1
|
Hu X, Yu X, Zhang L, Zhang Q, Ji M, Qi K, Wang S, Li Z, Xu K, Fu C. The aberrantly activated AURKB supports and complements the function of AURKA in CALR mutated cells through regulating the cell growth and differentiation. Exp Cell Res 2025; 444:114377. [PMID: 39706286 DOI: 10.1016/j.yexcr.2024.114377] [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: 06/14/2024] [Revised: 11/12/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
Aurora kinase B (AURKB) was reported to assist Aurora kinase A (AURKA) to regulate cellular mitosis. AURKA has been found activated in myeloproliferative neoplasms (MPNs) patients with CALR gene mutation, however, it's unclear whether AURKB displays a compensatory function of AURKA in regulation of CALR mutant cell growth and differentiation. Here, we found that AURKB, similar with AURKA, was aberrantly activated in CALR mutant patients, and displayed a more tolerance to the aurora kinase inhibitor. Inhibition of AURKA decreased cell growth and colony formation, induced cell differentiation and apoptosis, while, this inhibitive degree was further enhanced when AURKB was blocked by incremental inhibitor. Transcriptomic analyses revealed a more significant gene enrichment in cells with knockdown of AURKB than that of AURKA, mainly reflecting in oxidative phosphorylation, mitosis, proliferation and apoptosis signaling pathway. Moreover, downregulation of AURKB enhanced cell growth arrest and apoptosis more obviously than that of AURKA, and additionally promoted cell differentiation and metabolism-oxygen consumption rate (OCR). Otherwise, overexpression of AURKA or AURKB facilitated the cell proliferation of CALR mutant cells, and made cells more sensitive to the aurora kinase inhibitor. These results suggest that activated AURKB not only supports the functions of AURKA in promoting the growth of CALR mutated cells, but also has impeded the differentiation of these cells.
Collapse
Affiliation(s)
- Xueting Hu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Xiangru Yu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Liwei Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Qigang Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Mengchu Ji
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Kunming Qi
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Shujin Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.
| | - Chunling Fu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
2
|
Yamamoto Y, Iba S, Inaguma Y, Okamoto A, Abe A. ELC52: a novel megakaryocytic leukemia cell line with a CALR type 1 mutation. Leukemia 2025; 39:234-237. [PMID: 39379530 DOI: 10.1038/s41375-024-02434-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024]
Affiliation(s)
- Yukiya Yamamoto
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Aichi, Japan.
- School of Medicine, Fujita Health University, Toyoake, Aichi, Japan.
| | - Sachiko Iba
- Department of Hematology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Yoko Inaguma
- Department of Clinical General Medicine, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Akinao Okamoto
- Department of Hematology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Akihiro Abe
- Department of Hematology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| |
Collapse
|
3
|
Vadeikienė R, Jakštys B, Laukaitienė D, Šatkauskas S, Juozaitytė E, Ugenskienė R. The Role of Mutated Calreticulin in the Pathogenesis of BCR-ABL1-Negative Myeloproliferative Neoplasms. Int J Mol Sci 2024; 25:9873. [PMID: 39337361 PMCID: PMC11432199 DOI: 10.3390/ijms25189873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs) are characterized by increased proliferation of myeloid lineages in the bone marrow. Calreticulin (CALR) 52 bp deletion and CALR 5 bp insertion have been identified in essential thrombocythemia (ET) and primary myelofibrosis (PMF). There is not much data on the crosstalk between mutated CALR and MPN-related signaling pathways, such as JAK/STAT, PI3K/Akt/mTOR, and Hedgehog. Calreticulin, a multifunctional protein, takes part in many cellular processes. Nevertheless, there is little data on how mutated CALR affects the oxidative stress response and oxidative stress-induced DNA damage, apoptosis, and cell cycle progression. We aimed to investigate the role of the CALR 52 bp deletion and 5 bp insertion in the pathogenesis of MPN, including signaling pathway activation and functional analysis in CALR-mutated cells. Our data indicate that the JAK/STAT and PI3K/Akt/mTOR pathways are activated in CALR-mutated cells, and this activation does not necessarily depend on the CALR and MPL interaction. Moreover, it was found that CALR mutations impair calreticulin function, leading to reduced responses to oxidative stress and DNA damage. It was revealed that the accumulation of G2/M-CALR-mutated cells indicates that oxidative stress-induced DNA damage is difficult to repair. Taken together, this study contributes to a deeper understanding of the specific molecular mechanisms underlying CALR-mutated MPNs.
Collapse
Affiliation(s)
- Roberta Vadeikienė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Baltramiejus Jakštys
- Research on Delivery of Medicine and Genes Cluster, Faculty of Natural Sciences, Vytautas Magnus University, LT-44001 Kaunas, Lithuania
| | - Danguolė Laukaitienė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Saulius Šatkauskas
- Research on Delivery of Medicine and Genes Cluster, Faculty of Natural Sciences, Vytautas Magnus University, LT-44001 Kaunas, Lithuania
| | - Elona Juozaitytė
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Rasa Ugenskienė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| |
Collapse
|
4
|
Ushijima Y, Ishikawa Y, Nishiyama T, Kawashima N, Kanamori T, Sanada M, Kiyoi H. Clonal evolution process from essential thrombocythemia to acute myeloid leukemia in the original patient from whom the CALR-mutated Marimo cell line was established. NAGOYA JOURNAL OF MEDICAL SCIENCE 2024; 86:326-332. [PMID: 38962422 PMCID: PMC11219235 DOI: 10.18999/nagjms.86.2.326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/25/2023] [Indexed: 07/05/2024]
Abstract
We previously reported the Marimo cell line, which was established from the bone marrow cells of a patient with essential thrombocythemia (ET) at the last stage after transformation to acute myeloid leukemia (AML). This cell line is widely used for the biological analysis of ET because it harbors CALR mutation. However, genetic processes during disease progression in the original patient were not analyzed. We sequentially analyzed the genetic status in the original patient samples during disease progression. The ET clone had already acquired CALR and MPL mutations, and TP53 and NRAS mutations affected the disease progression from ET to AML in this patient. Particularly, the variant allele frequency of the NRAS mutation increased along with the disease progression after transformation, and the NRAS-mutated clone selectively proliferated in vitro, resulting in the establishment of the Marimo cell line. Although CALR and MPL mutations co-existed, MPL was not expressed in Marimo cells or any clinical samples. Furthermore, mitogen-activated protein kinase (MAPK) but not the JAK2-STAT pathway was activated. These results collectively indicate that MAPK activation is mainly associated with the proliferation ability of Marimo cells.
Collapse
Affiliation(s)
- Yoko Ushijima
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichi Ishikawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takahiro Nishiyama
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Hematology, Ichinomiya Municipal Hospital, Ichinomiya, Japan
| | - Naomi Kawashima
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Kanamori
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Masashi Sanada
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
5
|
Kramer F, Mullally A. Antibody targeting of mutant calreticulin in myeloproliferative neoplasms. J Cell Mol Med 2024; 28:e17896. [PMID: 37551061 PMCID: PMC10902560 DOI: 10.1111/jcmm.17896] [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: 05/27/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/09/2023] Open
Abstract
Mutations in calreticulin are one of the key disease-initiating mutations in myeloproliferative neoplasms (MPN). In MPN, mutant calreticulin translates with a novel C-terminus that leads to aberrant binding to the extracellular domain of the thrombopoietin receptor, MPL. This cell surface neoantigen has become an attractive target for immunological intervention. Here, we summarize recent advances in the development of mutant calreticulin targeting antibodies as a novel therapeutic approach in MPN.
Collapse
Affiliation(s)
- Frederike Kramer
- Division of Hematology, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Ann Mullally
- Division of Hematology, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Broad InstituteCambridgeMassachusettsUSA
| |
Collapse
|
6
|
Bigot T, Gabinaud E, Hannouche L, Sbarra V, Andersen E, Bastelica D, Falaise C, Bernot D, Ibrahim-Kosta M, Morange PE, Loosveld M, Saultier P, Payet-Bornet D, Alessi MC, Potier D, Poggi M. Single-cell analysis of megakaryopoiesis in peripheral CD34 + cells: insights into ETV6-related thrombocytopenia. J Thromb Haemost 2023; 21:2528-2544. [PMID: 37085035 DOI: 10.1016/j.jtha.2023.04.007] [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: 01/30/2023] [Revised: 03/21/2023] [Accepted: 04/04/2023] [Indexed: 04/23/2023]
Abstract
BACKGROUND Germline mutations in the ETV6 transcription factor gene are responsible for familial thrombocytopenia and leukemia predisposition syndrome. Although previous studies have shown that ETV6 plays an important role in megakaryocyte (MK) maturation and platelet formation, the mechanisms by which ETV6 dysfunction promotes thrombocytopenia remain unclear. OBJECTIVES To decipher the transcriptional mechanisms and gene regulatory network linking ETV6 germline mutations and thrombocytopenia. METHODS Presuming that ETV6 mutations result in selective effects at a particular cell stage, we applied single-cell RNA sequencing to understand gene expression changes during megakaryopoiesis in peripheral CD34+ cells from healthy controls and patients with ETV6-related thrombocytopenia. RESULTS Analysis of gene expression and regulon activity revealed distinct clusters partitioned into 7 major cell stages: hematopoietic stem/progenitor cells, common-myeloid progenitors (CMPs), MK-primed CMPs, granulocyte-monocyte progenitors, MK-erythroid progenitors (MEPs), progenitor MKs/mature MKs, and platelet-like particles. We observed a differentiation trajectory in which MEPs developed directly from hematopoietic stem/progenitor cells and bypassed the CMP stage. ETV6 deficiency led to the development of aberrant cells as early as the MEP stage, which intensified at the progenitor MK/mature MK stage, with a highly deregulated core "ribosome biogenesis" pathway. Indeed, increased translation levels have been documented in patient CD34+-derived MKs with overexpression of ribosomal protein S6 and phosphorylated ribosomal protein S6 in both CD34+-derived MKs and platelets. Treatment of patient MKs with the ribosomal biogenesis inhibitor CX-5461 resulted in an increase in platelet-like particles. CONCLUSION These findings provide novel insight into both megakaryopoiesis and the link among ETV6, translation, and platelet production.
Collapse
Affiliation(s)
- Timothée Bigot
- Aix-Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
| | - Elisa Gabinaud
- Aix-Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
| | | | | | - Elisa Andersen
- Aix-Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
| | | | | | - Denis Bernot
- Aix-Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
| | | | | | - Marie Loosveld
- Aix-Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Paul Saultier
- Aix-Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
| | | | - Marie-Christine Alessi
- Aix-Marseille Univ, INSERM, INRAe, C2VN, Marseille, France; AP-HM, CHU Timone, CRPP, Marseille, France
| | | | - Marjorie Poggi
- Aix-Marseille Univ, INSERM, INRAe, C2VN, Marseille, France.
| |
Collapse
|
7
|
Xue C, Yao Q, Gu X, Shi Q, Yuan X, Chu Q, Bao Z, Lu J, Li L. Evolving cognition of the JAK-STAT signaling pathway: autoimmune disorders and cancer. Signal Transduct Target Ther 2023; 8:204. [PMID: 37208335 DOI: 10.1038/s41392-023-01468-7] [Citation(s) in RCA: 202] [Impact Index Per Article: 101.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/22/2023] [Indexed: 05/21/2023] Open
Abstract
The Janus kinase (JAK) signal transducer and activator of transcription (JAK-STAT) pathway is an evolutionarily conserved mechanism of transmembrane signal transduction that enables cells to communicate with the exterior environment. Various cytokines, interferons, growth factors, and other specific molecules activate JAK-STAT signaling to drive a series of physiological and pathological processes, including proliferation, metabolism, immune response, inflammation, and malignancy. Dysregulated JAK-STAT signaling and related genetic mutations are strongly associated with immune activation and cancer progression. Insights into the structures and functions of the JAK-STAT pathway have led to the development and approval of diverse drugs for the clinical treatment of diseases. Currently, drugs have been developed to mainly target the JAK-STAT pathway and are commonly divided into three subtypes: cytokine or receptor antibodies, JAK inhibitors, and STAT inhibitors. And novel agents also continue to be developed and tested in preclinical and clinical studies. The effectiveness and safety of each kind of drug also warrant further scientific trials before put into being clinical applications. Here, we review the current understanding of the fundamental composition and function of the JAK-STAT signaling pathway. We also discuss advancements in the understanding of JAK-STAT-related pathogenic mechanisms; targeted JAK-STAT therapies for various diseases, especially immune disorders, and cancers; newly developed JAK inhibitors; and current challenges and directions in the field.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qinfan Yao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinyu Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhengyi Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
8
|
Guijarro-Hernández A, Eder-Azanza L, Hurtado C, Navarro-Herrera D, Ezcurra B, Novo FJ, Cabello J, Vizmanos JL. Transcriptomic Analysis Reveals JAK2/MPL-Independent Effects of Calreticulin Mutations in a C. elegans Model. Cells 2023; 12:186. [PMID: 36611979 PMCID: PMC9818371 DOI: 10.3390/cells12010186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/23/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
There is growing evidence that Ph-negative myeloproliferative neoplasms (MPNs) are disorders in which multiple molecular mechanisms are significantly disturbed. Since their discovery, CALR driver mutations have been demonstrated to trigger pathogenic mechanisms apart from the well-documented activation of JAK2/MPL-related pathways, but the lack of experimental models harboring CALR mutations in a JAK2/MPL knockout background has hindered the research on these non-canonical mechanisms. In this study, CRISPR/Cas9 was performed to introduce homozygous patient-like calreticulin mutations in a C. elegans model that naturally lacks JAK2 and MPL orthologs. Whole-genome transcriptomic analysis of these worms was conducted, and some of the genes identified to be associated with processes involved in the pathogenesis of MPNs were further validated by qPCR. Some of the transcriptomic alterations corresponded to typically altered genes and processes in cancer and Ph-negative MPN patients that are known to be triggered by mutant calreticulin without the intervention of JAK2/MPL. However, interestingly, we have also found altered other processes described in these diseases that had not been directly attributed to calreticulin mutations without the intervention of JAK2 or MPL. Thus, these results point to a new experimental model for the study of the JAK2/MPL-independent mechanisms of mutant calreticulin that induce these biological alterations, which could be useful to study unknown non-canonical effects of the mutant protein. The comparison with a calreticulin null strain revealed that the alteration of all of these processes seems to be a consequence of a loss of function of mutant calreticulin in the worm, except for the dysregulation of Hedgehog signaling and flh-3. Further analysis of this model could help to delineate these mechanisms, and the verification of these results in mammalian models may unravel new potential therapeutic targets in MPNs. As far as we know, this is the first time that a C. elegans strain with patient-like mutations is proposed as a potential model for leukemia research.
Collapse
Affiliation(s)
- Ana Guijarro-Hernández
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
| | - Laura Eder-Azanza
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
| | - Cristina Hurtado
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
| | - David Navarro-Herrera
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
| | - Begoña Ezcurra
- Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| | - Francisco Javier Novo
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Juan Cabello
- Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| | - José Luis Vizmanos
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
9
|
Zhan H, Kaushansky K. Megakaryocytes as the Regulator of the Hematopoietic Vascular Niche. Front Oncol 2022; 12:912060. [PMID: 35814384 PMCID: PMC9258777 DOI: 10.3389/fonc.2022.912060] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Megakaryocytes (MKs) are important components of the hematopoietic niche. Compared to the non-hematopoietic niche cells, MKs serving as part of the hematopoietic niche provides a mechanism for feedback regulation of hematopoietic stem cells (HSCs), in which HSC progeny (MKs) can modulate HSC adaptation to hematopoietic demands during both steady-state and stress hematopoiesis. MKs are often located adjacent to marrow sinusoids. Considering that most HSCs reside close to a marrow vascular sinusoid, as do MKs, the interactions between MKs and vascular endothelial cells are positioned to play important roles in modulating HSC function, and by extrapolation, might be dysregulated in various disease states. In this review, we discuss the interactions between MKs and the vascular niche in both normal and neoplastic hematopoiesis.
Collapse
Affiliation(s)
- Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, United States
- Medical Service, Northport Veterans Affairs (VA) Medical Center, Northport, NY, United States
- *Correspondence: Huichun Zhan,
| | - Kenneth Kaushansky
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, United States
| |
Collapse
|
10
|
Lee S, Wong H, Castiglione M, Murphy M, Kaushansky K, Zhan H. JAK2V617F Mutant Megakaryocytes Contribute to Hematopoietic Aging in a Murine Model of Myeloproliferative Neoplasm. Stem Cells 2022; 40:359-370. [PMID: 35260895 PMCID: PMC9199841 DOI: 10.1093/stmcls/sxac005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022]
Abstract
Megakaryocytes (MKs) is an important component of the hematopoietic niche. Abnormal MK hyperplasia is a hallmark feature of myeloproliferative neoplasms (MPNs). The JAK2V617F mutation is present in hematopoietic cells in a majority of patients with MPNs. Using a murine model of MPN in which the human JAK2V617F gene is expressed in the MK lineage, we show that the JAK2V617F-bearing MKs promote hematopoietic stem cell (HSC) aging, manifesting as myeloid-skewed hematopoiesis with an expansion of CD41+ HSCs, a reduced engraftment and self-renewal capacity, and a reduced differentiation capacity. HSCs from 2-year-old mice with JAK2V617F-bearing MKs were more proliferative and less quiescent than HSCs from age-matched control mice. Examination of the marrow hematopoietic niche reveals that the JAK2V617F-bearing MKs not only have decreased direct interactions with hematopoietic stem/progenitor cells during aging but also suppress the vascular niche function during aging. Unbiased RNA expression profiling reveals that HSC aging has a profound effect on MK transcriptomic profiles, while targeted cytokine array shows that the JAK2V617F-bearing MKs can alter the hematopoietic niche through increased levels of pro-inflammatory and anti-angiogenic factors. Therefore, as a hematopoietic niche cell, MKs represent an important connection between the extrinsic and intrinsic mechanisms for HSC aging.
Collapse
Affiliation(s)
- Sandy Lee
- Graduate Program in Molecular & Cellular Pharmacology, Stony Brook University, Stony Brook, NY, USA
| | - Helen Wong
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY, USA
| | | | | | - Kenneth Kaushansky
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, USA
| | - Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, USA
- Medical Service, Northport VA Medical Center, Northport, NY, USA
| |
Collapse
|
11
|
Inhibition of LDHA to Induce EEF2 Release Enhances Thrombocytopoiesis. Blood 2022; 139:2958-2971. [PMID: 35176139 DOI: 10.1182/blood.2022015620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/14/2022] [Indexed: 11/20/2022] Open
Abstract
Translation is essential for megakaryocyte (MK) maturation and platelet production. However, how the translational pathways are regulated in this process remains unknown. In this study, we found that megakaryocyte/platelet-specific lactate dehydrogenase A (LdhA)-knockout mice showed an increased number of platelets with remarkably accelerated MK maturation and proplatelet formation. Interestingly, the role of LDHA in MK maturation and platelet formation did not depend on lactate content, which was the major product of LDHA. Mechanism studies revealed that LDHA interacted with eukaryotic elongation factor 2 (eEF2) in the cytoplasm, controlling the participation of eEF2 in translation at the ribosome. Furthermore, the interaction of LDHA and eEF2 was dependent on NADH, a coenzyme of LDHA. NADH-competitive inhibitors of LDHA could release eEF2 from the LDHA pool, up-regulate translation and enhance MK maturation in vitro. Among LDHA inhibitors, stiripentol significantly promoted the production of platelets in vivo under physiological state and in the immune thrombocytopenia model. Moreover, stiripentol could promote platelet production from human cord blood mononuclear cells (CBMCs)-derived megakaryocytes, and also have a superposed effect with romiplostim. In short, this study reveals a novel non-classical function of LDHA in translation and may serve as a potential target for thrombocytopenia therapy.
Collapse
|
12
|
Moroi AJ, Newman PJ. Conditional CRISPR-mediated deletion of Lyn kinase enhances differentiation and function of iPSC-derived megakaryocytes. J Thromb Haemost 2022; 20:182-195. [PMID: 34624170 PMCID: PMC8712352 DOI: 10.1111/jth.15546] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/21/2021] [Accepted: 10/05/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Thrombocytopenia leading to life-threatening excessive bleeding can be treated by platelet transfusion. Currently, such treatments are totally dependent on donor-derived platelets. To support future applications in the use of in vitro-derived platelets, we sought to identify genes whose manipulation might improve the efficiency of megakaryocyte production and resulting hemostatic effectiveness. Disruption of Lyn kinase has previously been shown to improve cell survival, megakaryocyte ploidy and TPO-mediated activation in mice, but its role in human megakaryocytes and platelets has not been examined. METHODS To analyze the role of Lyn at defined differentiation stages during human megakaryocyte differentiation, conditional Lyn-deficient cells were generated using CRISPR/Cas9 technology in iPS cells. The efficiency of Lyn-deficient megakaryocytes to differentiate and become activated in response to a range of platelet agonists was analyzed in iPSC-derived megakaryocytes. RESULTS Temporally controlled deletion of Lyn improved the in vitro differentiation of hematopoietic progenitor cells into mature megakaryocytes, as measured by the rate and extent of appearance of CD41+ CD42+ cells. Lyn-deficient megakaryocytes also demonstrated improved hemostatic effectiveness, as reported by their ability to mediate clot formation in rotational thromboelastometry. Finally, Lyn-deficient megakaryocytes produced increased numbers of platelet-like particles (PLP) in vitro. CONCLUSIONS Conditional deletion of Lyn kinase increases the hemostatic effectiveness of megakaryocytes and their progeny as well as improving their yield. Adoption of this system during generation of in vitro-derived platelets may contribute to both their efficiency of production and their ability to support hemostasis.
Collapse
Affiliation(s)
- Alyssa J. Moroi
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI
| | - Peter J. Newman
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI
- Department of Cell biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
13
|
Olschok K, Han L, de Toledo MAS, Böhnke J, Graßhoff M, Costa IG, Theocharides A, Maurer A, Schüler HM, Buhl EM, Pannen K, Baumeister J, Kalmer M, Gupta S, Boor P, Gezer D, Brümmendorf TH, Zenke M, Chatain N, Koschmieder S. CALR frameshift mutations in MPN patient-derived iPSCs accelerate maturation of megakaryocytes. Stem Cell Reports 2021; 16:2768-2783. [PMID: 34678208 PMCID: PMC8581168 DOI: 10.1016/j.stemcr.2021.09.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
Calreticulin (CALR) mutations are driver mutations in myeloproliferative neoplasms (MPNs), leading to activation of the thrombopoietin receptor and causing abnormal megakaryopoiesis. Here, we generated patient-derived CALRins5- or CALRdel52-positive induced pluripotent stem cells (iPSCs) to establish an MPN disease model for molecular and mechanistic studies. We demonstrated myeloperoxidase deficiency in granulocytic cells derived from homozygous CALR mutant iPSCs, rescued by repairing the mutation using CRISPR/Cas9. iPSC-derived megakaryocytes showed characteristics of primary megakaryocytes such as formation of demarcation membrane system and cytoplasmic pro-platelet protrusions. Importantly, CALR mutations led to enhanced megakaryopoiesis and accelerated megakaryocytic development in a thrombopoietin-independent manner. Mechanistically, our study identified differentially regulated pathways in mutated versus unmutated megakaryocytes, such as hypoxia signaling, which represents a potential target for therapeutic intervention. Altogether, we demonstrate key aspects of mutated CALR-driven pathogenesis dependent on its zygosity, and found novel therapeutic targets, making our model a valuable tool for clinical drug screening in MPNs. CALR-mutated iPSCs allow efficient modeling of human MPN disease CRISPR-mediated repair of CALR mutations rescues normal iPSC function Megakaryopoiesis in CALR-mutated iPSCs is hyperplastic and accelerated Transcriptome screen of mutated megakaryocytes identifies novel therapeutic options
Collapse
Affiliation(s)
- Kathrin Olschok
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Lijuan Han
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany; Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Marcelo A S de Toledo
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Janik Böhnke
- Institute for Biomedical Engineering, Department of Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany; Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Martin Graßhoff
- Institute for Computational Genomics Joint Research Center for Computational Biomedicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Ivan G Costa
- Institute for Computational Genomics Joint Research Center for Computational Biomedicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Alexandre Theocharides
- Division of Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Angela Maurer
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Herdit M Schüler
- Institute for Human Genetics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Eva Miriam Buhl
- Institute for Pathology, Electron Microscopy Facility, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Kristina Pannen
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Julian Baumeister
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Milena Kalmer
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Siddharth Gupta
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Peter Boor
- Institute for Pathology, Electron Microscopy Facility, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Deniz Gezer
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Martin Zenke
- Institute for Biomedical Engineering, Department of Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany; Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Nicolas Chatain
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany.
| |
Collapse
|
14
|
Wang C, Hu X, Wan Y, Wang S, Qi K, Li Y, Qiao J, Zeng L, Li Z, Fu C, Xu K. The Synergistic Inhibitory Effect of Combining MK-2206 and AZD 6244 in MARIMO Cells Harboring a Calreticulin Gene Mutation. Chemotherapy 2021; 66:169-178. [PMID: 34666331 DOI: 10.1159/000518921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/05/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Somatic mutations in the calreticulin (CALR) gene occur in most myeloproliferative neoplasm (MPN) patients who lack Janus kinase 2 or thrombopoietin receptor (MPL) mutations, but the molecular pathogenesis of MPN with mutated CALR is unclear, which limited the further treatment for CALR gene mutant patients. OBJECTIVES Previous studies showed that CALR mutations not only activated serine/threonine protein kinase (AKT) in primary mouse bone marrow cells but also mitogen-activated protein kinases (MAPKs) in MARIMO cells harboring a heterozygous 61-bp deletion in CALR exon 9, which were responsible for mutant CALR cell survival, respectively. Hence, we aimed to initially explore the mechanism of AKT activation and observe the synergistic inhibitory effect of combining AKT (MK-2206) and MAPK kinase (AZD 6244) inhibitors in MARIMO cells. METHODS We detected the expression of phosphorylated AKT in MARIMO cells treated with inhibitors for 24 or 48 h by western blotting and analyzed cell proliferation, cell cycle, and apoptosis by flow cytometry. We further examined the synergistic inhibitory effect of combining MK-2206 and AZD 6244 in MARIMO cells using the median effect principle of Chou and Talalay. RESULTS We found that the AKT was activated in MARIMO cells, and blocking its activity significantly inhibited MARIMO cell growth with downregulation of cyclin D and E, and accelerated cell apoptosis by decreasing Bcl-2 but increasing Bax and cleaved caspase-3 levels in a dose-dependent manner. Further analysis showed that AKT activation was dependent on mammalian target of rapamycin but not on the JAK signaling pathway in MARIMO cells, displaying that inhibition of JAK activity by ruxolitinib (RUX) did not decrease the AKT phosphorylation. Furthermore, the combination of MK-2206 and AZD 6244 produced a significantly synergistic inhibitory effect on MARIMO cells. CONCLUSIONS AKT activation is a feature of MARIMO cells and co-targeting of AKT and MAPKs signaling pathways synergistically inhibits MARIMO cell growth.
Collapse
Affiliation(s)
- Chunqing Wang
- Nanjing Medical University, Nanjing, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xueting Hu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Yan Wan
- The affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital Huai'an, Huai'an, China
| | - Shujin Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Kunming Qi
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Yanjie Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Jianlin Qiao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Lingyu Zeng
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chunling Fu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Kailin Xu
- Nanjing Medical University, Nanjing, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
15
|
Giai V, Secreto C, Freilone R, Pregno P. Philadelphia-Negative MPN: A Molecular Journey, from Hematopoietic Stem Cell to Clinical Features. MEDICINA-LITHUANIA 2021; 57:medicina57101043. [PMID: 34684081 PMCID: PMC8537741 DOI: 10.3390/medicina57101043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 11/17/2022]
Abstract
Philadelphia negative Myeloproliferative Neoplasms (MPN) are a heterogeneous group of hematopoietic stem cell diseases. MPNs show different risk grades of thrombotic complications and acute myeloid leukemia evolution. In the last couple of decades, from JAK2 mutation detection in 2005 to the newer molecular trademarks studied through next generation sequencing, we are learning to approach MPNs from a deeper perspective. Here, we intend to elucidate the important factors affecting MPN clonal advantage and the reasons why some patients progress to more aggressive disease. Understanding these mechanisms is the key to developing new treatment approaches and targeted therapies for MPN patients.
Collapse
|
16
|
Hui W, Zhang W, Liu C, Wan S, Sun W, Su L. Alterations of Signaling Pathways in Essential Thrombocythemia with Calreticulin Mutation. Cancer Manag Res 2021; 13:6231-6238. [PMID: 34393515 PMCID: PMC8357313 DOI: 10.2147/cmar.s316919] [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: 04/21/2021] [Accepted: 07/26/2021] [Indexed: 12/25/2022] Open
Abstract
Purpose Though mutations of the calreticulin (CALR) gene have been identified in essential thrombocythemia patients, the detailed mechanisms for CALR mutations have not been completely clarified. Our study is aimed at characterizing alteration of protein expression in ET patients with mutated CALRdel52 and further recognizing possible involvement of signaling pathways associated with CALR mutations. Patients and Methods Protein pathway array was performed to analyze the expression levels of proteins involved in various signaling pathways in peripheral blood neutrophils from 18 ET patients with mutated CALRdel52, 20 ET patients with JAK2V617F mutation and 20 controls. Results We found 20 proteins differentially expressed in ET patients with mutated CALRdel52 compared with healthy controls. These proteins were associated with molecular mechanisms of cancer in ingenuity pathways analysis (IPA) network. We identified top ten canonical pathways which including apoptotic pathways and cellular cytokine pathways might participate in pathogenesis of ET with mutated CALRdel52. Additionally, there were 8 proteins found to be dysregulated differently between ET patients with mutated CALRdel52 and those with JAK2V617F mutation. These proteins might be related to the unique signaling pathways activated by CALRdel52 mutation which were different to JAK/STATs pathway by JAK2V617F mutation. Conclusion Our study demonstrated that numerous alterations of signaling proteins and pathways in ET patients with mutated CALRdel52. These findings could help to gain insights into the pathological mechanisms of ET.
Collapse
Affiliation(s)
- Wuhan Hui
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wei Zhang
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Congyan Liu
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Suigui Wan
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wanling Sun
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Li Su
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
17
|
Mapping human calreticulin regions important for structural stability. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140710. [PMID: 34358706 DOI: 10.1016/j.bbapap.2021.140710] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/24/2021] [Accepted: 08/02/2021] [Indexed: 12/28/2022]
Abstract
Calreticulin (CALR) is a highly conserved multifunctional chaperone protein primarily present in the endoplasmic reticulum, where it regulates Ca2+ homeostasis. Recently, CALR has gained special interest for its diverse functions outside the endoplasmic reticulum, including the cell surface and extracellular space. Although high-resolution structures of CALR exist, it has not yet been established how different regions and individual amino acid residues contribute to structural stability of the protein. In the present study, we have identified key residues determining the structural stability of CALR. We used a Saccharomyces cerevisiae expression system to express and purify 50 human CALR mutants, which were analysed for several parameters including secretion titer, melting temperature (Tm), stability and oligomeric state. Our results revealed the importance of a previously identified small patch of conserved surface residues, amino acids 166-187 ("cluster 2") for structural stability of the human CALR protein. Two residues, Tyr172 and Asp187, were critical for maintaining the native structure of the protein. Mutant D187A revealed a severe drop in secretion titer, it was thermally unstable, prone to degradation, and oligomer formation. Tyr172 was critical for thermal stability of CALR and interacted with the third free Cys163 residue. This illustrates an unusual thermal stability of CALR dominated by Asp187, Tyr172 and Cys163, which may interact as part of a conserved structural unit. Besides structural clusters, we found a correlation of some measured parameter values in groups of CALR mutants that cause myeloproliferative neoplasms (MPN) and in mutants that may be associated with sudden unexpected death (SUD).
Collapse
|
18
|
Jia R, Kutzner L, Koren A, Runggatscher K, Májek P, Müller AC, Schuster M, Bock C, Loizou JI, Kubicek S, Kralovics R. High-throughput drug screening identifies the ATR-CHK1 pathway as a therapeutic vulnerability of CALR mutated hematopoietic cells. Blood Cancer J 2021; 11:137. [PMID: 34333533 PMCID: PMC8325683 DOI: 10.1038/s41408-021-00531-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Mutations of calreticulin (CALR) are the second most prevalent driver mutations in essential thrombocythemia and primary myelofibrosis. To identify potential targeted therapies for CALR mutated myeloproliferative neoplasms, we searched for small molecules that selectively inhibit the growth of CALR mutated cells using high-throughput drug screening. We investigated 89 172 compounds using isogenic cell lines carrying CALR mutations and identified synthetic lethality with compounds targeting the ATR-CHK1 pathway. The selective inhibitory effect of these compounds was validated in a co-culture assay of CALR mutated and wild-type cells. Of the tested compounds, CHK1 inhibitors potently depleted CALR mutated cells, allowing wild-type cell dominance in the co-culture over time. Neither CALR deficient cells nor JAK2V617F mutated cells showed hypersensitivity to ATR-CHK1 inhibition, thus suggesting specificity for the oncogenic activation by the mutant CALR. CHK1 inhibitors induced replication stress in CALR mutated cells revealed by elevated pan-nuclear staining for γH2AX and hyperphosphorylation of RPA2. This was accompanied by S-phase cell cycle arrest due to incomplete DNA replication. Transcriptomic and phosphoproteomic analyses revealed a replication stress signature caused by oncogenic CALR, suggesting an intrinsic vulnerability to CHK1 perturbation. This study reveals the ATR-CHK1 pathway as a potential therapeutic target in CALR mutated hematopoietic cells.
Collapse
Affiliation(s)
- Ruochen Jia
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Leon Kutzner
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Anna Koren
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Kathrin Runggatscher
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Peter Májek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - André C Müller
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Michael Schuster
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Joanna I Loizou
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Robert Kralovics
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
19
|
Beuret L, Fortier-Beaulieu SP, Rondeau V, Roy S, Houde N, Balabanian K, Espéli M, Charron J. Mek1 and Mek2 Functional Redundancy in Erythropoiesis. Front Cell Dev Biol 2021; 9:639022. [PMID: 34386488 PMCID: PMC8353236 DOI: 10.3389/fcell.2021.639022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/21/2021] [Indexed: 12/23/2022] Open
Abstract
Several studies have established the crucial role of the extracellular signal–regulated kinase (ERK)/mitogen-activated protein kinase pathway in hematopoietic cell proliferation and differentiation. MEK1 and MEK2 phosphorylate and activate ERK1 and ERK2. However, whether MEK1 and MEK2 differentially regulate these processes is unknown. To define the function of Mek genes in the activation of the ERK pathway during hematopoiesis, we generated a mutant mouse line carrying a hematopoietic-specific deletion of the Mek1 gene function in a Mek2 null background. Inactivation of both Mek1 and Mek2 genes resulted in death shortly after birth with a severe anemia revealing the essential role of the ERK pathway in erythropoiesis. Mek1 and Mek2 functional ablation also affected lymphopoiesis and myelopoiesis. In contrast, mice that retained one functional Mek1 (1Mek1) or Mek2 (1Mek2) allele in hematopoietic cells were viable and fertile. 1Mek1 and 1Mek2 mutants showed mild signs of anemia and splenomegaly, but the half-life of their red blood cells and the response to erythropoietic stress were not altered, suggesting a certain level of Mek redundancy for sustaining functional erythropoiesis. However, subtle differences in multipotent progenitor distribution in the bone marrow were observed in 1Mek1 mice, suggesting that the two Mek genes might differentially regulate early hematopoiesis.
Collapse
Affiliation(s)
- Laurent Beuret
- Centre de Recherche sur le Cancer de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval (Oncology), Québec, QC, Canada
| | - Simon-Pierre Fortier-Beaulieu
- Centre de Recherche sur le Cancer de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval (Oncology), Québec, QC, Canada
| | - Vincent Rondeau
- Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160, Paris, France.,OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Sophie Roy
- Centre de Recherche sur le Cancer de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval (Oncology), Québec, QC, Canada
| | - Nicolas Houde
- Centre de Recherche sur le Cancer de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval (Oncology), Québec, QC, Canada
| | - Karl Balabanian
- Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160, Paris, France.,OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Marion Espéli
- Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160, Paris, France.,OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Jean Charron
- Centre de Recherche sur le Cancer de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval (Oncology), Québec, QC, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, QC, Canada
| |
Collapse
|
20
|
Morretta E, Belvedere R, Petrella A, Spallarossa A, Rapetti F, Bruno O, Brullo C, Monti MC. Novel insights on the molecular mechanism of action of the anti-angiogenic pyrazolyl-urea GeGe-3 by functional proteomics. Bioorg Chem 2021; 115:105168. [PMID: 34284173 DOI: 10.1016/j.bioorg.2021.105168] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/28/2021] [Accepted: 07/09/2021] [Indexed: 12/15/2022]
Abstract
In recent years, 5-pyrazolyl-ureas have mostly been known for their attractive poly-pharmacological outline and, in particular, ethyl 1-(2-hydroxypentyl)-5-(3-(3-(trifluoromethyl) phenyl) ureido)-1H-pyrazole-4-carboxylate (named GeGe-3) has emerged as a capable anti-angiogenic compound. This paper examines its interactome by functional proteomics using a label-free mass spectrometry based platform, coupling Drug Affinity Responsive Target Stability and targeted Limited Proteolysis-Multiple Reaction Monitoring. Calreticulin has been recognized as the GeGe-3 principal target and this evidence has been supported by immunoblotting and in silico molecular docking. Furthermore, cell studies have shown that GeGe-3 lowers cell calcium mobilization, cytoskeleton organization and focal adhesion kinase expression, thus linking its biological potential to calreticulin binding and, ultimately, shedding light on the reasonable action mechanism of this molecule as an anti-angiogenic factor.
Collapse
Affiliation(s)
- Elva Morretta
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Salerno, Italy.
| | - Raffaella Belvedere
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Salerno, Italy.
| | - Antonello Petrella
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Salerno, Italy.
| | - Andrea Spallarossa
- Department of Pharmacy, University of Genova, Viale Benedetto XV, 3, 16132 Genova, Italy.
| | - Federica Rapetti
- Department of Pharmacy, University of Genova, Viale Benedetto XV, 3, 16132 Genova, Italy.
| | - Olga Bruno
- Department of Pharmacy, University of Genova, Viale Benedetto XV, 3, 16132 Genova, Italy.
| | - Chiara Brullo
- Department of Pharmacy, University of Genova, Viale Benedetto XV, 3, 16132 Genova, Italy.
| | - Maria Chiara Monti
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Salerno, Italy.
| |
Collapse
|
21
|
Shide K. Calreticulin mutations in myeloproliferative neoplasms. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 365:179-226. [PMID: 34756244 DOI: 10.1016/bs.ircmb.2021.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Calreticulin (CALR) is a chaperone present in the endoplasmic reticulum, which is involved in the quality control of N-glycosylated proteins and storage of calcium ions. In 2013, the C-terminal mutation in CALR was identified in half of the patients with essential thrombocythemia and primary myelofibrosis who did not have a JAK2 or MPL mutation. The results of 8 years of intensive research are changing the clinical practice associated with treating myeloproliferative neoplasms (MPNs). The presence or absence of CALR mutations and their mutation types already provide important information for diagnosis and treatment decision making. In addition, the interaction with the thrombopoietin receptor MPL, which is the main mechanism of transformation by CALR mutation, and the expression of the mutant protein on the cell surface have a great potential as targets for molecular-targeted drugs and immunotherapy. This chapter presents recent findings on the clinical significance of the CALR mutation and the molecular basis by which this mutation drives MPNs.
Collapse
Affiliation(s)
- Kotaro Shide
- Division of Haematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan.
| |
Collapse
|
22
|
Greenfield G, McMullin MF, Mills K. Molecular pathogenesis of the myeloproliferative neoplasms. J Hematol Oncol 2021; 14:103. [PMID: 34193229 PMCID: PMC8246678 DOI: 10.1186/s13045-021-01116-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
The Philadelphia negative myeloproliferative neoplasms (MPN) compromise a heterogeneous group of clonal myeloid stem cell disorders comprising polycythaemia vera, essential thrombocythaemia and primary myelofibrosis. Despite distinct clinical entities, these disorders are linked by morphological similarities and propensity to thrombotic complications and leukaemic transformation. Current therapeutic options are limited in disease-modifying activity with a focus on the prevention of thrombus formation. Constitutive activation of the JAK/STAT signalling pathway is a hallmark of pathogenesis across the disease spectrum with driving mutations in JAK2, CALR and MPL identified in the majority of patients. Co-occurring somatic mutations in genes associated with epigenetic regulation, transcriptional control and splicing of RNA are variably but recurrently identified across the MPN disease spectrum, whilst epigenetic contributors to disease are increasingly recognised. The prognostic implications of one MPN diagnosis may significantly limit life expectancy, whilst another may have limited impact depending on the disease phenotype, genotype and other external factors. The genetic and clinical similarities and differences in these disorders have provided a unique opportunity to understand the relative contributions to MPN, myeloid and cancer biology generally from specific genetic and epigenetic changes. This review provides a comprehensive overview of the molecular pathophysiology of MPN exploring the role of driver mutations, co-occurring mutations, dysregulation of intrinsic cell signalling, epigenetic regulation and genetic predisposing factors highlighting important areas for future consideration.
Collapse
Affiliation(s)
- Graeme Greenfield
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK.
| | | | - Ken Mills
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| |
Collapse
|
23
|
Induced Pluripotent Stem Cells Enable Disease Modeling and Drug Screening in Calreticulin del52 and ins5 Myeloproliferative Neoplasms. Hemasphere 2021; 5:e593. [PMID: 34131633 PMCID: PMC8196125 DOI: 10.1097/hs9.0000000000000593] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Mutations in the calreticulin (CALR) gene are seen in about 30% of essential thrombocythemia and primary myelofibrosis patients. To address the contribution of the human CALR mutants to the pathogenesis of myeloproliferative neoplasms (MPNs) in an endogenous context, we modeled the CALRdel52 and CALRins5 mutants by induced pluripotent stem cell (iPSC) technology using CD34+ progenitors from 4 patients. We describe here the generation of several clones of iPSC carrying heterozygous CALRdel52 or CALRins5 mutations. We showed that CALRdel52 induces a stronger increase in progenitors than CALRins5 and that both CALRdel52 and CALRins5 mutants favor an expansion of the megakaryocytic lineage. Moreover, we found that both CALRdel52 and CALRins5 mutants rendered colony forming unit–megakaryocyte (CFU-MK) independent from thrombopoietin (TPO), and promoted a mild constitutive activation level of signal transducer and activator of transcription 3 in megakaryocytes. Unexpectedly, a mild increase in the sensitivity of colony forming unit-granulocyte (CFU-G) to granulocyte-colony stimulating factor was also observed in iPSC CALRdel52 and CALRins5 compared with control iPSC. Moreover, CALRdel52-induced megakaryocytic spontaneous growth is more dependent on Janus kinase 2/phosphoinositide 3-kinase/extracellular signal-regulated kinase than TPO-mediated growth and opens a therapeutic window for treatments in CALR-mutated MPN. The iPSC models described here represent an interesting platform for testing newly developed inhibitors. Altogether, this study shows that CALR-mutated iPSC recapitulate MPN phenotypes in vitro and may be used for drug screening.
Collapse
|
24
|
Activated IL-6 signaling contributes to the pathogenesis of, and is a novel therapeutic target for, CALR-mutated MPNs. Blood Adv 2021; 5:2184-2195. [PMID: 33890979 DOI: 10.1182/bloodadvances.2020003291] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 03/14/2021] [Indexed: 02/08/2023] Open
Abstract
Calreticulin (CALR), an endoplasmic reticulum-associated chaperone, is frequently mutated in myeloproliferative neoplasms (MPNs). Mutated CALR promotes downstream JAK2/STAT5 signaling through interaction with, and activation of, the thrombopoietin receptor (MPL). Here, we provide evidence of a novel mechanism contributing to CALR-mutated MPNs, represented by abnormal activation of the interleukin 6 (IL-6)-signaling pathway. We found that UT7 and UT7/mpl cells, engineered by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) to express the CALR type 1-like (DEL) mutation, acquired cytokine independence and were primed to the megakaryocyte (Mk) lineage. Levels of IL-6 messenger RNA (mRNA), extracellular-released IL-6, membrane-associated glycoprotein 130 (gp130), and IL-6 receptor (IL-6R), phosphorylated JAK1 and STAT3 (p-JAK1 and p-STAT3), and IL-6 promoter region occupancy by STAT3 all resulted in increased CALR DEL cells in the absence of MPL stimulation. Wild-type, but not mutated, CALR physically interacted with gp130 and IL-6R, downregulating their expression on the cell membrane. Agents targeting gp130 (SC-144), IL-6R (tocilizumab [TCZ]), and cell-released IL-6 reduced proliferation of CALR DEL as well as CALR knockout cells, supporting a mutated CALR loss-of-function model. CD34+ cells from CALR-mutated patients showed increased levels of IL-6 mRNA and p-STAT3, and colony-forming unit-Mk growth was inhibited by either SC144 or TCZ, as well as an IL-6 antibody, supporting cell-autonomous activation of the IL-6 pathway. Targeting IL-6 signaling also reduced colony formation by CD34+ cells of JAK2V617F-mutated patients. The combination of TCZ and ruxolitinib was synergistic at very low nanomolar concentrations. Overall, our results suggest that target inhibition of IL-6 signaling may have therapeutic potential in CALR, and possibly JAK2V617F, mutated MPNs.
Collapse
|
25
|
Venkatesan A, Geng J, Kandarpa M, Wijeyesakere SJ, Bhide A, Talpaz M, Pogozheva ID, Raghavan M. Mechanism of mutant calreticulin-mediated activation of the thrombopoietin receptor in cancers. J Cell Biol 2021; 220:212031. [PMID: 33909030 PMCID: PMC8085772 DOI: 10.1083/jcb.202009179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/10/2021] [Accepted: 03/17/2021] [Indexed: 12/21/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are frequently driven by mutations within the C-terminal domain (C-domain) of calreticulin (CRT). CRTDel52 and CRTIns5 are recurrent mutations. Oncogenic transformation requires both mutated CRT and the thrombopoietin receptor (Mpl), but the molecular mechanism of CRT-mediated constitutive activation of Mpl is unknown. We show that the acquired C-domain of CRTDel52 mediates both Mpl binding and disulfide-linked CRTDel52 dimerization. Cysteine mutations within the novel C-domain (C400A and C404A) and the conserved N-terminal domain (N-domain; C163A) of CRTDel52 are required to reduce disulfide-mediated dimers and multimers of CRTDel52. Based on these data and published structures of CRT oligomers, we identify an N-domain dimerization interface relevant to both WT CRT and CRTDel52. Elimination of disulfide bonds and ionic interactions at both N-domain and C-domain dimerization interfaces is required to abrogate the ability of CRTDel52 to mediate cell proliferation via Mpl. Thus, MPNs exploit a natural dimerization interface of CRT combined with C-domain gain of function to achieve cell transformation.
Collapse
Affiliation(s)
- Arunkumar Venkatesan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Jie Geng
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Malathi Kandarpa
- Department of Internal Medicine/Division of Hematology/Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | | | - Ashwini Bhide
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Moshe Talpaz
- Department of Internal Medicine/Division of Hematology/Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Irina D Pogozheva
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI
| | - Malini Raghavan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
26
|
Alshemmari SH, Rajan R, Ameen R, Almazyad M. Analysis of Common Driver Mutations in Philadelphia-Negative Myeloproliferative Neoplasms. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 21:483-488. [PMID: 33858806 DOI: 10.1016/j.clml.2021.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/04/2021] [Accepted: 03/15/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Philadelphia-negative myeloproliferative neoplasms (MPNs) are a group of hematopoietic stem cell disorders that include polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). This study examines the driver mutations among patients with MPNs in Kuwait. PATIENTS AND METHODS This study was a retrospective review of 942 MPN cases with a driver mutation from July 2007 to June 2019 to examine their demographic, clinical, and laboratory attributes. RESULTS The annual incidence of MPNs is 1.6 per 100,000 persons, and ET is the most common subtype. The median age of our cohort was 55 years, and the patients were predominantly male. We found that the most frequent gene mutation of MPNs in our cohort was the JAK2V617F mutation, which was present in 90% of cases, followed by the CALR exon 9, MPLW515L/K, and JAK2 exon 12 mutations. In our cohort, thrombotic events were observed in 18.7% of cases. CONCLUSION Although Philadelphia-negative MPNs are rare hematologic malignancies, thrombosis is a relatively common initial presentation. The JAK2V617F mutation was the driver mutation in the majority of patients with MPN.
Collapse
Affiliation(s)
- Salem H Alshemmari
- Department of Medicine, Faculty of Medicine, Kuwait University, Jabriya, Kuwait.
| | - Reshmi Rajan
- Department of Hematology, Kuwait Cancer Center, Shuwiakh, Jabriya
| | - Reem Ameen
- Department of Medical Laboratory Sciences, Faculty of Allied Sciences, Kuwait University, Jabriya, Kuwait
| | - Mazyad Almazyad
- Seventh Year Medical Student, Faculty of Medicine, Kuwait University, Jabriya, Kuwait
| |
Collapse
|
27
|
Benlabiod C, Cacemiro MDC, Nédélec A, Edmond V, Muller D, Rameau P, Touchard L, Gonin P, Constantinescu SN, Raslova H, Villeval JL, Vainchenker W, Plo I, Marty C. Calreticulin del52 and ins5 knock-in mice recapitulate different myeloproliferative phenotypes observed in patients with MPN. Nat Commun 2020; 11:4886. [PMID: 32985500 PMCID: PMC7522233 DOI: 10.1038/s41467-020-18691-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 09/04/2020] [Indexed: 12/20/2022] Open
Abstract
Somatic mutations in the calreticulin (CALR) gene are associated with approximately 30% of essential thrombocythemia (ET) and primary myelofibrosis (PMF). CALR mutations, including the two most frequent 52 bp deletion (del52) and 5 bp insertion (ins5), induce a frameshift to the same alternative reading frame generating new C-terminal tails. In patients, del52 and ins5 induce two phenotypically distinct myeloproliferative neoplasms (MPNs). They are equally found in ET, but del52 is more frequent in PMF. We generated heterozygous and homozygous conditional inducible knock-in (KI) mice expressing a chimeric murine CALR del52 or ins5 with the human mutated C-terminal tail to investigate their pathogenic effects on hematopoiesis. Del52 induces greater phenotypic changes than ins5 including thrombocytosis, leukocytosis, splenomegaly, bone marrow hypocellularity, megakaryocytic lineage amplification, expansion and competitive advantage of the hematopoietic stem cell compartment. Homozygosity amplifies these features, suggesting a distinct contribution of homozygous clones to human MPNs. Moreover, homozygous del52 KI mice display features of a penetrant myelofibrosis-like disorder with extramedullary hematopoiesis linked to splenomegaly, megakaryocyte hyperplasia and the presence of reticulin fibers. Overall, modeling del52 and ins5 mutations in mice successfully recapitulates the differences in phenotypes observed in patients. Calreticulin del52 and ins5 mutations induce two phenotypically distinct myeloproliferative neoplasms in patients. Here the authors show that modeling these mutations in knock-in mice recapitulate the two diseases and highlight how they impact the different hematopoietic compartments.
Collapse
Affiliation(s)
- Camélia Benlabiod
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Maira da Costa Cacemiro
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France.,Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, São Paulo, Brazil
| | - Audrey Nédélec
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Université catholique de Louvain, Brussels, Belgium
| | - Valérie Edmond
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Delphine Muller
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Philippe Rameau
- Integrated Biology Core Facility, Gustave Roussy, Villejuif, France
| | - Laure Touchard
- Preclinical Research Plateform, Unité Mixte de Service AMMICA 3655/US 23, Gustave Roussy, Villejuif, France
| | - Patrick Gonin
- Preclinical Research Plateform, Unité Mixte de Service AMMICA 3655/US 23, Gustave Roussy, Villejuif, France
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Université catholique de Louvain, Brussels, Belgium
| | - Hana Raslova
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Jean-Luc Villeval
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - William Vainchenker
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Caroline Marty
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France. .,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France. .,Gustave Roussy, UMR 1287, Villejuif, France.
| |
Collapse
|
28
|
Abstract
Calreticulin (CALR) is an endoplasmic reticulum (ER)-resident protein involved in a spectrum of cellular processes. In healthy cells, CALR operates as a chaperone and Ca2+ buffer to assist correct protein folding within the ER. Besides favoring the maintenance of cellular proteostasis, these cell-intrinsic CALR functions support Ca2+-dependent processes, such as adhesion and integrin signaling, and ensure normal antigen presentation on MHC Class I molecules. Moreover, cancer cells succumbing to immunogenic cell death (ICD) expose CALR on their surface, which promotes the uptake of cell corpses by professional phagocytes and ultimately supports the initiation of anticancer immunity. Thus, loss-of-function CALR mutations promote oncogenesis not only as they impair cellular homeostasis in healthy cells, but also as they compromise natural and therapy-driven immunosurveillance. However, the prognostic impact of total or membrane-exposed CALR levels appears to vary considerably with cancer type. For instance, while genetic CALR defects promote pre-neoplastic myeloproliferation, patients with myeloproliferative neoplasms bearing CALR mutations often experience improved overall survival as compared to patients bearing wild-type CALR. Here, we discuss the context-dependent impact of CALR on malignant transformation, tumor progression and response to cancer therapy.
Collapse
|
29
|
Different impact of calreticulin mutations on human hematopoiesis in myeloproliferative neoplasms. Oncogene 2020; 39:5323-5337. [PMID: 32572159 DOI: 10.1038/s41388-020-1368-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 01/05/2023]
Abstract
Mutations of calreticulin (CALRm) define a subtype of myeloproliferative neoplasms (MPN). We studied the biological and genetic features of CALR-mutated essential thrombocythemia and myelofibrosis patients. In most cases, CALRm were found in granulocytes, monocytes, B and NK cells, but also in T cells. However, the type 1 CALRm spreads more easily than the type 2 CALRm in lymphoid cells. The CALRm were also associated with an early clonal dominance at the level of hematopoietic stem and progenitor cells (HSPC) with no significant increase during granulo/monocytic differentiation in most cases. Moreover, we found that half of type 2 CALRm patients harbors some homozygous progenitors. Those patients were associated with a higher clonal dominance during granulo/monocytic differentiation than patients with only heterozygous type 2 CALRm progenitors. When associated mutations were present, CALRm were the first genetic event suggesting that they are both the initiating and phenotypic event. In blood, type 1 CALRm led to a greater increased number of all types of progenitors compared with the type 2 CALRm. However, both types of CALRm induced an increase in megakaryocytic progenitors associated with a ruxolitinib-sensitive independent growth and with a mild constitutive signaling in megakaryocytes. At the transcriptional level, type 1 CALRm seems to deregulate more pathways than the type 2 CALRm in megakaryocytes. Altogether, our results show that CALRm modify both the HSPC and megakaryocyte biology with a stronger effect for type 1 than for type 2 CALRm.
Collapse
|
30
|
Saleh LM, Algamal R, Abd Elmasseh H, Barber E, Abdel-ghaffar H. Different CALR mutation subtypes in essential thrombocythemia and primary myelofibrosis patients without JAK2 mutation. MEMO - MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2020; 13:235-243. [DOI: 10.1007/s12254-020-00584-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/11/2020] [Indexed: 09/02/2023]
|
31
|
Kurtovic-Kozaric A, Islamagic E, Komic H, Bilalovic N, Eminovic I, Burekovic A, Uzunovic A, Kurtovic S. The effects of mutational profiles on phenotypic presentation of myeloproliferative neoplasm subtypes in Bosnia: 18 year follow-up. Bosn J Basic Med Sci 2020; 20:236-247. [PMID: 31668145 PMCID: PMC7202189 DOI: 10.17305/bjbms.2019.4391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/25/2019] [Indexed: 11/28/2022] Open
Abstract
The identification of mutually exclusive somatic mutations shared among myeloproliferative neoplasm (MPN) subtypes has provided a powerful tool for studying disease evolution. Clinical features, gene mutations, and survival over 18 years were analyzed in MPN patients. One hundred thirty-eight MPN patients were subcategorized according to MPN subtypes: essential thrombocythemia (ET, n = 41), polycythemia vera (PV, n = 56), primary myelofibrosis (PMF, n = 10), and MPN unclassified (MPN-U, n = 31). Patient characteristics included clinical parameters, overall survival (OS), and mutational status of the Janus kinase 2 (JAK2), calreticulin (CALR), and myeloproliferative leukemia virus oncogene (MPL) genes. We compared hematologic and clinical features of JAK2V617F-ET vs. CALR-mutated ET vs. JAK2V617F-PV patients. JAK2V617F-patients had higher values of erythrocytes, hemoglobin, and hematocrit compared to CALR-mutated patients (p < 0.05). The mutant allele burden in JAK2V617F-PV and JAK2V617F-ET patients directly correlated with erythrocyte, hemoglobin, and hematocrit values, but it inversely correlated with platelet count. Thus, mutant allele burden was an indicator of the clinical phenotype in JAK2V617F-MPN patients. OS was not affected by the mutational status. In general, mutated JAK2, CALR, and MPL genes left specific hematological signatures.
Collapse
Affiliation(s)
- Amina Kurtovic-Kozaric
- Department of Clinical Pathology, Cytology and Human Genetics, Clinical Center of the University of Sarajevo, Sarajevo, Bosnia and Herzegovina; Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Erna Islamagic
- Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Hana Komic
- Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Nurija Bilalovic
- Department of Clinical Pathology, Cytology and Human Genetics, Clinical Center of the University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Izet Eminovic
- Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Adnan Burekovic
- Department of Internal Medicine, Clinical Hospital, Zenica, Bosnia and Herzegovina
| | - Amna Uzunovic
- Department of Internal Medicine, Clinical Hospital, Zenica, Bosnia and Herzegovina
| | - Sabira Kurtovic
- Department of Hematology, Clinical Center of the University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
32
|
Mutant Calreticulin in the Myeloproliferative Neoplasms. Hemasphere 2020; 4:e333. [PMID: 32382708 PMCID: PMC7000472 DOI: 10.1097/hs9.0000000000000333] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/03/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022] Open
Abstract
Mutations in the gene for calreticulin (CALR) were identified in the myeloproliferative neoplasms (MPNs) essential thrombocythaemia (ET) and primary myelofibrosis (MF) in 2013; in combination with previously described mutations in JAK2 and MPL, driver mutations have now been described for the majority of MPN patients. In subsequent years, researchers have begun to unravel the mechanisms by which mutant CALR drives transformation and to understand their clinical implications. Mutant CALR activates the thrombopoietin receptor (MPL), causing constitutive activation of Janus kinase 2 (JAK2) signaling and cytokine independent growth in vitro. Mouse models show increased numbers of hematopoietic stem cells (HSCs) and overproduction of megakaryocytic lineage cells with associated thrombocytosis. In the clinic, detection of CALR mutations has been embedded in World Health Organization and other international diagnostic guidelines. Distinct clinical and laboratory associations of CALR mutations have been identified together with their prognostic significance, with CALR mutant patients showing increased overall survival. The discovery and subsequent study of CALR mutations have illuminated novel aspects of megakaryopoiesis and raised the possibility of new therapeutic approaches.
Collapse
|
33
|
The role of calreticulin mutations in myeloproliferative neoplasms. Int J Hematol 2019; 111:200-205. [DOI: 10.1007/s12185-019-02800-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022]
|
34
|
The Expression of Myeloproliferative Neoplasm-Associated Calreticulin Variants Depends on the Functionality of ER-Associated Degradation. Cancers (Basel) 2019; 11:cancers11121921. [PMID: 31810292 PMCID: PMC6966542 DOI: 10.3390/cancers11121921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Mutations in CALR observed in myeloproliferative neoplasms (MPN) were recently shown to be pathogenic via their interaction with MPL and the subsequent activation of the Janus Kinase – Signal Transducer and Activator of Transcription (JAK-STAT) pathway. However, little is known on the impact of those variant CALR proteins on endoplasmic reticulum (ER) homeostasis. Methods: The impact of the expression of Wild Type (WT) or mutant CALR on ER homeostasis was assessed by quantifying the expression level of Unfolded Protein Response (UPR) target genes, splicing of X-box Binding Protein 1 (XBP1), and the expression level of endogenous lectins. Pharmacological and molecular (siRNA) screens were used to identify mechanisms involved in CALR mutant proteins degradation. Coimmunoprecipitations were performed to define more precisely actors involved in CALR proteins disposal. Results: We showed that the expression of CALR mutants alters neither ER homeostasis nor the sensitivity of hematopoietic cells towards ER stress-induced apoptosis. In contrast, the expression of CALR variants is generally low because of a combination of secretion and protein degradation mechanisms mostly mediated through the ER-Associated Degradation (ERAD)-proteasome pathway. Moreover, we identified a specific ERAD network involved in the degradation of CALR variants. Conclusions: We propose that this ERAD network could be considered as a potential therapeutic target for selectively inhibiting CALR mutant-dependent proliferation associated with MPN, and therefore attenuate the associated pathogenic outcomes.
Collapse
|
35
|
Nam AS, Kim KT, Chaligne R, Izzo F, Ang C, Taylor J, Myers RM, Abu-Zeinah G, Brand R, Omans ND, Alonso A, Sheridan C, Mariani M, Dai X, Harrington E, Pastore A, Cubillos-Ruiz JR, Tam W, Hoffman R, Rabadan R, Scandura JM, Abdel-Wahab O, Smibert P, Landau DA. Somatic mutations and cell identity linked by Genotyping of Transcriptomes. Nature 2019; 571:355-360. [PMID: 31270458 PMCID: PMC6782071 DOI: 10.1038/s41586-019-1367-0] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 06/05/2019] [Indexed: 02/06/2023]
Abstract
Defining the transcriptomic identity of malignant cells is challenging in the absence of surface markers that distinguish cancer clones from one another, or from admixed non-neoplastic cells. To address this challenge, here we developed Genotyping of Transcriptomes (GoT), a method to integrate genotyping with high-throughput droplet-based single-cell RNA sequencing. We apply GoT to profile 38,290 CD34+ cells from patients with CALR-mutated myeloproliferative neoplasms to study how somatic mutations corrupt the complex process of human haematopoiesis. High-resolution mapping of malignant versus normal haematopoietic progenitors revealed an increasing fitness advantage with myeloid differentiation of cells with mutated CALR. We identified the unfolded protein response as a predominant outcome of CALR mutations, with a considerable dependency on cell identity, as well as upregulation of the NF-κB pathway specifically in uncommitted stem cells. We further extended the GoT toolkit to genotype multiple targets and loci that are distant from transcript ends. Together, these findings reveal that the transcriptional output of somatic mutations in myeloproliferative neoplasms is dependent on the native cell identity.
Collapse
Affiliation(s)
- Anna S Nam
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Kyu-Tae Kim
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ronan Chaligne
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Franco Izzo
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Chelston Ang
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Justin Taylor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert M Myers
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional MD-PhD Program, Weill Cornell Medicine, Rockefeller University, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ghaith Abu-Zeinah
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Richard T. Silver MD Myeloproliferative Neoplasms Center, Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ryan Brand
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Nathaniel D Omans
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional Training Program in Computational Biology and Medicine, Memorial Sloan Kettering Cancer Center, Cornell University, Weill Cornell Medicine, New York, NY, USA
| | - Alicia Alonso
- Epigenomics Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Caroline Sheridan
- Epigenomics Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Marisa Mariani
- Epigenomics Core Facility, Weill Cornell Medicine, New York, NY, USA
| | | | | | - Alessandro Pastore
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juan R Cubillos-Ruiz
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
| | - Wayne Tam
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ronald Hoffman
- Division of Hematology and Medical Oncology, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raul Rabadan
- Department of Systems Biology, Columbia University Medical Center, New York, NY, USA
| | - Joseph M Scandura
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Richard T. Silver MD Myeloproliferative Neoplasms Center, Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Peter Smibert
- Technology Innovation Lab, New York Genome Center, New York, NY, USA
| | - Dan A Landau
- New York Genome Center, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
36
|
Merlinsky TR, Levine RL, Pronier E. Unfolding the Role of Calreticulin in Myeloproliferative Neoplasm Pathogenesis. Clin Cancer Res 2019; 25:2956-2962. [PMID: 30655313 DOI: 10.1158/1078-0432.ccr-18-3777] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/18/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022]
Abstract
In 2013, two seminal studies identified gain-of-function mutations in the Calreticulin (CALR) gene in a subset of JAK2/MPL-negative myeloproliferative neoplasm (MPN) patients. CALR is an endoplasmic reticulum (ER) chaperone protein that normally binds misfolded proteins in the ER and prevents their export to the Golgi and had never previously been reported mutated in cancer or to be associated with hematologic disorders. Further investigation determined that mutated CALR is able to achieve oncogenic transformation primarily through constitutive activation of the MPL-JAK-STAT signaling axis. Here we review our current understanding of the role of CALR mutations in MPN pathogenesis and how these insights can lead to innovative therapeutics approaches.
Collapse
Affiliation(s)
- Tiffany R Merlinsky
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ross L Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. .,Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York.,Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elodie Pronier
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
37
|
Greenfield G, McPherson S, Mills K, McMullin MF. The ruxolitinib effect: understanding how molecular pathogenesis and epigenetic dysregulation impact therapeutic efficacy in myeloproliferative neoplasms. J Transl Med 2018; 16:360. [PMID: 30558676 PMCID: PMC6296062 DOI: 10.1186/s12967-018-1729-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 12/05/2018] [Indexed: 12/20/2022] Open
Abstract
The myeloproliferative neoplasms (MPN), polycythaemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF) are linked by a propensity to thrombosis formation and a risk of leukaemic transformation. Activation of cytokine independent signalling through the JAK/STAT cascade is a feature of these disorders. A point mutation in exon 14 of the JAK2 gene resulting in the formation of the JAK2 V617F transcript occurs in 95% of PV patients and around 50% of ET and PMF patients driving constitutive activation of the JAK/STAT pathway. Mutations in CALR or MPL are present as driving mutations in the majority of remaining ET and PMF patients. Ruxolitinib is a tyrosine kinase inhibitor which inhibits JAK1 and JAK2. It is approved for use in intermediate and high risk PMF, and in PV patients who are resistant or intolerant to hydroxycarbamide. In randomised controlled trials it has demonstrated efficacy in spleen volume reduction and symptom burden reduction with a moderate improvement in overall survival in PMF. In PV, there is demonstrated benefit in haematocrit control and spleen volume. Despite these benefits, there is limited impact to induce complete haematological remission with normalisation of blood counts, reduce the mutant allele burden or reverse bone marrow fibrosis. Clonal evolution has been observed on ruxolitinib therapy and transformation to acute leukaemia can still occur. This review will concentrate on understanding the clinical and molecular effects of ruxolitinib in MPN. We will focus on understanding the limitations of JAK inhibition and the challenges to improving therapeutic efficacy in these disorders. We will explore the demonstrated benefits and disadvantages of ruxolitinib in the clinic, the role of genomic and clonal variability in pathogenesis and response to JAK inhibition, epigenetic changes which impact on response to therapy, the role of DNA damage and the role of inflammation in these disorders. Finally, we will summarise the future prospects for improving therapy in MPN in the JAK inhibition era.
Collapse
Affiliation(s)
- Graeme Greenfield
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, UK
| | - Suzanne McPherson
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, UK
| | - Ken Mills
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, UK
| | | |
Collapse
|
38
|
O'Sullivan J, Mead AJ. Heterogeneity in myeloproliferative neoplasms: Causes and consequences. Adv Biol Regul 2018; 71:55-68. [PMID: 30528537 DOI: 10.1016/j.jbior.2018.11.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 01/09/2023]
Abstract
Myeloproliferative neoplasms (MPNs) are haematopoietic stem cell-derived clonal disorders characterised by proliferation of some or all myeloid lineages, depending on the subtype. MPNs are classically categorized into three disease subgroups; essential thrombocythaemia (ET), polycythaemia vera (PV) and primary myelofibrosis (PMF). The majority (>85%) of patients carry a disease-initiating or driver mutation, the most prevalent occurring in the janus kinase 2 gene (JAK2 V617F), followed by calreticulin (CALR) and myeloproliferative leukaemia virus (MPL) genes. Although these diseases are characterised by shared clinical, pathological and molecular features, one of the most challenging aspects of these disorders is the diverse clinical features which occur in each disease type, with marked variability in risks of disease complications and progression to leukaemia. A remarkable aspect of MPN biology is that the JAK2 V617F mutation, often occurring in the absence of additional mutations, generates a spectrum of phenotypes from asymptomatic ET through to aggressive MF, associated with a poor outcome. The mechanisms promoting MPN heterogeneity remain incompletely understood, but contributing factors are broad and include patient characteristics (gender, age, comorbidities and environmental exposures), additional somatic mutations, target disease-initiating cell, bone marrow microenvironment and germline genetic associations. In this review, we will address these in detail and discuss their role in heterogeneity of MPN disease phenotypes. Tailoring patient management according to the multiple different factors that influence disease phenotype may prove to be the most effective approach to modify the natural history of the disease and ultimately improve outcomes for patients.
Collapse
Affiliation(s)
- Jennifer O'Sullivan
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, United Kingdom.
| | - Adam J Mead
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, United Kingdom; NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
39
|
Pronier E, Cifani P, Merlinsky TR, Berman KB, Somasundara AVH, Rampal RK, LaCava J, Wei KE, Pastore F, Maag JL, Park J, Koche R, Kentsis A, Levine RL. Targeting the CALR interactome in myeloproliferative neoplasms. JCI Insight 2018; 3:122703. [PMID: 30429377 DOI: 10.1172/jci.insight.122703] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 09/19/2018] [Indexed: 02/06/2023] Open
Abstract
Mutations in the ER chaperone calreticulin (CALR) are common in myeloproliferative neoplasm (MPN) patients, activate the thrombopoietin receptor (MPL), and mediate constitutive JAK/STAT signaling. The mechanisms by which CALR mutations cause myeloid transformation are incompletely defined. We used mass spectrometry proteomics to identify CALR-mutant interacting proteins. Mutant CALR caused mislocalization of binding partners and increased recruitment of FLI1, ERP57, and CALR to the MPL promoter to enhance transcription. Consistent with a critical role for CALR-mediated JAK/STAT activation, we confirmed the efficacy of JAK2 inhibition on CALR-mutant cells in vitro and in vivo. Due to the altered interactome induced by CALR mutations, we hypothesized that CALR-mutant MPNs may be vulnerable to disruption of aberrant CALR protein complexes. A synthetic peptide designed to competitively inhibit the carboxy terminal of CALR specifically abrogated MPL/JAK/STAT signaling in cell lines and primary samples and improved the efficacy of JAK kinase inhibitors. These findings reveal what to our knowledge is a novel potential therapeutic approach for patients with CALR-mutant MPN.
Collapse
Affiliation(s)
- Elodie Pronier
- Human Oncology and Pathogenesis Program.,Center for Epigenetics Research, and
| | - Paolo Cifani
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, New York, USA
| | - Tiffany R Merlinsky
- Human Oncology and Pathogenesis Program.,Center for Epigenetics Research, and
| | | | | | - Raajit K Rampal
- Human Oncology and Pathogenesis Program.,Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - John LaCava
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Karen E Wei
- Human Oncology and Pathogenesis Program.,Center for Epigenetics Research, and
| | - Friederike Pastore
- Human Oncology and Pathogenesis Program.,Center for Epigenetics Research, and
| | | | - Jane Park
- Center for Epigenetics Research, and
| | | | - Alex Kentsis
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, New York, USA.,Department of Pediatrics, Weill Cornell Medical College of Cornell University and Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program.,Center for Epigenetics Research, and.,Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
40
|
Mutant NRAS Q61K is responsible for MAPK pathway activation in the MARIMO cell line and renders these cells independent of the CALR-MPL-JAK2-STAT5 pathway. Leukemia 2018; 32:2087-2090. [PMID: 30115971 DOI: 10.1038/s41375-018-0234-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 07/10/2018] [Accepted: 07/23/2018] [Indexed: 11/09/2022]
|
41
|
AKT activation is a feature of CALR mutant myeloproliferative neoplasms. Leukemia 2018; 33:271-274. [PMID: 30082822 DOI: 10.1038/s41375-018-0224-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 07/06/2018] [Accepted: 07/11/2018] [Indexed: 12/29/2022]
|
42
|
Lim KH, Chen CGS, Chang YC, Chiang YH, Kao CW, Wang WT, Chang CY, Huang L, Lin CS, Cheng CC, Cheng HI, Su NW, Lin J, Chang YF, Chang MC, Hsieh RK, Lin HC, Kuo YY. Increased B cell activation is present in JAK2V617F-mutated, CALR-mutated and triple-negative essential thrombocythemia. Oncotarget 2018; 8:32476-32491. [PMID: 28415571 PMCID: PMC5464803 DOI: 10.18632/oncotarget.16381] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 02/28/2017] [Indexed: 01/19/2023] Open
Abstract
Essential thrombocythemia (ET) is a BCL-ABL1-negative myeloproliferative neoplasm. We have reported that increased activated B cells can facilitate platelet production mediated by cytokines regardless JAK2 mutational status in ET. Recently, calreticulin (CALR) mutations were discovered in ~30% JAK2/MPL-unmutated ET and primary myelofibrosis. Here we sought to screen for CALR mutations and to evaluate B cell immune profiles in a cohort of adult Taiwanese ET patients. B cell populations, granulocytes/monocytes membrane-bound B cell-activating factor (mBAFF) levels, B cells toll-like receptor 4 (TLR4) expression and intracellular levels of interleukin (IL)-1β/IL-6 and the expression of CD69, CD80, and CD86 were quantified by flow cytometry. Serum BAFF concentration was measured by ELISA. 48 healthy adults were used for comparison. 19 (35.2%) of 54 ET patients harbored 8 types of CALR exon 9 mutations including 4 (7.4%) patients with concomitant JAK2V617F mutations. Compared to JAK2V617F mutation, CALR mutations correlated with younger age at diagnosis (p=0.04), higher platelet count (p=0.004), lower hemoglobin level (p=0.013) and lower leukocyte count (p=0.013). Multivariate analysis adjusted for age, sex, follow-up period and hematological parameters confirmed that increased activated B cells were universally present in JAK2-mutated, CALR-mutated and triple-negative ET patients when compared to healthy adults. JAK2- and CALR-mutated ET have significantly higher fraction of B cells with TLR4 expression when compared to triple-negative ET (p=0.019 and 0.02, respectively). CALR-mutated ET had significantly higher number of CD69-positive activated B cells when compared to triple-negative ET (p=0.035). In conclusion, increased B cell activation is present in ET patients across different mutational subgroups.
Collapse
Affiliation(s)
- Ken-Hong Lim
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Internal Medicine, Division of Hematology and Oncology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Caleb Gon-Shen Chen
- Department of Internal Medicine, Division of Hematology and Oncology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.,Institute of Molecular and Cellular Biology, National Tsing-Hua University, Hsinchu, Taiwan
| | - Yu-Cheng Chang
- Department of Internal Medicine, Division of Hematology and Oncology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Yi-Hao Chiang
- Department of Internal Medicine, Division of Hematology and Oncology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Chen-Wei Kao
- Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Wei-Ting Wang
- Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Chiao-Yi Chang
- Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Ling Huang
- Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Ching-Sung Lin
- Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Chun-Chia Cheng
- Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Hung-I Cheng
- Department of Internal Medicine, Division of Hematology and Oncology, MacKay Memorial Hospital, Hsinchu, Taiwan
| | - Nai-Wen Su
- Department of Internal Medicine, Division of Hematology and Oncology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Johnson Lin
- Department of Internal Medicine, Division of Hematology and Oncology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yi-Fang Chang
- Department of Internal Medicine, Division of Hematology and Oncology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Ming-Chih Chang
- Department of Internal Medicine, Division of Hematology and Oncology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Ruey-Kuen Hsieh
- Department of Internal Medicine, Division of Hematology and Oncology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Huan-Chau Lin
- Department of Internal Medicine, Division of Hematology and Oncology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Laboratory of Good Clinical Research Center, MacKay Memorial Hospital, Tamsui District, New Taipei City, Taiwan
| | - Yuan-Yeh Kuo
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
43
|
Mughal TI, Gotlib J, Mesa R, Koschmieder S, Khoury HJ, Cortes JE, Barbui T, Hehlmann R, Mauro M, Saussele S, Radich JP, Van Etten RA, Saglio G, Verstovek S, Gale RP, Abdel-Wahab O. Recent advances in the genomics and therapy of BCR/ABL1-positive and -negative chronic myeloproliferative neoplasms. Leuk Res 2018; 67:67-74. [PMID: 29466766 PMCID: PMC6613209 DOI: 10.1016/j.leukres.2018.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 01/08/2023]
Abstract
This review is based on the presentations and deliberations at the 7th John Goldman Chronic Myeloid Leukemia (CML) and Myeloproliferative Neoplasms (MPN) Colloquium which took place in Estoril, Portugal on the 15th October 2017, and the 11th post-ASH International Workshop on CML and MPN which took place on the 6th-7th December 2016, immediately after the 58th American Society of Hematology Annual Meeting. Rather than present a resume of the proceedings, we have elected to address some of the topical translational research and clinically relevant topics in greater detail. We address recent updates in the genetics and epigenetics of MPN, the mechanisms of transformation by mutant calreticulin, advances in the biology and therapy of systemic mastocytosis, clinical updates on JAK2 inhibitors and other therapeutic approaches for patients with MPNs, cardiovascular toxicity related to tyrosine kinase inhibitors and the concept of treatment-free remission for patients with CML.
Collapse
Affiliation(s)
| | | | - Ruben Mesa
- UT Health San Antonio Cancer Center, San Antonio, TX, USA
| | | | | | | | - Tiziano Barbui
- Papa Giovani XXIII Hospital and Research Center, Bergamo, Italy
| | | | - Michael Mauro
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Jerald P Radich
- Fredreick Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
Essential thrombocythemia (ET) is an indolent myeloproliferative neoplasm characterized by megakaryocyte hyperplasia, thrombocytosis, thrombotic and hemorrhagic complications, and potential transformation into myelofibrosis and acute myeloid leukemia. The vast majority of cases are driven by a somatic mutation in JAK2, CALR, or MPL. CALR, a gene that codes for the calcium-binding chaperone calreticulin, is the predominant mutation in patients with non-mutated JAK2 essential thrombocythemia, accounting for 20-25% of the overall somatic mutation frequency in ET. In this brief review of ET, we introduce a rare CALR mutation through a case presentation of a 58-year-old man with diffuse pulmonary emboli in the setting of thrombocytosis. We subsequently characterize the main types of CALR mutations and their value in diagnosis and prognosis of disease course, and lastly discuss the current clinical approach to ET.
Collapse
|
45
|
Rosso V, Petiti J, Bracco E, Pedrola R, Carnuccio F, Signorino E, Carturan S, Calabrese C, Bot-Sartor G, Ronconi M, Serra A, Saglio G, Frassoni F, Cilloni D. A novel assay to detect calreticulin mutations in myeloproliferative neoplasms. Oncotarget 2018; 8:6399-6405. [PMID: 28031530 PMCID: PMC5351640 DOI: 10.18632/oncotarget.14113] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 12/15/2016] [Indexed: 11/27/2022] Open
Abstract
The myeloproliferative neoplasms are chronic myeloid cancers divided in Philadelphia positive (Ph+), chronic myeloid leukemia, or negative: polycythemia vera (PV) essential thrombocythemia (ET), and primary myelofibrosis (PMF). Most Ph negative cases have an activating JAK2 or MPL mutation. Recently, somatic mutations in the calreticulin gene (CALR) were detected in 56–88% of JAK2/MPL-negative patients affected by ET or PMF. The most frequent mutations in CARL gene are type-1 and 2. Currently, CALR mutations are evaluated by sanger sequencing. The evaluation of CARL mutations increases the diagnostic accuracy in patients without other molecular markers and could represent a new therapeutic target for molecular drugs. We developed a novel detection assay in order to identify type-1 and 2 CALR mutations by PNA directed PCR clamping. Seventy-five patients affected by myeloproliferative neoplasms and seven controls were examined by direct DNA sequencing and by PNA directed PCR clamping. The assay resulted to be more sensitive, specific and cheaper than sanger sequencing and it could be applied even in laboratory not equipped for more sophisticated analysis. Interestingly, we report here a case carrying both type 1 and type2 mutations in CALR gene.
Collapse
Affiliation(s)
- Valentina Rosso
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Jessica Petiti
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Enrico Bracco
- Department of Oncology, University of Turin, Turin, Italy
| | - Roberto Pedrola
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Francesca Carnuccio
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Elisabetta Signorino
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Sonia Carturan
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Chiara Calabrese
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Giada Bot-Sartor
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Michela Ronconi
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Anna Serra
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Giuseppe Saglio
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Francesco Frassoni
- Department of Pediatric Hemato-Oncology and Stem Cell, Cellular Therapy Laboratory, Institute G. Gaslini, Genova, Italy
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
46
|
Li J, Prins D, Park HJ, Grinfeld J, Gonzalez-Arias C, Loughran S, Dovey OM, Klampfl T, Bennett C, Hamilton TL, Pask DC, Sneade R, Williams M, Aungier J, Ghevaert C, Vassiliou GS, Kent DG, Green AR. Mutant calreticulin knockin mice develop thrombocytosis and myelofibrosis without a stem cell self-renewal advantage. Blood 2018; 131:649-661. [PMID: 29282219 DOI: 10.1182/blood-2017-09-806356] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/15/2017] [Indexed: 02/02/2023] Open
Abstract
Somatic mutations in the endoplasmic reticulum chaperone calreticulin (CALR) are detected in approximately 40% of patients with essential thrombocythemia (ET) and primary myelofibrosis (PMF). Multiple different mutations have been reported, but all result in a +1-bp frameshift and generate a novel protein C terminus. In this study, we generated a conditional mouse knockin model of the most common CALR mutation, a 52-bp deletion. The mutant novel human C-terminal sequence is integrated into the otherwise intact mouse CALR gene and results in mutant CALR expression under the control of the endogenous mouse locus. CALRdel/+ mice develop a transplantable ET-like disease with marked thrombocytosis, which is associated with increased and morphologically abnormal megakaryocytes and increased numbers of phenotypically defined hematopoietic stem cells (HSCs). Homozygous CALRdel/del mice developed extreme thrombocytosis accompanied by features of MF, including leukocytosis, reduced hematocrit, splenomegaly, and increased bone marrow reticulin. CALRdel/+ HSCs were more proliferative in vitro, but neither CALRdel/+ nor CALRdel/del displayed a competitive transplantation advantage in primary or secondary recipient mice. These results demonstrate the consequences of heterozygous and homozygous CALR mutations and provide a powerful model for dissecting the pathogenesis of CALR-mutant ET and PMF.
Collapse
Affiliation(s)
- Juan Li
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Daniel Prins
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Hyun Jung Park
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Jacob Grinfeld
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Carlos Gonzalez-Arias
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Stephen Loughran
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Oliver M Dovey
- Wellcome Trust Sanger Institute, Cambridge, United Kingdom; and
| | - Thorsten Klampfl
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Cavan Bennett
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge, United Kingdom
| | - Tina L Hamilton
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Dean C Pask
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Rachel Sneade
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Matthew Williams
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Juliet Aungier
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Cedric Ghevaert
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge, United Kingdom
| | - George S Vassiliou
- Department of Haematology, Addenbrooke's Hospital, Cambridge, United Kingdom
- Wellcome Trust Sanger Institute, Cambridge, United Kingdom; and
| | - David G Kent
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Anthony R Green
- Cambridge Institute for Medical Research and Wellcome Trust/Medical Research Council Stem Cell Institute and
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, Addenbrooke's Hospital, Cambridge, United Kingdom
| |
Collapse
|
47
|
Iborra FJ, Papadopoulos P. Calreticulin in Essential Thrombocythemia: StressINg OUT the Megakaryocyte Nucleus. Front Oncol 2017; 7:103. [PMID: 28589084 PMCID: PMC5438987 DOI: 10.3389/fonc.2017.00103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 05/02/2017] [Indexed: 12/29/2022] Open
Abstract
Calreticulin (CALR) is a multifaceted protein primarily involved in intracellular protein control processes. The identification of CALR mutations in essential thrombocythemia (ET) and primary myelofibrosis that are mutually exclusive with the JAK2 V617F mutation has stirred an intensive research interest about the molecular functions of CALR and its mutants in myeloproliferative neoplasms (MPNs) and its diagnostic/prognostic value. The recently characterized protein–protein interaction of CALR mutants and MPL receptor has advanced our knowledge on the functional role of CALR mutants in thrombocythemia but it has also uncovered limitations of the current established research models. Human cell lines and mouse models provide useful information but they lack the advantages provided by ex vivo primary cultures of physiologically relevant to the disease cell types [i.e., megakaryocytes (MKs), platelets]. The results from gene expression and chromatin occupancy analysis have focused on the JAK-STAT pathway activated in both JAK2 V617F- and CALR-mutated MPN patient groups, although a more complete analysis is needed to be performed in MKs. Stress related processes seem to be affected in CALR mutant ET-MKs, but the precise mechanism is not known yet. Herein, we describe a culture method for human MKs from peripheral blood progenitors, which could help further toward an unbiased characterization of the role of CALR in ET and MK differentiation.
Collapse
Affiliation(s)
- Francisco Jose Iborra
- Department of Molecular Cell Biology, Centro Nacional de Biotecnologia, Madrid, Spain
| | - Petros Papadopoulos
- Department of Hematology, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| |
Collapse
|
48
|
Somatic mutations of calreticulin in myeloproliferative neoplasms. Int J Hematol 2017; 105:743-747. [PMID: 28470469 DOI: 10.1007/s12185-017-2246-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 11/27/2022]
Abstract
Recurrent somatic mutations in calreticulin (CALR) gene that encodes a molecular chaperone residing in the endoplasmic reticulum were identified in 2013 in a subset of patients with myeloproliferative neoplasms (MPNs). All of these mutations found in patients were either small insertion or deletion in a narrow region on exon 9 of CALR gene, and caused +1 frameshift in the reading frame for the translation of the carboxyl-terminus of CALR. Because of this unique feature, the CALR mutation is believed to be a gain-of-function mutation. However, there was essentially no rationale model to implicate the involvement of mutant CALR in the pathogenesis of MPN or other malignancies. Based on the recent findings, this review summarizes a novel molecular mechanism by which this mutant molecular chaperone constitutively activates the cytokine receptor to induce cellular transformation in MPNs.
Collapse
|