells and induce bone marrow fibrosis. The pathogenic mechanism and the cells responsible for progression to bone marrow fibrosis in PMF are not completely understood. This review article aims to provide an overview of the crucial role of megakaryocytes in myelofibrosis by discussing the role and the altered secretion of megakaryocyte-derived soluble factors, enzymes and extracellular matrices that are known to induce bone marrow fibrosis.
Collapse
|
33
|
Olcay L, Ünal Ş, Onay H, Erdemli E, Öztürk A, Billur D, Metin A, Okur H, Yıldırmak Y, Büyükaşık Y, İkincioğulları A, Falay M, Özet G, Yetgin S. Both Granulocytic and Non-Granulocytic Blood Cells Are Affected in Patients with Severe Congenital Neutropenia and Their Non-Neutropenic Family Members: An Evaluation of Morphology, Function, and Cell Death. Turk J Haematol 2018; 35:229-259. [PMID: 30040071 PMCID: PMC6256814 DOI: 10.4274/tjh.2017.0160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Objective: To examine granulocytic and non-granulocytic cells in children with severe congenital neutropenia (SCN) and their non-neutropenic parents. Materials and Methods: Fifteen patients with SCN and 21 non-neutropenic parents were evaluated for a) CD95, CD95 ligand, annexin V, propidium iodide, cell cycle, and lymphocyte subsets by flow cytometry; b) rapid cell senescence (of leukocytes) by senescence-associated β-galactosidase stain; c) aggregation tests by aggregometer; d) in vitro bleeding time by PFA-100 instrument; e) mepacrine-labeled dense granule number of thrombocytes by fluorescence microscope; and f) hematomorphology by light and electron microscope. HAX1, ELANE, G6PC3, CSF3R, and JAGN1 mutations associated with SCN were studied in patients and several parents. Results: Significant increase in apoptosis and secondary necrosis in monocytes, lymphocytes, and granulocytes of the patients and parents was detected, irrespective of the mutation type. CD95 and CD95 ligand results implied that apoptosis was non-CD95-mediated. Leukocytes of 25%, 12.5%, and 0% of patients, parents, and controls showed rapid cell senescence. The cell cycle analysis testable in four cases showed G1 arrest and apoptosis in lymphocytes of three. The patients had HAX1 (n=6), ELANE (n=2), G6PC3 (n=2), and unidentified (n=5) mutations. The CD3, CD4, and NK lymphocytes were below normal levels in 16.6%, 8.3%, and 36.4% of the patients and in 0%, 0%, and 15.4% of the parents (controls: 0%, 0%, 5.6%). The thrombocytes aggregated at low rates, dense granule number/thrombocyte ratio was low, and in vitro bleeding time was prolonged in 37.5%-66.6% of patients and 33.3%-63.2% of parents (vs. 0% in controls). Under electron and/or light microscope, the neutrophils, monocytes, lymphocytes, and thrombocytes in the peripheral blood of both patients and parents were dysplastic and the bone marrow of patients revealed increased phagocytic activity, dysmegakaryopoiesis, and necrotic and apoptotic cells. Ultrastructurally, thrombocyte adhesion, aggregation, and release were inadequate. Conclusion: In cases of SCN, patients’ pluripotent hematopoietic stem cells and their non-neutropenic parents are both affected irrespective of the genetic defect.
Collapse
Affiliation(s)
- Lale Olcay
- Ankara Oncology Training and Research Hospital, Clinic of Pediatric Hematology, Ankara, Turkey
| | - Şule Ünal
- Hacettepe University Faculty of Medicine, İhsan Doğramacı Children’s Hospital, Clinic of Pediatric Hematology, Ankara, Turkey
| | - Hüseyin Onay
- Ege University Faculty of Medicine, Department of Medical Genetics, İzmir, Turkey
| | - Esra Erdemli
- Ankara University Faculty of Medicine, Department of Histology Embryology, Ankara, Turkey
| | - Ayşenur Öztürk
- Ankara University Faculty of Medicine, Department of Pediatric Molecular Genetics, Ankara, Turkey
| | - Deniz Billur
- Ankara University Faculty of Medicine, Department of Histology Embryology, Ankara, Turkey
| | - Ayşe Metin
- Ankara Children’s Hematology Oncology Training and Research Hospital, Clinic of Pediatric Immunology, Ankara, Turkey
| | - Hamza Okur
- Hacettepe University Faculty of Medicine, İhsan Doğramacı Children’s Hospital, Clinic of Pediatric Hematology, Ankara, Turkey
| | - Yıldız Yıldırmak
- Şişli Etfal Children’s Training and Research Hospital, Clinic of Pediatric Hematology, İstanbul, Turkey
| | - Yahya Büyükaşık
- Hacettepe University Faculty of Medicine, Department of Internal Medicine, Unit of Hematology, Ankara, Turkey
| | - Aydan İkincioğulları
- Ankara University Faculty of Medicine, Department of Pediatric Immunology and Allergy and Pediatric Molecular Genetics, Ankara, Turkey
| | - Mesude Falay
- Ankara Numune Training and Research Hospital, Clinic of Hematology, Ankara, Turkey
| | - Gülsüm Özet
- Ankara Numune Training and Research Hospital, Clinic of Hematology, Ankara, Turkey,Yıldırım Beyazıt University Faculty of Medicine, Department of Internal Medicine, Clinic of Hematology, Ankara, Turkey
| | - Sevgi Yetgin
- Hacettepe University Faculty of Medicine, İhsan Doğramacı Children’s Hospital, Clinic of Pediatric Hematology, Ankara, Turkey
| |
Collapse
|
34
|
Mapping the physical network of cellular interactions. Nat Methods 2018; 15:547-553. [PMID: 29786092 DOI: 10.1038/s41592-018-0009-z] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 03/20/2018] [Indexed: 01/03/2023]
Abstract
A cell's function is influenced by the environment, or niche, in which it resides. Studies of niches usually require assumptions about the cell types present, which impedes the discovery of new cell types or interactions. Here we describe ProximID, an approach for building a cellular network based on physical cell interaction and single-cell mRNA sequencing, and show that it can be used to discover new preferential cellular interactions without prior knowledge of component cell types. ProximID found specific interactions between megakaryocytes and mature neutrophils and between plasma cells and myeloblasts and/or promyelocytes (precursors of neutrophils) in mouse bone marrow, and it identified a Tac1+ enteroendocrine cell-Lgr5+ stem cell interaction in small intestine crypts. This strategy can be used to discover new niches or preferential interactions in a variety of organs.
Collapse
|
35
|
Ling T, Crispino JD, Zingariello M, Martelli F, Migliaccio AR. GATA1 insufficiencies in primary myelofibrosis and other hematopoietic disorders: consequences for therapy. Expert Rev Hematol 2018; 11:169-184. [PMID: 29400094 PMCID: PMC6108178 DOI: 10.1080/17474086.2018.1436965] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION GATA1, the founding member of a family of transcription factors, plays important roles in the development of hematopoietic cells of several lineages. Although loss of GATA1 has been known to impair hematopoiesis in animal models for nearly 25 years, the link between GATA1 defects and human blood diseases has only recently been realized. Areas covered: Here the current understanding of the functions of GATA1 in normal hematopoiesis and how it is altered in disease is reviewed. GATA1 is indispensable mainly for erythroid and megakaryocyte differentiation. In erythroid cells, GATA1 regulates early stages of differentiation, and its deficiency results in apoptosis. In megakaryocytes, GATA1 controls terminal maturation and its deficiency induces proliferation. GATA1 alterations are often found in diseases involving these two lineages, such as congenital erythroid and/or megakaryocyte deficiencies, including Diamond Blackfan Anemia (DBA), and acquired neoplasms, such as acute megakaryocytic leukemia (AMKL) and the myeloproliferative neoplasms (MPNs). Expert commentary: Since the first discovery of GATA1 mutations in AMKL, the number of diseases that are associated with impaired GATA1 function has increased to include DBA and MPNs. With respect to the latter, we are only just now appreciating the link between enhanced JAK/STAT signaling, GATA1 deficiency and disease pathogenesis.
Collapse
Affiliation(s)
- Te Ling
- Division of Hematology/Oncology, Northwestern University, Chicago, IL, USA
| | - John D. Crispino
- Division of Hematology/Oncology, Northwestern University, Chicago, IL, USA
| | | | - Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Roma, Italy
| | - Anna Rita Migliaccio
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, Bologna, Italy
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
| |
Collapse
|
36
|
Extensive Megakaryocytic Emperipolesis in an Adult Male with Immune Thrombocytopenia. Indian J Hematol Blood Transfus 2018; 34:158-160. [PMID: 29398820 DOI: 10.1007/s12288-017-0834-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 05/19/2017] [Indexed: 10/19/2022] Open
|
37
|
Goyal M, Thekkelakayil ST, Gupta A. Megakaryocytic Emperipolesis Associated with Thrombocytopenia: Causative or Coincidence? Turk J Haematol 2017; 34:370-371. [PMID: 28832006 PMCID: PMC5774352 DOI: 10.4274/tjh.2017.0211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- Manu Goyal
- AmPath Hyderabad Hospital, Clinics of Hematopathology and Molecular Hematopathology, Telangana, India
| | | | - Anurag Gupta
- AmPath Hyderabad Hospital, Clinic of Hematopathology, Telangana, India.,AmPath Hyderabad Hospital, Clinic of Cytogenetics, Telangana, India
| |
Collapse
|
38
|
Filippini A, Bonini D, Lacoux C, Pacini L, Zingariello M, Sancillo L, Bosisio D, Salvi V, Mingardi J, La Via L, Zalfa F, Bagni C, Barbon A. Absence of the Fragile X Mental Retardation Protein results in defects of RNA editing of neuronal mRNAs in mouse. RNA Biol 2017. [PMID: 28640668 PMCID: PMC5785225 DOI: 10.1080/15476286.2017.1338232] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
The fragile X syndrome (FXS), the most common form of inherited intellectual disability, is due to the absence of FMRP, a protein regulating RNA metabolism. Recently, an unexpected function of FMRP in modulating the activity of Adenosine Deaminase Acting on RNA (ADAR) enzymes has been reported both in Drosophila and Zebrafish. ADARs are RNA-binding proteins that increase transcriptional complexity through a post-transcriptional mechanism called RNA editing. To evaluate the ADAR2-FMRP interaction in mammals we analyzed several RNA editing re-coding sites in the fmr1 knockout (KO) mice. Ex vivo and in vitro analysis revealed that absence of FMRP leads to an increase in the editing levels of brain specific mRNAs, indicating that FMRP might act as an inhibitor of editing activity. Proximity Ligation Assay (PLA) in mouse primary cortical neurons and in non-neuronal cells revealed that ADAR2 and FMRP co-localize in the nucleus. The ADAR2-FMRP co-localization was further observed by double-immunogold Electron Microscopy (EM) in the hippocampus. Moreover, ADAR2-FMRP interaction appeared to be RNA independent. Because changes in the editing pattern are associated with neuropsychiatric and neurodevelopmental disorders, we propose that the increased editing observed in the fmr1-KO mice might contribute to the FXS molecular phenotypes.
Collapse
Affiliation(s)
- Alice Filippini
- a Biology and Genetic Division; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| | - Daniela Bonini
- a Biology and Genetic Division; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| | - Caroline Lacoux
- b Department of Biomedicine and Prevention , University of Rome Tor Vergata , Rome , Italy
| | - Laura Pacini
- b Department of Biomedicine and Prevention , University of Rome Tor Vergata , Rome , Italy
| | - Maria Zingariello
- c Department of Medicine , Campus Bio-Medico University , via Álvaro del Portillo 21, Rome , Italy
| | - Laura Sancillo
- d Department of Medicine and Aging Sciences, Section of Human Morphology , University G. D'Annunzio of Chieti-Pescara , Chieti , Italy
| | - Daniela Bosisio
- e Immunology Unit; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| | - Valentina Salvi
- e Immunology Unit; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| | - Jessica Mingardi
- a Biology and Genetic Division; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| | - Luca La Via
- a Biology and Genetic Division; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| | - Francesca Zalfa
- c Department of Medicine , Campus Bio-Medico University , via Álvaro del Portillo 21, Rome , Italy
| | - Claudia Bagni
- b Department of Biomedicine and Prevention , University of Rome Tor Vergata , Rome , Italy.,f VIB Center for the Biology of Disease and Center for Human Genetics , Leuven , Belgium.,g Department of Fundamental Neuroscience , University of Lausanne , Lausanne , Switzerland
| | - Alessandro Barbon
- a Biology and Genetic Division; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| |
Collapse
|
39
|
Hasegawa A, Shimizu R. GATA1 Activity Governed by Configurations of cis-Acting Elements. Front Oncol 2017; 6:269. [PMID: 28119852 PMCID: PMC5220053 DOI: 10.3389/fonc.2016.00269] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/19/2016] [Indexed: 01/19/2023] Open
Abstract
The transcription factor GATA1 regulates the expression of essential erythroid and megakaryocytic differentiation genes through binding to the DNA consensus sequence WGATAR. The GATA1 protein has four functional domains, including two centrally located zinc-finger domains and two transactivation domains at the N- and C-termini. These functional domains play characteristic roles in the elaborate regulation of diversified GATA1 target genes, each of which exhibits a unique expression profile. Three types of GATA1-related hematological malignancies have been reported. One is a structural mutation in the GATA1 gene, resulting in the production of a short form of GATA1 that lacks the N-terminal transactivation domain and is found in Down syndrome-related acute megakaryocytic leukemia. The other two are cis-acting regulatory mutations affecting expression of the Gata1 gene, which have been shown to cause acute erythroblastic leukemia and myelofibrosis in mice. Therefore, imbalanced gene regulation caused by qualitative and quantitative changes in GATA1 is thought to be involved in specific hematological disease pathogenesis. In the present review, we discuss recent advances in understanding the mechanisms of differential transcriptional regulation by GATA1 during erythroid differentiation, with special reference to the binding kinetics of GATA1 at conformation-specific binding sites.
Collapse
Affiliation(s)
- Atsushi Hasegawa
- Department of Molecular Hematology, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Ritsuko Shimizu
- Department of Molecular Hematology, Tohoku University Graduate School of Medicine, Sendai, Japan; Medical Mega-Bank Organization, Tohoku University, Sendai, Japan
| |
Collapse
|
40
|
Gupta N, Jadhav K, Shah V. Emperipolesis, entosis and cell cannibalism: Demystifying the cloud. J Oral Maxillofac Pathol 2017; 21:92-98. [PMID: 28479694 PMCID: PMC5406827 DOI: 10.4103/0973-029x.203763] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
There are intense published data in literature related to cell engulfment phenomena such as emperipolesis, entosis and cell cannibalism. All these are closely related phenomena with a very fine line of differences. Its correct identification has a significant diagnostic and prognostic value. After extensive literature search, a gap of knowledge was found in concept designing and clarity about understanding of aforementioned terminologies. The authors have attempted to review data of these closely knit terminologies and further organize its characteristic appearances, pathogenetic aspects and prognostic implications. The data published in English Language, from 1925 to 2015, were collected using keywords such as emperipolesis, entosis and cell cannibalism through scientific database systems such as MEDLINE, Science Direct, Cochrane Library and Google Scholar. Articles were selected which have focused to explain the phenomenon, presentation and pathogenesis of one or more of this phenomenon. A total of 48 articles were retrieved, thirty of which were selected. The various cell engulfment phenomena are very similar looking but operate through entirely different pathways.
Collapse
Affiliation(s)
- Nidhi Gupta
- Department of Oral Pathology and Microbiology, KM Shah Dental College, Sumandeep Vidyapeeth University, Vadodara, Gujarat, India
| | - Kiran Jadhav
- Department of Oral Pathology and Microbiology, KM Shah Dental College, Sumandeep Vidyapeeth University, Vadodara, Gujarat, India
| | - Vandana Shah
- Department of Oral Pathology and Microbiology, KM Shah Dental College, Sumandeep Vidyapeeth University, Vadodara, Gujarat, India
| |
Collapse
|
41
|
Ceglia I, Dueck AC, Masiello F, Martelli F, He W, Federici G, Petricoin EF, Zeuner A, Iancu-Rubin C, Weinberg R, Hoffman R, Mascarenhas J, Migliaccio AR. Preclinical rationale for TGF-β inhibition as a therapeutic target for the treatment of myelofibrosis. Exp Hematol 2016; 44:1138-1155.e4. [PMID: 27592389 PMCID: PMC5778911 DOI: 10.1016/j.exphem.2016.08.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 08/16/2016] [Accepted: 08/19/2016] [Indexed: 01/20/2023]
Abstract
To assess the role of abnormal transforming growth factor-beta (TGF-β) signaling in the pathogenesis of primary myelofibrosis (PMF), the effects of the TGF-β receptor-1 kinase inhibitor SB431542 on ex vivo expansion of hematopoietic cells in cultures from patients with JAK2V617+-polycythemia vera (PV) or PMF (JAK2V617F+, CALRpQ365f+, or unknown) and from normal sources (adult blood, AB, or cord blood, CB) were compared. In cultures of normal sources, SB431542 significantly increased by 2.5-fold the number of progenitor cells generated by days 1-2 (CD34+) and 6 (colony-forming cells) (CB) and that of precursor cells, mostly immature erythroblasts, by days 14-17 (AB and CB). In cultures of JAK2V617F+-PV, SB431542 increased by twofold the numbers of progenitor cells by day 10 and had no effect on that of precursors cells by days 12-17 (∼fourfold increase in all cases). In contrast, SB431542 had no effect on the number of either progenitor or precursor cells in cultures of JAK2V617F+ and CALR pQ365fs+ PMF. These ontogenetic- and disease-specific effects were associated with variegation in the ability of SB431542 to induce CD34+ cells from AB (increased), CB (decreased), or PV and PMF (unaffected) into cycle and erythroblasts in proliferation (increased for AB and PV and unaffected for CB and PMF). Differences in expansion of erythroblasts from AB, CB, and PV were associated with differences in activation of TGF-β signaling (SHCY317, SMAD2S245/250/255, and SMAD1S/S/SMAD5S/S/SMAD8S/S) detectable in these cells by phosphoproteomic profiling. In conclusion, treatment with TGF-β receptor-1 kinase inhibitors may reactivate normal hematopoiesis in PMF patients, providing a proliferative advantage over the unresponsive malignant clone.
Collapse
Affiliation(s)
- Ilaria Ceglia
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Francesca Masiello
- Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Fabrizio Martelli
- Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Wu He
- Flow Cytometry Shared Resource Facility, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulia Federici
- Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy; Regina Elena National Cancer Institute, Rome, Italy
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Ann Zeuner
- Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Camelia Iancu-Rubin
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Rita Migliaccio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Biomedical and Neuromotorial Sciences, Alma Mater University, Bologna, Italy.
| |
Collapse
|
42
|
Naspi A, Zingariello M, Sancillo L, Panasiti V, Polinari D, Martella M, Rosa Alba R, Londei P. IGFBP-3 inhibits Wnt signaling in metastatic melanoma cells. Mol Carcinog 2016; 56:681-693. [PMID: 27377812 PMCID: PMC5213668 DOI: 10.1002/mc.22525] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 06/17/2016] [Accepted: 07/01/2016] [Indexed: 12/27/2022]
Abstract
In previous works, we have shown that insulin‐like growth factor‐binding protein‐3 (IGFBP‐3), a tissue and circulating protein able to bind to IGFs, decreases drastically in the blood serum of patients with diffuse metastatic melanoma. In agreement with the clinical data, recombinant IGFBP‐3 was found to inhibit the motility and invasiveness of cultured metastatic melanoma cells and to prevent growth of grafted melanomas in mice. The present work was aimed at identifying the signal transduction pathways underlying the anti‐tumoral effects of IGFBP‐3. We show that the anti‐tumoral effect of IGFBP‐3 is due to inhibition of the Wnt pathway and depends upon the presence of CD44, a receptor protein known to modulate Wnt signaling. Once it has entered the cell, IGFBP‐3 binds the Wnt signalosome interacting specifically with its component GSK‐3β. As a consequence, the β‐catenin destruction complex dissociates from the LRP6 Wnt receptor and GSK‐3β is activated through dephosphorylation, becoming free to target cytoplasmic β‐catenin which is degraded by the proteasomal pathway. Altogether, the results suggest that IGFBP‐3 is a novel and effective inhibitor of Wnt signaling. As IGFBP‐3 is a physiological protein which has no detectable toxic effects either on cultured cells or live mice, it might qualify as an interesting new therapeutic agent in melanoma, and potentially many other cancers with a hyperactive Wnt signaling. © 2016 The Authors. Molecular Carcinogenesis Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Antimo Naspi
- Department of Cellular Biotechnologies and Haematology, Istituto Pasteur Fondazione Cenci-Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Maria Zingariello
- Laboratory of Microscopic and Ultrastructural Anatomy, School of Medicine, Campus Bio-Medico University of Rome, Rome, Italy
| | - Laura Sancillo
- Department of Medicine and Aging Sciences, Section of Human Morphology, University G. D'Annunzio, Chieti, Italy
| | - Vincenzo Panasiti
- Plastic and Reconstructive Surgery Unit, Campus Bio-Medico University of Rome, Rome, Italy
| | - Dorina Polinari
- Department of Cellular Biotechnologies and Haematology, Istituto Pasteur Fondazione Cenci-Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Marianna Martella
- Department of Cellular Biotechnologies and Haematology, Istituto Pasteur Fondazione Cenci-Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Rana Rosa Alba
- Department of Medicine and Aging Sciences, Section of Human Morphology, University G. D'Annunzio, Chieti, Italy
| | - Paola Londei
- Department of Cellular Biotechnologies and Haematology, Istituto Pasteur Fondazione Cenci-Bolognetti, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
43
|
Shimizu R, Yamamoto M. GATA-related hematologic disorders. Exp Hematol 2016; 44:696-705. [PMID: 27235756 DOI: 10.1016/j.exphem.2016.05.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/15/2016] [Accepted: 05/17/2016] [Indexed: 01/09/2023]
Abstract
The transcription factors GATA1 and GATA2 are fundamental regulators of hematopoiesis and have overlapping expression profiles. GATA2 is expressed in hematopoietic stem cells and early erythroid-megakaryocytic progenitors and activates a certain set of early-phase genes, including the GATA2 gene itself. GATA2 also initiates GATA1 gene expression. In contrast, GATA1 is expressed in relatively mature erythroid progenitors and facilitates the expression of genes associated with differentiation, including the GATA1 gene itself; however, GATA1 represses the expression of GATA2. Switching the GATA factors from GATA2 to GATA1 appears to be one of the key regulatory mechanisms underlying erythroid differentiation. Loss-of-function analyses using mice in vivo have indicated that GATA2 and GATA1 are functionally nonredundant and that neither can compensate for the absence of the other. However, transgenic expression of GATA2 under the transcriptional regulation of the Gata1 gene rescues lethal dyserythropoiesis in GATA1-deficient mice, illustrating that the dynamic expression profiles of these GATA factors are critically important for the maintenance of hematopoietic homeostasis. Analysis of naturally occurring leukemias in GATA1-knockdown mice revealed that leukemic stem cells undergo functional alterations in response to exposure to chemotherapeutic agents. This mechanism may also underlie the aggravating features of relapsing leukemias. Recent hematologic analyses have suggested that disturbances in the balance of the GATA factors are associated with specific types of hematopoietic disorders. Here, we describe GATA1- and GATA2-related hematologic diseases, focusing on the regulation of GATA factor gene expression.
Collapse
Affiliation(s)
- Ritsuko Shimizu
- Department of Molecular Hematology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
44
|
Spangrude GJ, Lewandowski D, Martelli F, Marra M, Zingariello M, Sancillo L, Rana RA, Migliaccio AR. P-Selectin Sustains Extramedullary Hematopoiesis in the Gata1 low Model of Myelofibrosis. Stem Cells 2015; 34:67-82. [PMID: 26439305 DOI: 10.1002/stem.2229] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 08/07/2015] [Accepted: 08/28/2015] [Indexed: 01/03/2023]
Abstract
Splenomegaly is a major manifestation of primary myelofibrosis (PMF) contributing to clinical symptoms and hematologic abnormalities. The spleen from PMF patients contains increased numbers of hematopoietic stem cells (HSC) and megakaryocytes (MK). These MK express high levels of P-selectin (P-sel) that, by triggering neutrophil emperipolesis, may cause TGF-β release and disease progression. This hypothesis was tested by deleting the P-sel gene in the myelofibrosis mouse model carrying the hypomorphic Gata1(low) mutation that induces megakaryocyte abnormalities that recapitulate those observed in PMF. P-sel(null) Gata1(low) mice survived splenectomy and lived 3 months longer than P-sel(WT) Gata1(low) littermates and expressed limited fibrosis and osteosclerosis in the marrow or splenomegaly. Furthermore, deletion of P-sel disrupted megakaryocyte/neutrophil interactions in spleen, reduced TGF-β content, and corrected the HSC distribution that in Gata1(low) mice, as in PMF patients, is abnormally expanded in spleen. Conversely, pharmacological inhibition of TGF-β reduced P-sel expression in MK and corrected HSC distribution. Spleens, but not marrow, of Gata1(low) mice contained numerous cKIT(pos) activated fibrocytes, probably of dendritic cell origin, whose membrane protrusions interacted with MK establishing niches hosting immature cKIT(pos) hematopoietic cells. These activated fibrocytes were not detected in spleens from P-sel(null) Gata1(low) or TGF-β-inhibited Gata1(low) littermates and were observed in spleen, but not in marrow, from PMF patients. Therefore, in Gata1(low) mice, and possibly in PMF, abnormal P-sel expression in MK may mediate the pathological cell interactions that increase TGF-β content in MK and favor establishment of a microenvironment that supports myelofibrosis-related HSC in spleen.
Collapse
Affiliation(s)
- Gerald J Spangrude
- Department of Medicine, Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, Utah, USA
| | | | - Fabrizio Martelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità
| | - Manuela Marra
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità
| | | | - Laura Sancillo
- Istituto Genetica Medica, Centro Nazionale Ricerche, and Medicine and Aging Sciences, Section of Human Momorphology, University G. D'Annunzio, Chieti, Italy
| | - Rosa Alba Rana
- Istituto Genetica Medica, Centro Nazionale Ricerche, and Medicine and Aging Sciences, Section of Human Momorphology, University G. D'Annunzio, Chieti, Italy
| | - Anna Rita Migliaccio
- Department of Biomedical Sciences, Alma Mater University, Bologna, Italy.,Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA
| |
Collapse
|
45
|
Recombinant interferon-α in myelofibrosis reduces bone marrow fibrosis, improves its morphology and is associated with clinical response. Mod Pathol 2015; 28:1315-23. [PMID: 26271725 DOI: 10.1038/modpathol.2015.93] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/19/2015] [Accepted: 06/21/2015] [Indexed: 12/14/2022]
Abstract
Recombinant interferon-α represents a well-established therapeutic option for the treatment of polycythemia vera and essential thrombocythemia. Recent studies also suggest a role for recombinant interferon-α in the treatment of 'early stage' primary myelofibrosis, but few studies have reported the bone marrow changes after clinically successful interferon therapy. The aim of the present study is to detail the histological responses to recombinant interferon-α in primary myelofibrosis and post-polycythemia vera/post-essential thrombocythemia myelofibrosis and to correlate these with clinical findings. We retrospectively studied 12 patients with primary myelofibrosis or post-polycythemia vera/post-essential thrombocythemia myelofibrosis, who had been treated with recombinant interferon-α. Six patients had received other prior cytoreductive therapies. Bone marrow biopsy was assessed for the following histological parameters: (i) cellularity; (ii) myeloid-to-erythroid ratio; (iii) megakaryocyte tight clusters; (iv) megakaryocyte and naked nuclei density; (v) megakaryocytic atypia; (vi) fibrosis; and (vii) the percentage of blasts. Clinical and laboratory data were included: (i) constitutional symptoms; (ii) splenomegaly, if present; and (iii) complete cell blood count. The clinical response to therapy was evaluated using the International Working Group for Myelofibrosis Research and Treatment/European LeukemiaNet response criteria. The Dynamic International Prognostic Scoring System (DIPSS) score was calculated before and after recombinant interferon-α administration. Successful interferon therapy for myelofibrosis was associated with a significant reduction of marrow fibrosis, cellularity, megakaryocyte density and naked nuclei density. The presence of JAK2(V617F) mutation correlated with improved DIPSS score. JAK2(V617F)-negative cases showed worsening of such score or evolution to acute myeloid leukemia. Cytogenetic analysis documented a normal karyotype in all cases. In conclusion, successful clinical response to interferon-α correlates well with an improvement of bone marrow morphology. The prognostic effect of such therapy may be influenced by the JAK2 mutational status. Additional studies are needed to confirm these preliminary data.
Collapse
|
46
|
Larocca LM, Heller PG, Podda G, Pujol-Moix N, Glembotsky AC, Pecci A, Alberelli MA, Balduini CL, Landolfi R, Cattaneo M, De Candia E. Megakaryocytic emperipolesis and platelet function abnormalities in five patients with gray platelet syndrome. Platelets 2015; 26:751-7. [PMID: 25806575 DOI: 10.3109/09537104.2014.994093] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The gray platelet syndrome (GPS) is a rare congenital platelet disorder characterized by mild to moderate bleeding diathesis, macrothrombocytopenia and lack of azurophilic α-granules in platelets. Some platelet and megakaryocyte (MK) abnormalities have been described, but confirmative studies of the defects in larger patient cohorts have not been undertaken. We studied platelet function and bone marrow (BM) features in five GPS patients with NBEAL2 autosomal recessive mutations from four unrelated families. In 3/3 patients, we observed a defect in platelet responses to protease-activated receptor (PAR)1-activating peptide as the most consistent finding, either isolated or combined to defective responses to other agonists. A reduction of PAR1 receptors with normal expression of major glycoproteins on the platelet surface was also found. Thrombin-induced fibrinogen binding to platelets was severely impaired in 2/2 patients. In 4/4 patients, the BM biopsy showed fibrosis (grade 2-3) and extensive emperipolesis, with many (36-65%) MKs containing 2-4 leukocytes engulfed within the cytoplasm. Reduced immunolabeling for platelet factor 4 together with normal immunolabeling for CD63 in MKs of two patients demonstrated that GPS MKs display an alpha granule-specific defect. Increased immunolabeling for P-selectin and decreased immunolabeling for PAR1, PAR4 and c-MPL were also observed in MKs of two patients. Marked emperipolesis, specific defect of MK alpha-granule content and defect of PAR1-mediated platelet responses are present in all GPS patients that we could study in detail. These results help to further characterize the disease.
Collapse
Affiliation(s)
- Luigi M Larocca
- a Department of Pathology , Policlinico A. Gemelli, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Paula G Heller
- b Department of Hematology Research , Instituto de Investigaciones Médicas Alfredo Lanari, University of Buenos Aires, CONICET , Buenos Aires , Argentina
| | - Gianmarco Podda
- c Medicina III, Azienda Ospedaliera San Paolo, Dipartimento di Scienze della Salute , Università degli Studi di Milano , Milano , Italy
| | - Nuria Pujol-Moix
- d Hemostasis and Thrombosis Unit, Department of Medicine , Institut de Recerca Sant Pau, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Ana C Glembotsky
- b Department of Hematology Research , Instituto de Investigaciones Médicas Alfredo Lanari, University of Buenos Aires, CONICET , Buenos Aires , Argentina
| | - Alessandro Pecci
- e Department of Internal Medicine , IRCCS Policlinico San Matteo Foundation, University of Pavia , Pava , Italy , and
| | - Maria Adele Alberelli
- f Department of Internal Medicine , Policlinico A. Gemelli, Università Cattolica del Sacro Cuore , Roma , Italy
| | - Carlo L Balduini
- e Department of Internal Medicine , IRCCS Policlinico San Matteo Foundation, University of Pavia , Pava , Italy , and
| | - Raffaele Landolfi
- f Department of Internal Medicine , Policlinico A. Gemelli, Università Cattolica del Sacro Cuore , Roma , Italy
| | - Marco Cattaneo
- c Medicina III, Azienda Ospedaliera San Paolo, Dipartimento di Scienze della Salute , Università degli Studi di Milano , Milano , Italy
| | - Erica De Candia
- f Department of Internal Medicine , Policlinico A. Gemelli, Università Cattolica del Sacro Cuore , Roma , Italy
| |
Collapse
|
47
|
Evaluation of plitidepsin in patients with primary myelofibrosis and post polycythemia vera/essential thrombocythemia myelofibrosis: results of preclinical studies and a phase II clinical trial. Blood Cancer J 2015; 5:e286. [PMID: 25768401 PMCID: PMC4382667 DOI: 10.1038/bcj.2015.5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 01/09/2015] [Accepted: 01/21/2015] [Indexed: 12/12/2022] Open
Abstract
Previous data established that plitidepsin, a cyclic depsipeptide, exerted activity in a mouse model of myelofibrosis (MF). New preclinical experiments reported herein found that low nanomolar plitidepsin concentrations potently inhibited the proliferation of JAK2V617F-mutated cell lines and reduced colony formation by CD34+ cells of individuals with MF, at least in part through modulation of p27 levels. Cells of MF patients had significantly reduced p27 content, that were modestly increased upon plitidepsin exposure. On these premise, an exploratory phase II trial evaluated plitidepsin 5 mg/m(2) 3-h intravenous infusion administered on days 1 and 15 every 4 weeks (q4wk). Response rate (RR) according to the International Working Group for Myelofibrosis Research and Treatment consensus criteria was 9.1% (95% CI, 0.2-41.3%) in 11 evaluable patients during the first trial stage. The single responder achieved a red cell transfusion independence and stable disease was reported in nine additional patients (81.8%). Eight patients underwent a short-lasting improvement of splenomegaly. In conclusion, plitidepsin 5 mg/m(2) 3-h infusion q4wk was well tolerated but had a modest activity in patients with primary, post-polycythaemia vera or post-essential thrombocythaemia MF. Therefore, this trial was prematurely terminated and we concluded that further clinical trials with plitidepsin as single agent in MF are not warranted.
Collapse
|
48
|
Ciaffoni F, Cassella E, Varricchio L, Massa M, Barosi G, Migliaccio AR. Activation of non-canonical TGF-β1 signaling indicates an autoimmune mechanism for bone marrow fibrosis in primary myelofibrosis. Blood Cells Mol Dis 2015; 54:234-41. [PMID: 25703685 DOI: 10.1016/j.bcmd.2014.12.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 12/31/2014] [Indexed: 11/25/2022]
Abstract
Primary myelofibrosis (PMF) is characterized by megakaryocyte hyperplasia, dysplasia and death with progressive reticulin/collagen fibrosis in marrow and hematopoiesis in extramedullary sites. The mechanism of fibrosis was investigated by comparing TGF-β1 signaling of marrow and spleen of patients with PMF and of non-diseased individuals. Expression of 39 (23 up-regulated and 16 down-regulated) and 38 (8 up-regulated and 30 down-regulated) TGF-β1 signaling genes was altered in the marrow and spleen of PMF patients, respectively. Abnormalities included genes of TGF-β1 signaling, cell cycling and abnormal in chronic myeloid leukemia (EVI1 and p21(CIP)) (both marrow and spleen) and Hedgehog (marrow only) and p53 (spleen only) signaling. Pathway analyses of these alterations predict an increased osteoblast differentiation, ineffective hematopoiesis and fibrosis driven by non-canonical TGF-β1 signaling in marrow and increased proliferation and defective DNA repair in spleen. Since activation of non-canonical TGF-β1 signaling is associated with fibrosis in autoimmune diseases, the hypothesis that fibrosis in PMF results from an autoimmune process triggered by dead megakaryocytes was tested by determining that PMF patients expressed plasma levels of mitochondrial DNA and anti-mitochondrial antibodies greater than normal controls. These data identify autoimmunity as a possible cause of marrow fibrosis in PMF.
Collapse
Affiliation(s)
- Fiorella Ciaffoni
- Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Cassella
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - Lilian Varricchio
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - Margherita Massa
- Biotechnology Research Area, Center for the Study of Myelofibrosis, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Giovanni Barosi
- Biotechnology Research Area, Center for the Study of Myelofibrosis, IRCCS Policlinico San Matteo Foundation, Pavia, Italy; Myeloproliferative Disease Research Consortium, New York, NY, USA
| | - Anna Rita Migliaccio
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, USA; Myeloproliferative Disease Research Consortium, New York, NY, USA; Department of Biomedical Sciences, Alma Mater University, Bologna, Italy.
| |
Collapse
|
49
|
Falchi M, Varricchio L, Martelli F, Masiello F, Federici G, Zingariello M, Girelli G, Whitsett C, Petricoin EF, Moestrup SK, Zeuner A, Migliaccio AR. Dexamethasone targeted directly to macrophages induces macrophage niches that promote erythroid expansion. Haematologica 2014; 100:178-87. [PMID: 25533803 DOI: 10.3324/haematol.2014.114405] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Cultures of human CD34(pos) cells stimulated with erythroid growth factors plus dexamethasone, a model for stress erythropoiesis, generate numerous erythroid cells plus a few macrophages (approx. 3%; 3:1 positive and negative for CD169). Interactions occurring between erythroblasts and macrophages in these cultures and the biological effects associated with these interactions were documented by live phase-contrast videomicroscopy. Macrophages expressed high motility interacting with hundreds/thousands of erythroblasts per hour. CD169(pos) macrophages established multiple rapid 'loose' interactions with proerythroblasts leading to formation of transient erythroblastic island-like structures. By contrast, CD169(neg) macrophages established 'tight' interactions with mature erythroblasts and phagocytosed these cells. 'Loose' interactions of CD169(pos) macrophages were associated with proerythroblast cytokinesis (the M phase of the cell cycle) suggesting that these interactions may promote proerythroblast duplication. This hypothesis was tested by experiments that showed that as few as 103 macrophages significantly increased levels of 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide incorporation frequency in S/G2/M and cytokinesis expressed by proerythroblasts over 24 h of culture. These effects were observed also when macrophages were co-cultured with dexamethasone directly conjugated to a macrophage-specific CD163 antibody. In conclusion, in addition to promoting proerythroblast proliferation directly, dexamethasone stimulates expansion of these cells indirectly by stimulating maturation and cytokinesis supporting activity of macrophages.
Collapse
Affiliation(s)
- Mario Falchi
- National AIDS Center, New York, NY, USA Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - Lilian Varricchio
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - Fabrizio Martelli
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, USA Hematology/Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Masiello
- Hematology/Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giulia Federici
- Regina Elena National Cancer Institute, Rome, Italy Hematology/Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Carolyn Whitsett
- Kings County Hospital and Downstate Medical Center, Brooklyn, NY, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Søren Kragh Moestrup
- Department of Biomedicine, University of Aarhus, Aarhus C, Denmark Institute of Molecular Medicine, University of Souther Denmark, Denmark
| | - Ann Zeuner
- Hematology/Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Anna Rita Migliaccio
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, USA Hematology/Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
50
|
Varricchio L, Mancini A, Migliaccio AR. Pathological interactions between hematopoietic stem cells and their niche revealed by mouse models of primary myelofibrosis. Expert Rev Hematol 2014; 2:315-334. [PMID: 20352017 DOI: 10.1586/ehm.09.17] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Primary myelofibrosis (PMF) belongs to the Philadelphia-negative myeloproliferative neoplasms and is a hematological disorder caused by abnormal function of the hematopoietic stem cells. The disease manifests itself with a plethora of alterations, including anemia, splenomegaly and extramedullary hematopoiesis. Its hallmarks are progressive marrow fibrosis and atypical megakaryocytic hyperplasia, two distinctive features used to clinically monitor disease progression. In an attempt to investigate the role of abnormal megakaryocytopoiesis in the pathogenesis of PMF, several transgenic mouse models have been generated. These models are based either on mutations that interfere with the extrinsic (thrombopoietin and its receptor, MPL) and intrinsic (the GATA1 transcription factor) control of normal megakaryocytopoiesis, or on known genetic lesions associated with the human disease. Here we provide an up-to-date review on the insights into the pathobiology of human PMF achieved by studying these animal models, with particular emphasis on results obtained with Gata1(low) mice.
Collapse
Affiliation(s)
- Lilian Varricchio
- Department of Medicine, Division of Hematology/Oncology, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1079, New York, NY 10029, USA Tel.: +1 212 241 6974
| | | | | |
Collapse
|