201
|
Zhang Z, Yang W, Zhu T, Wang L, Zhao X, Zhao G, Qu L, Jia Y. Genetic Parameter Estimation and Whole Sequencing Analysis of the Genetic Architecture of Chicken Keel Bending. Front Genet 2022; 13:833132. [PMID: 35401685 PMCID: PMC8984200 DOI: 10.3389/fgene.2022.833132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/24/2022] [Indexed: 11/18/2022] Open
Abstract
Bone health is particularly important for high-yielding commercial layer chickens. The keel of poultry is an extension of the abdomen side of the sternum along the sagittal plane and is one of the most important bones. In this study, the keel phenotype of White Leghorns laying hen flocks showed significant individual differences. To clarify its genetic mechanism, we first estimated the heritability of keel bend (KB) in White Leghorn, recorded the production performance of the chicken flock, examined the blood biochemical indexes and bone quality in KB and keel normal (KN) chickens, and performed whole-genome pooled sequencing in KB and KN chickens. We then performed selection elimination analysis to determine the genomic regions that may affect the keel phenotypes. The results show that KB is a medium heritability trait. We found that cage height had a significant effect on the KB (p < 0.01). At 48 weeks, there were significant differences in the number of eggs, the number of normal eggs, and eggshell strength (p < 0.05). The content of parathyroid hormone was lower (p < 0.01) and that of calcitonin was higher (p < 0.01) in KB chickens than in KN chickens. The differences in bone mineral density, bone strength, and bone cortical thickness of the humerus and femur were extremely significant (p < 0.01), with all being lower in KB chickens than in KN chickens. In addition, the bones of KB chickens contained more fat organization. A total of 128 genes were identified in selective sweep regions. We identified 10 important candidate genes: ACP5, WNT1, NFIX, CNN1, CALR, FKBP11, TRAPPC5, MAP2K7, RELA, and ENSGALG00000047166. Among the significantly enriched Kyoto Encyclopedia of Genes and Genomes pathways found, we identifed two bone-related pathways, one involving “osteoclast differentiation” and the other the “MAPK signaling pathway.” These results may help us better understand the molecular mechanism of bone traits in chickens and other birds and provide new insights for the genetic breeding of chickens.
Collapse
Affiliation(s)
- Zhihao Zhang
- Institute of Animal Sciences, Chinese Academy of Agricultural Science, Beijing, China
| | - Weifang Yang
- Beijing General Station of Animal Husbandry, Beijing, China
| | - Tao Zhu
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Liang Wang
- Beijing General Station of Animal Husbandry, Beijing, China
| | - Xiaoyu Zhao
- Hebei Dawu Poultry Breeding Co., Ltd., Hebei, China
| | | | - Lujiang Qu
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- *Correspondence: Lujiang Qu, ; Yaxiong Jia,
| | - Yaxiong Jia
- Institute of Animal Sciences, Chinese Academy of Agricultural Science, Beijing, China
- *Correspondence: Lujiang Qu, ; Yaxiong Jia,
| |
Collapse
|
202
|
Bhuria V, Baldauf CK, Schraven B, Fischer T. Thromboinflammation in Myeloproliferative Neoplasms (MPN)-A Puzzle Still to Be Solved. Int J Mol Sci 2022; 23:ijms23063206. [PMID: 35328626 PMCID: PMC8954909 DOI: 10.3390/ijms23063206] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs), a group of malignant hematological disorders, occur as a consequence of somatic mutations in the hematopoietic stem cell compartment and show excessive accumulation of mature myeloid cells in the blood. A major cause of morbidity and mortality in these patients is the marked prothrombotic state leading to venous and arterial thrombosis, including myocardial infarction (MI), deep vein thrombosis (DVT), and strokes. Additionally, many MPN patients suffer from inflammation-mediated constitutional symptoms, such as fever, night sweats, fatigue, and cachexia. The chronic inflammatory syndrome in MPNs is associated with the up-regulation of various inflammatory cytokines in patients and is involved in the formation of the so-called MPN thromboinflammation. JAK2-V617F, the most prevalent mutation in MPNs, has been shown to activate a number of integrins on mature myeloid cells, including granulocytes and erythrocytes, which increase adhesion and drive venous thrombosis in murine knock-in/out models. This review aims to shed light on the current understanding of thromboinflammation, involvement of neutrophils in the prothrombotic state, plausible molecular mechanisms triggering the process of thrombosis, and potential novel therapeutic targets for developing effective strategies to reduce the MPN disease burden.
Collapse
Affiliation(s)
- Vikas Bhuria
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention—ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Conny K. Baldauf
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention—ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Correspondence: ; Tel.: +49-391-67-15338; Fax: +49-391-67-15852
| | - Thomas Fischer
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention—ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
203
|
Bhuria V, Baldauf CK, Schraven B, Fischer T. Thromboinflammation in Myeloproliferative Neoplasms (MPN)-A Puzzle Still to Be Solved. Int J Mol Sci 2022. [PMID: 35328626 DOI: 10.3390/ijms23063206.pmid:35328626;pmcid:pmc8954909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs), a group of malignant hematological disorders, occur as a consequence of somatic mutations in the hematopoietic stem cell compartment and show excessive accumulation of mature myeloid cells in the blood. A major cause of morbidity and mortality in these patients is the marked prothrombotic state leading to venous and arterial thrombosis, including myocardial infarction (MI), deep vein thrombosis (DVT), and strokes. Additionally, many MPN patients suffer from inflammation-mediated constitutional symptoms, such as fever, night sweats, fatigue, and cachexia. The chronic inflammatory syndrome in MPNs is associated with the up-regulation of various inflammatory cytokines in patients and is involved in the formation of the so-called MPN thromboinflammation. JAK2-V617F, the most prevalent mutation in MPNs, has been shown to activate a number of integrins on mature myeloid cells, including granulocytes and erythrocytes, which increase adhesion and drive venous thrombosis in murine knock-in/out models. This review aims to shed light on the current understanding of thromboinflammation, involvement of neutrophils in the prothrombotic state, plausible molecular mechanisms triggering the process of thrombosis, and potential novel therapeutic targets for developing effective strategies to reduce the MPN disease burden.
Collapse
Affiliation(s)
- Vikas Bhuria
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention-ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Conny K Baldauf
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention-ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Thomas Fischer
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention-ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
204
|
Nurgat Z, Lawrence M. Management of Myeloproliferative Neoplasms (MPNs). J Oncol Pharm Pract 2022; 28:1400-1410. [PMID: 35296179 DOI: 10.1177/10781552221082293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To provide up to date guidance, practice recommendations and highlight barriers to medication adherence in the long-term management of chronic myeloproliferative neoplasms (MPNs). AIM Current drug therapy for MPN is not curative and has not been shown to prolong survival. The main indication for treatment is the prevention of thrombosis and medication adherence remains a challenge in this group of patients. Identifying potentially modifiable barriers to medication adherence including primary nonadherence and non-persistent adherence enables timely interventions to be put in place and improve overall medication adherence. METHODS A systematic review of peer-reviewed literature and expert opinions was performed using electronic databases (PubMed, EMBASE, MEDLINE, and Web of Science) that were searched for articles reporting MPN and medication adherence. Discussions A case vignette is discussed throughout the article and expert opinion with international peer reviewed guidelines that are authored to support clinical decision making at the point of care were utilised. The evidence base was combined with more practical/clinical (data based) insight from real world clinical practice. Adoption of a broad range of digital health care activities and services in the health care system (telehealth applications) by the advanced practice providers (Non-Medical Prescribers-NMPs) in MPN clinics included medication prescribing and management, oral drug compliance and adherence evaluations, interventions, chronic care management, counselling and patient education on treatments. CONCLUSION Current drug therapy for MPN is neither curative nor has it been shown to prolong survival, and medication adherence remains a challenge in this group of patients. The longevity of the patients' disease course may contribute to the high risk of non-adherence in this patient cohort. Poor adherence to long-term therapies severely compromises the effectiveness of treatment. Adherence interventions should be tailored to the needs of the patient in order to achieve maximum impact. Interventions aimed at improving adherence provide the best experience and outcome for the patient and their families and can have a profound impact on the quality of life and mitigation of disease consequences.
Collapse
Affiliation(s)
- Zubeir Nurgat
- Lead Pharmacist, Haematology / Oncology & Clinical Trials, Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent, UK
| | - Myer Lawrence
- Lead Nurse Acute Oncology Service, East Suffolk and North Essex NHS Foundation Trust, Colchester General & Ipswich Hospital, England, UK
| |
Collapse
|
205
|
Hodeib H, Abd EL Hai D, Tawfik MA, Allam AA, Selim A, Elsawy AA, Youssef A. CCL2 rs1024611Gene Polymorphism in Philadelphia-Negative Myeloproliferative Neoplasms. Genes (Basel) 2022; 13:genes13030492. [PMID: 35328046 PMCID: PMC8948730 DOI: 10.3390/genes13030492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/20/2022] [Accepted: 03/08/2022] [Indexed: 02/05/2023] Open
Abstract
Introduction: The onset of the Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs) is caused by acquired somatic mutations in target myeloid genes “driver mutations”. The CCL2 gene is overexpressed by non-Hodgkin lymphomas and multiple solid tumors. Aim of the study: to evaluate the possible association of CCL2 rs1024611 SNP and its expression level and the risk of developing Philadelphia-negative MPNs. Patients and methods: A total of 128 newly diagnosed Philadelphia-negative MPN patient and 141 healthy subjects were evaluated for the genotype distribution of CCL2 rs1024611 and CCL2 expression levels. Results: The CCL2 rs1024611 G/G genotype was more frequent and significantly frequent among PMF and Post-PV/ET-MF patients and the mean CCL2 expression levels were significantly higher in PMF and Post-PV/ET-MF compared to the healthy subjects. The CCL2 rs1024611 SNP was significantly correlated to the CCL2 gene expression level and fibrosis grade. ROC analysis for the CCL2 gene expression level that discriminates MF patients from PV + ET patients revealed a sensitivity of 80.43% and a specificity of 73.17% with an AUC of 0.919 (p < 0.001). Conclusion: The CCL2 rs1024611 polymorphism could be an independent risk factor for developing MF (PMF and Post-PV/ET-MF). Moreover, CCL2 gene expression could be potential genetic biomarker of fibrotic progression.
Collapse
Affiliation(s)
- Hossam Hodeib
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (H.H.); (D.A.E.H.); (A.Y.)
| | - Dina Abd EL Hai
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (H.H.); (D.A.E.H.); (A.Y.)
| | - Mohamed A Tawfik
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (A.A.A.); (A.S.); (A.A.E.)
- Correspondence: ; Tel.: +20-1550844075
| | - Alzahraa A. Allam
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (A.A.A.); (A.S.); (A.A.E.)
| | - Amal Selim
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (A.A.A.); (A.S.); (A.A.E.)
| | - Abdallah Ahmed Elsawy
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (A.A.A.); (A.S.); (A.A.E.)
| | - Amira Youssef
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (H.H.); (D.A.E.H.); (A.Y.)
| |
Collapse
|
206
|
Dutta A, Nath D, Yang Y, Le BT, Rahman MFU, Faughnan P, Wang Z, Stuver M, He R, Tan W, Hutchison RE, Foulks JM, Warner SL, Zang C, Mohi G. Genetic ablation of Pim1 or pharmacologic inhibition with TP-3654 ameliorates myelofibrosis in murine models. Leukemia 2022; 36:746-759. [PMID: 34741118 PMCID: PMC8891046 DOI: 10.1038/s41375-021-01464-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/14/2022]
Abstract
Myelofibrosis (MF) is the deadliest form of myeloproliferative neoplasm (MPN). The JAK inhibitor Ruxolitinib can reduce constitutional symptoms but it does not substantially improve bone marrow fibrosis. Pim1 expression is significantly elevated in MPN/MF hematopoietic progenitors. Here, we show that genetic ablation of Pim1 blocked the development of myelofibrosis induced by Jak2V617F and MPLW515L. Pharmacologic inhibition of Pim1 with a second-generation Pim kinase inhibitor TP-3654 significantly reduced leukocytosis and splenomegaly, and attenuated bone marrow fibrosis in Jak2V617F and MPLW515L mouse models of MF. Combined treatment of TP-3654 and Ruxolitinib resulted in greater reduction of spleen size, normalization of blood leukocyte counts and abrogation of bone marrow fibrosis in murine models of MF. TP-3654 treatment also preferentially inhibited Jak2V617F mutant hematopoietic progenitors in mice. Mechanistically, we show that TP-3654 treatment significantly inhibits mTORC1, MYC and TGF-β signaling in Jak2V617F mutant hematopoietic cells and diminishes the expression of fibrotic markers in the bone marrow. Collectively, our results suggest that Pim1 plays an important role in the pathogenesis of MF, and inhibition of Pim1 with TP-3654 might be useful for treatment of MF.
Collapse
Affiliation(s)
- Avik Dutta
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Dipmoy Nath
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Yue Yang
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Bao T Le
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Mohammad Ferdous-Ur Rahman
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Patrick Faughnan
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Zhenjia Wang
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Matthew Stuver
- Department of Pharmacology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, USA
| | - Rongquan He
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Wuwei Tan
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Robert E Hutchison
- Department of Pathology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, USA
| | - Jason M Foulks
- Sumitomo Dainippon Pharma Oncology, Inc (formerly Tolero Pharmaceuticals, Inc), Lehi, UT, USA
| | - Steven L Warner
- Sumitomo Dainippon Pharma Oncology, Inc (formerly Tolero Pharmaceuticals, Inc), Lehi, UT, USA
| | - Chongzhi Zang
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA, USA
- University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Golam Mohi
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA.
- University of Virginia Cancer Center, Charlottesville, VA, USA.
| |
Collapse
|
207
|
Tefferi A, Gangat N, Pardanani A, Crispino JD. Myelofibrosis: Genetic Characteristics and the Emerging Therapeutic Landscape. Cancer Res 2022; 82:749-763. [PMID: 34911786 PMCID: PMC9306313 DOI: 10.1158/0008-5472.can-21-2930] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/18/2021] [Accepted: 11/30/2021] [Indexed: 01/07/2023]
Abstract
Primary myelofibrosis (PMF) is one of three myeloproliferative neoplasms (MPN) that are morphologically and molecularly inter-related, the other two being polycythemia vera (PV) and essential thrombocythemia (ET). MPNs are characterized by JAK-STAT-activating JAK2, CALR, or MPL mutations that give rise to stem cell-derived clonal myeloproliferation, which is prone to leukemic and, in case of PV and ET, fibrotic transformation. Abnormal megakaryocyte proliferation is accompanied by bone marrow fibrosis and characterizes PMF, while the clinical phenotype is pathogenetically linked to ineffective hematopoiesis and aberrant cytokine expression. Among MPN-associated driver mutations, type 1-like CALR mutation has been associated with favorable prognosis in PMF, while ASXL1, SRSF2, U2AF1-Q157, EZH2, CBL, and K/NRAS mutations have been shown to be prognostically detrimental. Such information has enabled development of exclusively genetic (GIPSS) and clinically integrated (MIPSSv2) prognostic models that facilitate individualized treatment decisions. Allogeneic stem cell transplantation remains the only treatment modality in MF with the potential to prolong survival, whereas drug therapy, including JAK2 inhibitors, is directed mostly at the inflammatory component of the disease and is therefore palliative in nature. Similarly, disease-modifying activity remains elusive for currently available investigational drugs, while their additional value in symptom management awaits controlled confirmation. There is a need for genetic characterization of clinical observations followed by in vitro and in vivo preclinical studies that will hopefully identify therapies that target the malignant clone in MF to improve patient outcomes.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota.,Corresponding Author: Ayalew Tefferi, Division of Hematology, Department of Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905. Phone: 507-284-2511; Fax: 507-266-4972; E-mail:
| | - Naseema Gangat
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Animesh Pardanani
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - John D. Crispino
- Division of Experimental Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
208
|
Thomas SJ, Dash DP. A Rare Co-Occurrence of Triple Mutations in JAK2, CALR, and MPL in the Same Patient with Myelofibrosis. Case Rep Hematol 2022; 2022:4579122. [PMID: 35237453 PMCID: PMC8885281 DOI: 10.1155/2022/4579122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 11/08/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022] Open
Abstract
Background. The diagnosis and prognostication of myeloproliferative neoplasm rely on the presence of driver mutations in JAK2, calreticulin (CALR), and MPL mutations. In the past, the presence of these mutations was thought to be mutually exclusive. Since then, there have been multiple reports of the presence of dual mutations. The presence of all three driver mutations in the same patient with myelofibrosis has not been previously described. CASE A 73-year-old female underwent a hematological workup in our facility after a routine hemogram performed prior to complex ophthalmological surgery revealed severe thrombocytosis. A comprehensive workup including an NGS panel for MPN driver mutations demonstrated that she had a calreticulin type-1 mutation, a JAK2 exon 14 (JAK2L611S) mutation, and an abnormal hotspot variant for MPL with VAF1%. A bone marrow biopsy confirmed a myeloproliferative neoplasm with grade 2 reticulin fibrosis suggesting primary myelofibrosis. Molecular profiling of bone marrow confirmed the previously noted mutations and an MPLW515R mutation. The patient was started on treatment with hydroxyurea and aspirin with improvement in platelet count and resolution of anemia. DISCUSSION The clinical significance of the presence of multiple driver mutations in the same patient is not well understood at this time. There have been 11 publications between 2014 and 2020 that have described dual mutations of JAK2V617F, MPL, and CALR mutations. The JAK2 exon 14 mutation noted, in this case, is JAK2L611S which has not previously been reported in MPN and only reported in 5 cases in the COSMIC database. The JAK2 exon 14 mutation identified in this case is not an established driver mutation for myeloproliferative neoplasm, and its clinical implication remains unknown. CONCLUSIONS The above case in addition to recent case reports and case series supports the use of broader NGS sequencing panels for diagnosis and prognostication of MPN. These mutations should not be considered mutually exclusive. The clinical behavior and prognosis of the subgroup with multiple mutations need to be studied in larger series.
Collapse
Affiliation(s)
- Sherine J. Thomas
- Hematology and Oncology, Northside Hospital Cancer Institute, Georgia Cancer Specialists, Atlanta, GA, USA
| | - D. P. Dash
- Molecular Oncology & Genetics (MOGL), Versiti Wisconsin (Formerly Known as Blood Center Wisconsin), Milwaukee, WI, USA
| |
Collapse
|
209
|
Bochicchio MT, Di Battista V, Poggio P, Carrà G, Morotti A, Brancaccio M, Lucchesi A. Understanding Aberrant Signaling to Elude Therapy Escape Mechanisms in Myeloproliferative Neoplasms. Cancers (Basel) 2022; 14:cancers14040972. [PMID: 35205715 PMCID: PMC8870427 DOI: 10.3390/cancers14040972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/01/2023] Open
Abstract
Aberrant signaling in myeloproliferative neoplasms may arise from alterations in genes coding for signal transduction proteins or epigenetic regulators. Both mutated and normal cells cooperate, altering fragile balances in bone marrow niches and fueling persistent inflammation through paracrine or systemic signals. Despite the hopes placed in targeted therapies, myeloid proliferative neoplasms remain incurable diseases in patients not eligible for stem cell transplantation. Due to the emergence of drug resistance, patient management is often very difficult in the long term. Unexpected connections among signal transduction pathways highlighted in neoplastic cells suggest new strategies to overcome neoplastic cell adaptation.
Collapse
Affiliation(s)
- Maria Teresa Bochicchio
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Valeria Di Battista
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Pietro Poggio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Giovanna Carrà
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy;
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| | - Alessandro Lucchesi
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| |
Collapse
|
210
|
Saito N, Yamauchi T, Kawano N, Ono R, Yoshida S, Miyamoto T, Kamimura T, Shultz LD, Saito Y, Takenaka K, Shimoda K, Harada M, Akashi K, Ishikawa F. Circulating CD34+ cells of primary myelofibrosis patients contribute to myeloid-dominant hematopoiesis and bone marrow fibrosis in immunodeficient mice. Int J Hematol 2022; 115:198-207. [PMID: 34773575 PMCID: PMC8905546 DOI: 10.1007/s12185-021-03239-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 10/13/2021] [Accepted: 10/17/2021] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Primary myelofibrosis (PMF) is a clonal stem cell disorder characterized by myeloid dominant hematopoiesis and dysregulated proliferation of fibroblasts in the bone marrow. However, how these aberrant myeloid cells and fibroblasts are produced remains unclear. AIM AND METHODS In this study, we examined in vivo engraftment kinetics of PMF patient-derived CD34+ cells in immunecompromised NOD/SCID/IL2rgKO (NSG) mice. Engrafted human cells were analyzed with flow cytometry, and proliferation of fibroblastic cells and bone marrow fibrosis were assessed with the histo-pathological examination. RESULTS Transplantation of PMF patient-derived circulating CD34+ fractions into NSG newborns recapitulates clinical features of human PMF. Engraftment of human CD45+ leukocytes resulted in anemia and myeloid hyperplasia accompanied by bone marrow fibrosis by six months post-transplantation. Fibrotic bone marrow contained CD45-vimentin+ cells of both human and mouse origin, suggesting that circulating malignant CD34+ subsets contribute to myelofibrotic changes in PMF through direct and indirect mechanisms. CONCLUSION A patient-derived xenotransplantation (PDX) model of PMF allows in vivo examination of disease onset and propagation originating from immature CD34+ cells and will support the investigation of pathogenesis and development of therapeutic modalities for the disorder.
Collapse
Affiliation(s)
- Noriyuki Saito
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medicine, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Department of Hematology, Saiseikai Fukuoka General Hospital, 1-3-46 Tenjin, Chuo-ku, Fukuoka, 810-0001, Japan
| | - Takuji Yamauchi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medicine, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Noriaki Kawano
- Department of Internal Medicine, Miyazaki Prefectural Miyazaki Hospital, Miyazaki, Japan
| | - Rintaro Ono
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shuro Yoshida
- Department of Hematology, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Toshihiro Miyamoto
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medicine, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | | | | | - Yoriko Saito
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Katsuto Takenaka
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Kazuya Shimoda
- Division of Hematology, Diabetes, and Endocrinology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Mine Harada
- Karatsu Higashimatsuura Medical Center, Karatsu, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medicine, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Fumihiko Ishikawa
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
| |
Collapse
|
211
|
Williams N, Lee J, Mitchell E, Moore L, Baxter EJ, Hewinson J, Dawson KJ, Menzies A, Godfrey AL, Green AR, Campbell PJ, Nangalia J. Life histories of myeloproliferative neoplasms inferred from phylogenies. Nature 2022; 602:162-168. [PMID: 35058638 DOI: 10.1038/s41586-021-04312-6] [Citation(s) in RCA: 182] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/06/2021] [Indexed: 12/16/2022]
Abstract
Mutations in cancer-associated genes drive tumour outgrowth, but our knowledge of the timing of driver mutations and subsequent clonal dynamics is limited1-3. Here, using whole-genome sequencing of 1,013 clonal haematopoietic colonies from 12 patients with myeloproliferative neoplasms, we identified 580,133 somatic mutations to reconstruct haematopoietic phylogenies and determine clonal histories. Driver mutations were estimated to occur early in life, including the in utero period. JAK2V617F was estimated to have been acquired by 33 weeks of gestation to 10.8 years of age in 5 patients in whom JAK2V617F was the first event. DNMT3A mutations were acquired by 8 weeks of gestation to 7.6 years of age in 4 patients, and a PPM1D mutation was acquired by 5.8 years of age. Additional genomic events occurred before or following JAK2V617F acquisition and as independent clonal expansions. Sequential driver mutation acquisition was separated by decades across life, often outcompeting ancestral clones. The mean latency between JAK2V617F acquisition and diagnosis was 30 years (range 11-54 years). Estimated historical rates of clonal expansion varied substantially (3% to 190% per year), increased with additional driver mutations, and predicted latency to diagnosis. Our study suggests that early driver mutation acquisition and life-long growth and evolution underlie adult myeloproliferative neoplasms, raising opportunities for earlier intervention and a new model for cancer development.
Collapse
Affiliation(s)
| | - Joe Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Emily Mitchell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Luiza Moore
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - E Joanna Baxter
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - James Hewinson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Kevin J Dawson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Andrew Menzies
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Anna L Godfrey
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Anthony R Green
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Peter J Campbell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Jyoti Nangalia
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| |
Collapse
|
212
|
Bader MS, Meyer SC. JAK2 in Myeloproliferative Neoplasms: Still a Protagonist. Pharmaceuticals (Basel) 2022; 15:ph15020160. [PMID: 35215273 PMCID: PMC8874480 DOI: 10.3390/ph15020160] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
The discovery of the activating V617F mutation in Janus kinase 2 (JAK2) has been decisive for the understanding of myeloproliferative neoplasms (MPN). Activated JAK2 signaling by JAK2, CALR, and MPL mutations has become a focus for the development of targeted therapies for patients with MPN. JAK2 inhibitors now represent a standard of clinical care for certain forms of MPN and offer important benefits for MPN patients. However, several key aspects remain unsolved regarding the targeted therapy of MPN with JAK2 inhibitors, such as reducing the MPN clone and how to avoid or overcome a loss of response. Here, we summarize the current knowledge on the structure and signaling of JAK2 as central elements of MPN pathogenesis and feature benefits and limitations of therapeutic JAK2 targeting in MPN.
Collapse
Affiliation(s)
| | - Sara Christina Meyer
- Division of Hematology, University Hospital Basel, CH-4031 Basel, Switzerland;
- Department of Biomedicine, University Hospital Basel and University of Basel, CH-4031 Basel, Switzerland
- Correspondence:
| |
Collapse
|
213
|
Majeti R, Jamieson C, Pang WW, Jaiswal S, Leeper NJ, Wernig G, Weissman IL. Clonal Expansion of Stem/Progenitor Cells in Cancer, Fibrotic Diseases, and Atherosclerosis, and CD47 Protection of Pathogenic Cells. Annu Rev Med 2022; 73:307-320. [PMID: 35084991 DOI: 10.1146/annurev-med-042420-104436] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We proposed and demonstrated that myelogenous leukemia has a preleukemic phase. In the premalignant phase, normal hematopoietic stem cells (HSCs) gradually accumulate mutations leading to HSC clonal expansion, resulting in the emergence of leukemic stem cells (LSCs). Here, we show that preleukemic HSCs are the basis of clonal hematopoiesis, as well as late-onset blood diseases (chronic-phase chronic myeloid leukemia, myeloproliferative neoplasms, and myelodysplastic disease). The clones at some point each trigger surface expression of "eat me" signals for macrophages, and in the clones and their LSC progeny, this is countered by upregulation of "don't eat me" signals for macrophages such as CD47,opening the possibility of CD47-based therapies. We include evidence that similar processes result in fibroblast expansion in a variety of fibrotic diseases, and arterial smooth muscle clonal expansion is a basis of atherosclerosis, including upregulation of both "eat me" and "don't eat me" molecules on the pathogenic cells.
Collapse
Affiliation(s)
- R Majeti
- Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University Medical Center, Stanford, California 94305, USA;
| | - C Jamieson
- Sanford Stem Cell Clinical Center, University of California, San Diego, La Jolla, California 92093, USA
| | - W W Pang
- Jasper Therapeutics, Redwood City, California 94065, USA
| | - S Jaiswal
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - N J Leeper
- Department of Surgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | - G Wernig
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University Medical Center, Stanford, California 94305, USA;
| | - I L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University Medical Center, Stanford, California 94305, USA;
| |
Collapse
|
214
|
Marković D, Maslovarić I, Djikić D, Čokić VP. Neutrophil Death in Myeloproliferative Neoplasms: Shedding More Light on Neutrophils as a Pathogenic Link to Chronic Inflammation. Int J Mol Sci 2022; 23:1490. [PMID: 35163413 PMCID: PMC8836089 DOI: 10.3390/ijms23031490] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 12/15/2022] Open
Abstract
Neutrophils are an essential component of the innate immune response, but their prolonged activation can lead to chronic inflammation. Consequently, neutrophil homeostasis is tightly regulated through balance between granulopoiesis and clearance of dying cells. The bone marrow is both a site of neutrophil production and the place they return to and die. Myeloproliferative neoplasms (MPN) are clonal hematopoietic disorders characterized by the mutations in three types of molecular markers, with emphasis on Janus kinase 2 gene mutation (JAK2V617F). The MPN bone marrow stem cell niche is a site of chronic inflammation, with commonly increased cells of myeloid lineage, including neutrophils. The MPN neutrophils are characterized by the upregulation of JAK target genes. Additionally, MPN neutrophils display malignant nature, they are in a state of activation, and with deregulated apoptotic machinery. In other words, neutrophils deserve to be placed in the midst of major events in MPN. Our crucial interest in this review is better understanding of how neutrophils die in MPN mirrored by defects in apoptosis and to what possible extent they can contribute to MPN pathophysiology. We tend to expect that reduced neutrophil apoptosis will establish a pathogenic link to chronic inflammation in MPN.
Collapse
Affiliation(s)
- Dragana Marković
- Group for Immunology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, POB 39, 11129 Belgrade, Serbia;
| | - Irina Maslovarić
- Group for Immunology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, POB 39, 11129 Belgrade, Serbia;
| | - Dragoslava Djikić
- Group for Molecular Oncology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, POB 39, 11129 Belgrade, Serbia; (D.D.); (V.P.Č.)
| | - Vladan P. Čokić
- Group for Molecular Oncology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, POB 39, 11129 Belgrade, Serbia; (D.D.); (V.P.Č.)
| |
Collapse
|
215
|
Baba T, Hashimoto Y, Yasuda H, Araki M, Edahiro Y, Morishita S, Ochiai T, Shirane S, Ando J, Komatsu N. Validation and reliability of current guidelines for the treatment of essential thrombocythemia under real-world clinical settings in Japan. Hematology 2022; 27:157-166. [DOI: 10.1080/16078454.2021.2022848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Terumi Baba
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshinori Hashimoto
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Laboratory for the Development of Therapies Against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hajime Yasuda
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Marito Araki
- Laboratory for the Development of Therapies Against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Transfusion Medicine and Stem Cell Regulation, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoko Edahiro
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Laboratory for the Development of Therapies Against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Soji Morishita
- Laboratory for the Development of Therapies Against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomonori Ochiai
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Laboratory for the Development of Therapies Against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shuichi Shirane
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Laboratory for the Development of Therapies Against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Jun Ando
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Transfusion Medicine and Stem Cell Regulation, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Norio Komatsu
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Laboratory for the Development of Therapies Against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
216
|
Battukh N, Ali E, Yassin M. Osteolytic bone lesions in patients with primary myelofibrosis: A systematic review. ACTA BIO-MEDICA : ATENEI PARMENSIS 2022; 92:e2021478. [PMID: 35075062 PMCID: PMC8823556 DOI: 10.23750/abm.v92i6.12350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 01/17/2023]
Abstract
Philadelphia negative myeloproliferative neoplasms (MPNs) are classically characterized by excess production of terminal myeloid cells in the peripheral blood. They include polycythemia vera, essential thrombocythemia, and primary myelofibrosis. Among this group, primary myelofibrosis is the least common and usually carries the worst prognosis. Bone involvement in primary myelofibrosis has many forms; it affects bone marrow leading to bone marrow fibrosis, it can cause periostitis, in addition to bone and joint pain. A common radiologic finding in primary myelofibrosis is the presence of osteosclerotic lesions. However, the presence of osteolytic lesions in bone imaging was described in few reports. In this review, we searched English literature using the PRISMA guidelines looking for patients with Primary myelofibrosis who had osteolytic bone lesions to assess the impact of such findings on the disease and its effect on prognosis. We found the vast majority of lesions were painful affecting most commonly the vertebral column, pelvis, and ribs, and were detected in patients above 50 years of age with no gender preference, unfortunately they represented advanced disease stages, resulting in inadequate treatment response and poor outcome.
Collapse
Affiliation(s)
| | | | - Mohamed Yassin
- Medical Oncology, Hematology Section, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
217
|
Mori N, Ohwashi-Miyazaki M, Yoshinaga K, Ogasawara T, Marshall S, Shiseki M, Sakura H, Tanaka J. Genetic alterations in patients with chronic leucocytosis and persistent thrombocytosis. J Genet 2022. [DOI: 10.1007/s12041-021-01354-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
218
|
Tan J, Chow YP, Zainul Abidin N, Chang KM, Selvaratnam V, Tumian NR, Poh YM, Veerakumarasivam A, Laffan MA, Wong CL. Analysis of genetic variants in myeloproliferative neoplasms using a 22-gene next-generation sequencing panel. BMC Med Genomics 2022; 15:10. [PMID: 35033063 PMCID: PMC8760696 DOI: 10.1186/s12920-021-01145-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 12/06/2021] [Indexed: 12/18/2022] Open
Abstract
Background The Philadelphia (Ph)-negative myeloproliferative neoplasms (MPNs), namely essential thrombocythaemia (ET), polycythaemia vera (PV) and primary myelofibrosis (PMF), are a group of chronic clonal haematopoietic disorders that have the propensity to advance into bone marrow failure or acute myeloid leukaemia; often resulting in fatality. Although driver mutations have been identified in these MPNs, subtype-specific markers of the disease have yet to be discovered. Next-generation sequencing (NGS) technology can potentially improve the clinical management of MPNs by allowing for the simultaneous screening of many disease-associated genes. Methods The performance of a custom, in-house designed 22-gene NGS panel was technically validated using reference standards across two independent replicate runs. The panel was subsequently used to screen a total of 10 clinical MPN samples (ET n = 3, PV n = 3, PMF n = 4). The resulting NGS data was then analysed via a bioinformatics pipeline. Results The custom NGS panel had a detection limit of 1% variant allele frequency (VAF). A total of 20 unique variants with VAFs above 5% (4 of which were putatively novel variants with potential biological significance) and one pathogenic variant with a VAF of between 1 and 5% were identified across all of the clinical MPN samples. All single nucleotide variants with VAFs ≥ 15% were confirmed via Sanger sequencing. Conclusions The high fidelity of the NGS analysis and the identification of known and novel variants in this study cohort support its potential clinical utility in the management of MPNs. However, further optimisation is needed to avoid false negatives in regions with low sequencing coverage, especially for the detection of driver mutations in MPL. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-021-01145-0.
Collapse
Affiliation(s)
- Jaymi Tan
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Selangor, Malaysia
| | - Yock Ping Chow
- Clinical Research Centre, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Norziha Zainul Abidin
- Molecular Diagnostics Laboratory, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Kian Meng Chang
- Haematology Unit, Department of Medicine, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | | | - Nor Rafeah Tumian
- Haematology Unit, Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Yang Ming Poh
- School of Data Sciences, Perdana University, Serdang, Selangor, Malaysia
| | - Abhi Veerakumarasivam
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Selangor, Malaysia
| | - Michael Arthur Laffan
- Centre for Haematology, Hammersmith Hospital, London, UK.,Faculty of Medicine, Imperial College London, London, UK
| | - Chieh Lee Wong
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Selangor, Malaysia. .,Clinical Research Centre, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia. .,Molecular Diagnostics Laboratory, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia. .,Haematology Unit, Department of Medicine, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia. .,Centre for Haematology, Hammersmith Hospital, London, UK. .,Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
219
|
Fabris S, Cattaneo D, Salerio S, Bucelli C, Ciceri G, Pasquale R, Todoerti K, Gianelli U, Baldini L, Neri A, Iurlo A. Impact on thrombotic risk of canonical and atypical CALR mutations in essential thrombocythemia. A single-center cohort study. Thromb Res 2022; 210:67-69. [PMID: 35016075 DOI: 10.1016/j.thromres.2021.12.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 01/05/2023]
Affiliation(s)
- Sonia Fabris
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Simone Salerio
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cristina Bucelli
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gabriella Ciceri
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Raffaella Pasquale
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Katia Todoerti
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Umberto Gianelli
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy; Division of Pathology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Baldini
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Antonino Neri
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Alessandra Iurlo
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
220
|
Genovese E, Mirabile M, Rontauroli S, Sartini S, Fantini S, Tavernari L, Maccaferri M, Guglielmelli P, Bianchi E, Parenti S, Carretta C, Mallia S, Castellano S, Colasante C, Balliu M, Bartalucci N, Palmieri R, Ottone T, Mora B, Potenza L, Passamonti F, Voso MT, Luppi M, Vannucchi AM, Tagliafico E, Manfredini R, on behalf of the Mynerva (MYeloid NEoplasms Research Venture AIRC). The Response to Oxidative Damage Correlates with Driver Mutations and Clinical Outcome in Patients with Myelofibrosis. Antioxidants (Basel) 2022; 11:antiox11010113. [PMID: 35052617 PMCID: PMC8772737 DOI: 10.3390/antiox11010113] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/29/2021] [Accepted: 01/01/2022] [Indexed: 12/12/2022] Open
Abstract
Myelofibrosis (MF) is the Philadelphia-negative myeloproliferative neoplasm characterized by the worst prognosis and no response to conventional therapy. Driver mutations in JAK2 and CALR impact on JAK-STAT pathway activation but also on the production of reactive oxygen species (ROS). ROS play a pivotal role in inflammation-induced oxidative damage to cellular components including DNA, therefore leading to greater genomic instability and promoting cell transformation. In order to unveil the role of driver mutations in oxidative stress, we assessed ROS levels in CD34+ hematopoietic stem/progenitor cells of MF patients. Our results demonstrated that ROS production in CD34+ cells from CALR-mutated MF patients is far greater compared with patients harboring JAK2 mutation, and this leads to increased oxidative DNA damage. Moreover, CALR-mutant cells show less superoxide dismutase (SOD) antioxidant activity than JAK2-mutated ones. Here, we show that high plasma levels of total antioxidant capacity (TAC) correlate with detrimental clinical features, such as high levels of lactate dehydrogenase (LDH) and circulating CD34+ cells. Moreover, in JAK2-mutated patients, high plasma level of TAC is also associated with a poor overall survival (OS), and multivariate analysis demonstrated that high TAC classification is an independent prognostic factor allowing the identification of patients with inferior OS in both DIPSS lowest and highest categories. Altogether, our data suggest that a different capability to respond to oxidative stress can be one of the mechanisms underlying disease progression of myelofibrosis.
Collapse
Affiliation(s)
- Elena Genovese
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Margherita Mirabile
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Sebastiano Rontauroli
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Stefano Sartini
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Sebastian Fantini
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Lara Tavernari
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Monica Maccaferri
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy;
| | - Paola Guglielmelli
- Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, University of Florence, Careggi University Hospital, 50134 Florence, Italy; (P.G.); (M.B.); (N.B.); (A.M.V.)
| | - Elisa Bianchi
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Sandra Parenti
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Chiara Carretta
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Selene Mallia
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Sara Castellano
- Center for Genome Research, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.C.); (E.T.)
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Corrado Colasante
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
| | - Manjola Balliu
- Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, University of Florence, Careggi University Hospital, 50134 Florence, Italy; (P.G.); (M.B.); (N.B.); (A.M.V.)
| | - Niccolò Bartalucci
- Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, University of Florence, Careggi University Hospital, 50134 Florence, Italy; (P.G.); (M.B.); (N.B.); (A.M.V.)
| | - Raffaele Palmieri
- Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy; (R.P.); (T.O.); (M.T.V.)
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy; (R.P.); (T.O.); (M.T.V.)
- Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, 00179 Rome, Italy
| | - Barbara Mora
- Division of Hematology, Ospedale ASST Sette Laghi, University of Insubria, 21110 Varese, Italy; (B.M.); (F.P.)
| | - Leonardo Potenza
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
| | - Francesco Passamonti
- Division of Hematology, Ospedale ASST Sette Laghi, University of Insubria, 21110 Varese, Italy; (B.M.); (F.P.)
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy; (R.P.); (T.O.); (M.T.V.)
- Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, 00179 Rome, Italy
| | - Mario Luppi
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
| | - Alessandro Maria Vannucchi
- Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, University of Florence, Careggi University Hospital, 50134 Florence, Italy; (P.G.); (M.B.); (N.B.); (A.M.V.)
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.C.); (E.T.)
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
| | - Rossella Manfredini
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
- Correspondence:
| | | |
Collapse
|
221
|
Germline ATG2B/GSKIP-containing 14q32 duplication predisposes to early clonal hematopoiesis leading to myeloid neoplasms. Leukemia 2022; 36:126-137. [PMID: 34172895 DOI: 10.1038/s41375-021-01319-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/26/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023]
Abstract
The germline predisposition associated with the autosomal dominant inheritance of the 14q32 duplication implicating ATG2B/GSKIP genes is characterized by a wide clinical spectrum of myeloid neoplasms. We analyzed 12 asymptomatic carriers and 52 patients aged 18-74 years from six families, by targeted sequencing of 41 genes commonly mutated in myeloid malignancies. We found that 75% of healthy carriers displayed early clonal hematopoiesis mainly driven by TET2 mutations. Molecular landscapes of patients revealed two distinct routes of clonal expansion and leukemogenesis. The first route is characterized by the clonal dominance of myeloproliferative neoplasms (MPN)-driver events associated with TET2 mutations in half of cases and mutations affecting splicing and/or the RAS pathway in one-third of cases, leading to the early development of MPN, mostly essential thrombocythemia, with a high risk of transformation (50% after 10 years). The second route is distinguished by the absence of MPN-driver mutations and leads to AML without prior MPN. These patients mostly harbored a genomic landscape specific to acute myeloid leukemia secondary to myelodysplastic syndrome. An unexpected result was the total absence of DNMT3A mutations in this cohort. Our results suggest that the germline duplication constitutively mimics hematopoiesis aging by favoring TET2 clonal hematopoiesis.
Collapse
|
222
|
Bartalucci N, Galluzzi L. Philadelphia-negative myeloproliferative neoplasms: From origins to new perspectives. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 366:ix-xx. [PMID: 35153008 DOI: 10.1016/s1937-6448(22)00019-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Affiliation(s)
- Niccolò Bartalucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; DENOThe Excellence Center, University of Florence, Florence, Italy.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States; Department of Dermatology, Yale School of Medicine, New Haven, CT, United States; Université de Paris, Paris, France.
| |
Collapse
|
223
|
Edahiro Y, Kurokawa Y, Morishita S, Yamamoto T, Araki M, Komatsu N. Causes of Thrombocytosis: A Single-center Retrospective Study of 1,202 Patients. Intern Med 2022; 61:3323-3328. [PMID: 36385045 PMCID: PMC9751737 DOI: 10.2169/internalmedicine.9282-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective Thrombocytosis can occur as a primary event accompanying hematological diseases or as a secondary event. Since the publication of the World Health Organization classification in 2008, thrombocytosis is now generally defined as a platelet count above 450×109/L. Furthermore, the discovery of driver-gene mutations in myeloproliferative neoplasms (MPNs) has simplified the diagnostic approach for thrombocytosis. To identify the causes of thrombocytosis using this new definition, we conducted a retrospective study. Methods We identified outpatients and inpatients aged 20 years or older with platelet counts >450×109/L in a half-year period at a single institute and analyzed the causes of thrombocytosis and associated clinical characteristics. Results Among 1,202 patients with thrombocytosis, 150 (12.5%) had primary and 999 (83.1%) had secondary thrombocytosis. Of these patients with primary thrombocytosis, 129 (86%) had at least 1 molecular marker indicative of MPNs. The major causes of secondary thrombocytosis were tissue injury (32.2%), infection (17.1%), chronic inflammatory disorders (11.7%) and iron deficiency anemia (11.1%). The median platelet count and the incidence of thrombosis were significantly higher in patients with primary thrombocytosis than in those with secondary thrombocytosis. Conclusion Thrombocytosis mainly occurs as a secondary event; however, it is important to determine the cause of and prevent thrombosis, particularly in cases of primary thrombocytosis.
Collapse
Affiliation(s)
- Yoko Edahiro
- Department of Hematology, Juntendo University School of Medicine, Japan
- Laboratory for the Development of Therapies against MPN, Juntendo University Graduate School of Medicine, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Japan
| | - Yasumitsu Kurokawa
- Department of Hematology, Juntendo University School of Medicine, Japan
- Department of Hematology, Ibaraki Prefectural Central Hospital, Japan
| | - Soji Morishita
- Laboratory for the Development of Therapies against MPN, Juntendo University Graduate School of Medicine, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Japan
| | - Takamasa Yamamoto
- Department of Clinical Laboratory, Juntendo University Hospital, Japan
| | - Marito Araki
- Laboratory for the Development of Therapies against MPN, Juntendo University Graduate School of Medicine, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Japan
| | - Norio Komatsu
- Department of Hematology, Juntendo University School of Medicine, Japan
- Laboratory for the Development of Therapies against MPN, Juntendo University Graduate School of Medicine, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Japan
| |
Collapse
|
224
|
Calabresi L, Balliu M, Bartalucci N. Immunoblotting-assisted assessment of JAK/STAT and PI3K/Akt/mTOR signaling in myeloproliferative neoplasms CD34+ stem cells. Methods Cell Biol 2022; 171:81-109. [DOI: 10.1016/bs.mcb.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
225
|
Veitia R, Innan H. Pathogenic 'germline' variants associated with myeloproliferative disorders in apparently normal individuals: inherited or acquired genetic alterations? Clin Genet 2021; 101:371-374. [PMID: 34958119 DOI: 10.1111/cge.14104] [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: 11/18/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 11/26/2022]
Abstract
Myeloproliferative syndromes (MPS) are hematologic malignancies due to the expansion of an abnormal hematopoietic stem cell. They include chronic myeloid leukemia (CML) and non-CML MPS such as polycythemia vera, essential thrombocythemia and primary myelofibrosis. The latter are distinguished by somatic pathogenic variants affecting JAK2, CALR or MPL genes. Apparent germline pathogenic variants have been reported in the general population. Here, we found that two gnomAD data-sets report more homozygotes than expected for the JAK2 c.1849G > T(Val617Phe) variant. We propose that somatic gene conversion underlies the presence of those unexpected homozygotes in normal populations. Consistently, homozygous individuals are older than 65 years. We also found a lower-than-expected frequency of the JAK2 variant in younger individuals suggesting that somatic mutation can underlie its presence in (at least some) heterozygotes. Regarding pathogenic variants in MPL and CALR, they are also present in the gnomAD data-sets explored. However, we cannot conclude that such seemingly germline variants are in fact somatic alterations. These results suggest that apparently normal individuals bearing MPS-related variants can be subclinical/undiagnosed MPS cases of somatic origin. It would be interesting to assess the hematologic phenotype of such individuals and the presence of the relevant variants in other tissues.
Collapse
Affiliation(s)
- Reiner Veitia
- Université de Paris, 752052, CNRS UMR7592, Paris 75013, France. Paris Cedex 13, France.,Institut Jacques Monod, CNRS UMR7592, Paris 75013, France. Paris Cedex 13, France.,Université Paris Saclay, CEA, Fontenay aux Roses, France.,Institut de Biologie François Jacob, CEA, Fontenay aux Roses, France
| | - Hideki Innan
- Graduate University for Advanced Studies, Hayama, Kanagawa, Japan
| |
Collapse
|
226
|
Belčič Mikič T, Vratanar B, Pajič T, Anžej Doma S, Debeljak N, Preložnik Zupan I, Sever M, Zver S. Is It Possible to Predict Clonal Thrombocytosis in Triple-Negative Patients with Isolated Thrombocytosis Based Only on Clinical or Blood Findings? J Clin Med 2021; 10:jcm10245803. [PMID: 34945099 PMCID: PMC8706709 DOI: 10.3390/jcm10245803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 11/29/2022] Open
Abstract
JAK2, MPL, and CALR mutations define clonal thrombocytosis in about 90% of patients with sustained isolated thrombocytosis. In the remainder of patients (triple-negative patients) diagnosing clonal thrombocytosis is especially difficult due to the different underlying conditions and possible inconclusive bone marrow biopsy results. The ability to predict patients with sustained isolated thrombocytosis with a potential clonal origin has a prognostic value and warrants further examination. The aim of our study was to define a non-invasive clinical or blood parameter that could help predict clonal thrombocytosis in triple-negative patients. We studied 237 JAK2 V617-negative patients who were diagnosed with isolated thrombocytosis and referred to the haematology service. Sixteen routine clinical and blood parameters were included in the logistic regression model which was used to predict the type of thrombocytosis (reactive/clonal). Platelet count and lactate dehydrogenase (LDH) were the only statistically significant predictors of clonal thrombocytosis. The platelet count threshold for the most accurate prediction of clonal or reactive thrombocytosis was 449 × 109/L. Other tested clinical and blood parameters were not statistically significant predictors of clonal thrombocytosis. The level of LDH was significantly higher in CALR-positive patients compared to CALR-negative patients. We did not identify any new clinical or blood parameters that could distinguish clonal from reactive thrombocytosis. When diagnosing clonal thrombocytosis triple-negative patients are most likely to be misdiagnosed. Treatment in patients with suspected triple negative clonal thrombocytosis should not be delayed if cardiovascular risk factors or pregnancy coexist, even in the absence of firm diagnostic criteria. In those cases the approach “better treat more than less” should be followed.
Collapse
Affiliation(s)
- Tanja Belčič Mikič
- Department of Haematology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (T.P.); (S.A.D.); (I.P.Z.); (M.S.); (S.Z.)
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence:
| | - Bor Vratanar
- Institute of Biomedical Statistics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Tadej Pajič
- Department of Haematology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (T.P.); (S.A.D.); (I.P.Z.); (M.S.); (S.Z.)
- Clinical Institute for Genomic Medicine, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Clinical Biochemistry, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Saša Anžej Doma
- Department of Haematology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (T.P.); (S.A.D.); (I.P.Z.); (M.S.); (S.Z.)
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Nataša Debeljak
- Medical Centre for Molecular Biology, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Irena Preložnik Zupan
- Department of Haematology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (T.P.); (S.A.D.); (I.P.Z.); (M.S.); (S.Z.)
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Matjaž Sever
- Department of Haematology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (T.P.); (S.A.D.); (I.P.Z.); (M.S.); (S.Z.)
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Samo Zver
- Department of Haematology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (T.P.); (S.A.D.); (I.P.Z.); (M.S.); (S.Z.)
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
227
|
Spivak JL. Advances in polycythemia vera and lessons for acute leukemia. Best Pract Res Clin Haematol 2021; 34:101330. [PMID: 34865702 DOI: 10.1016/j.beha.2021.101330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The myeloproliferative neoplasms (MPN), polycythemia vera (PV), essential thrombocytosis and primary myelofibrosis, are an unusual group of myeloid neoplasms, which arise in a pluripotent hematopoietic stem cell (HSC) due to gain of function driver mutations in the JAK2, CALR and MPL genes that constitutively activate JAK2, the cognate tyrosine kinase of the type 1 hematopoietic growth factor (HGF) receptors. PV is the ultimate phenotypic expression of constitutive JAK2 activation since it alone of the three MPN is characterized by overproduction of normal red cells, white cells and platelets. Paradoxically, however, although PV is a panmyelopathy involving myeloid, erythroid and megakaryocytic progenitor cells, pluripotent HSC only express a single type of HGF receptor, the thrombopoietin receptor, MPL. In this review, the basis for how a pluripotent HSC with one type of HGF can give rise to three separate types of myeloid cells will be explained and it will be demonstrated that PV is actually a hormone-sensitive disorder, characterized by elevated thrombopoietin levels. Finally, it will be shown that the most common form of acute leukemia in PV is due to the inappropriate use of chemotherapy, including hydroxyurea, which facilitates expansion of DNA-damaged, mutated HSC at the expense of their normal counterparts.
Collapse
Affiliation(s)
- Jerry L Spivak
- Division of Hematology, Johns Hopkins University School of Medicine, Traylor 924, 720 Rutland Avenue, Baltimore, MD, 20037, USA.
| |
Collapse
|
228
|
Essential Thrombocythemia in Children and Adolescents. Cancers (Basel) 2021; 13:cancers13236147. [PMID: 34885256 PMCID: PMC8656963 DOI: 10.3390/cancers13236147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 02/02/2023] Open
Abstract
Simple Summary Among chronic Ph-negative myeloproliferative neoplasms, essential thrombocythemia is found in children with low but increasing incidence. The diagnostic and clinical features do not completely overlap with ET of adult age. A significant number of cases, in fact, do not meet the criteria of clonality, and many cases require extensive clinical evaluation to exclude secondary, reactive forms. Therefore, histological analysis of bone marrow biopsy is necessary, and its use should be enforced. The clinical course appears to be more benign, at least within the first decades of observation, with the incidence of thrombotic events being much lower than in adults (4 % vs. 30%). Hemorrhages are mostly irrelevant. Therefore, the management should be carefully adapted to the individual patient, balancing the risk of future complications with long-term collateral effects of any drug. This review analyzes the peculiarities of the disease facing similarities and differences with adult scenarios. Abstract This paper reviews the features of pediatric essential thrombocythemia (ET). ET is a rare disease in children, challenging pediatric and adult hematologists alike. The current WHO classification acknowledges classical Philadelphia-negative MPNs and defines diagnostic criteria, mainly encompassing adult cases. The presence of one of three driver mutations (JAK2V617F, CALR, and MPL mutations) represent the proof of clonality typical of ET. Pediatric ET cases are thus usually confronted by adult approaches. These can fit only some patients, because only 25–40% of cases present one of the driver mutations. The diagnosis of hereditary, familial thrombocytosis and the exclusion of reactive/secondary thrombocytosis must be part of the diagnostic process in children and can clarify most of the negative cases. Still, many children present a clinical, histological picture of ET, with a molecular triple wild-type status. Moreover, prognosis seems more benign, at least within the first few decades of follow-up. Thrombotic events are rare, and only minor hemorrhages are ordinarily observed. As per the management, the need to control symptoms must be balanced with the collateral effects of lifelong drug therapy. We conclude that these differences concert a compelling case for a very careful therapeutic approach and advocate for the importance of further cooperative studies.
Collapse
|
229
|
Mosca M, Hermange G, Tisserand A, Noble R, Marzac C, Marty C, Le Sueur C, Campario H, Vertenoeil G, El-Khoury M, Catelain C, Rameau P, Gella C, Lenglet J, Casadevall N, Favier R, Solary E, Cassinat B, Kiladjian JJ, Constantinescu SN, Pasquier F, Hochberg ME, Raslova H, Villeval JL, Girodon F, Vainchenker W, Cournède PH, Plo I. Inferring the dynamics of mutated hematopoietic stem and progenitor cells induced by IFNα in myeloproliferative neoplasms. Blood 2021; 138:2231-2243. [PMID: 34407546 PMCID: PMC8641097 DOI: 10.1182/blood.2021010986] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 07/15/2021] [Indexed: 11/20/2022] Open
Abstract
Classical BCR-ABL-negative myeloproliferative neoplasms (MPNs) are clonal disorders of hematopoietic stem cells (HSCs) caused mainly by recurrent mutations in genes encoding JAK2 (JAK2), calreticulin (CALR), or the thrombopoietin receptor (MPL). Interferon α (IFNα) has demonstrated some efficacy in inducing molecular remission in MPNs. To determine factors that influence molecular response rate, we evaluated the long-term molecular efficacy of IFNα in patients with MPN by monitoring the fate of cells carrying driver mutations in a prospective observational and longitudinal study of 48 patients over more than 5 years. We measured the clonal architecture of early and late hematopoietic progenitors (84 845 measurements) and the global variant allele frequency in mature cells (409 measurements) several times per year. Using mathematical modeling and hierarchical Bayesian inference, we further inferred the dynamics of IFNα-targeted mutated HSCs. Our data support the hypothesis that IFNα targets JAK2V617F HSCs by inducing their exit from quiescence and differentiation into progenitors. Our observations indicate that treatment efficacy is higher in homozygous than heterozygous JAK2V617F HSCs and increases with high IFNα dose in heterozygous JAK2V617F HSCs. We also found that the molecular responses of CALRm HSCs to IFNα were heterogeneous, varying between type 1 and type 2 CALRm, and a high dose of IFNα correlates with worse outcomes. Our work indicates that the long-term molecular efficacy of IFNα implies an HSC exhaustion mechanism and depends on both the driver mutation type and IFNα dose.
Collapse
Affiliation(s)
- Matthieu Mosca
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
- Laboratoire d'Excellence GR-Ex, Paris, France
| | - Gurvan Hermange
- Université Paris-Saclay, CentraleSupélec, Laboratory MICS (Laboratory of Applied Mathematics and Computer Science), Gif-sur-Yvette, France
| | - Amandine Tisserand
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Laboratoire d'Excellence GR-Ex, Paris, France
- Université de Paris, Paris, France
| | - Robert Noble
- Department of Biosciences and Engineering, ETH Zurich, Basel, Switzerland
- Institut des Sciences de l'Evolution, University of Montpellier, Montpellier, France
- Institute of Evolutionary Biology and Environmental Studies (IEU), University of Zurich, Zurich, Switzerland
- University of London, London, United Kingdom
| | - Christophe Marzac
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
- Laboratoire d'Immuno-Hématologie, Gustave Roussy, Villejuif, France
| | - Caroline Marty
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
- Laboratoire d'Excellence GR-Ex, Paris, France
| | - Cécile Le Sueur
- Department of Biosciences and Engineering, ETH Zurich, Basel, Switzerland
| | | | - Gaëlle Vertenoeil
- Ludwig Institute for Cancer Research and Université Catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Mira El-Khoury
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Laboratoire d'Excellence GR-Ex, Paris, France
| | - Cyril Catelain
- UMS AMMICa-Plateforme Imagerie et Cytométries, Gustave Roussy, Villejuif, France
| | - Philippe Rameau
- UMS AMMICa-Plateforme Imagerie et Cytométries, Gustave Roussy, Villejuif, France
| | - Cyril Gella
- Laboratoire d'Immuno-Hématologie, Gustave Roussy, Villejuif, France
| | | | - Nicole Casadevall
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Assistance Publique des Hôpitaux de Paris, Laboratoire d'Hématologie, Hôpital Saint-Antoine, Paris, France
| | - Rémi Favier
- Assistance Publique des Hôpitaux de Paris, Service d'Hématologie Biologique, Hôpital d'Enfants Armand-Trousseau, Paris, France
| | - Eric Solary
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Département d'Hématologie, Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Bruno Cassinat
- Université de Paris, INSERM UMR-S 1131, Institut de Recherche Saint-Louis (IRSL), Hôpital Saint-Louis, Paris, France
- Assistance Publique des Hôpitaux de Paris, Laboratoire de Biologie Cellulaire
| | - Jean-Jacques Kiladjian
- Université de Paris, INSERM UMR-S 1131, Institut de Recherche Saint-Louis (IRSL), Hôpital Saint-Louis, Paris, France
- Assistance Publique des Hôpitaux de Paris, Centre d'Investigations Cliniques, Hôpital Saint-Louis, Paris, France
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research and Université Catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Florence Pasquier
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
- Département d'Hématologie, Gustave Roussy, Villejuif, France
| | - Michael E Hochberg
- Institut des Sciences de l'Evolution, University of Montpellier, Montpellier, France
- Santa Fe Institute, Santa Fe, NM
| | - Hana Raslova
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
| | - Jean-Luc Villeval
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
| | - François Girodon
- Laboratoire d'Hématologie, CHU Dijon, Dijon, France
- INSERM, UMR 866, Centre de Recherche, Dijon, France; and
| | - William Vainchenker
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
- Laboratoire d'Excellence GR-Ex, Paris, France
- Assistance Publique des Hôpitaux de Paris, Service d'Immunopathologie Clinique, Polyclinique d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - Paul-Henry Cournède
- Université Paris-Saclay, CentraleSupélec, Laboratory MICS (Laboratory of Applied Mathematics and Computer Science), Gif-sur-Yvette, France
| | - Isabelle Plo
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
- Laboratoire d'Excellence GR-Ex, Paris, France
| |
Collapse
|
230
|
Thrombosis in myeloproliferative neoplasms: A clinical and pathophysiological perspective. THROMBOSIS UPDATE 2021. [DOI: 10.1016/j.tru.2021.100081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
231
|
Celik H, Krug E, Zhang CR, Han W, Issa N, Koh WK, Bjeije H, Kukhar O, Allen M, Li T, Fisher DAC, Fowles JS, Wong TN, Stubbs MC, Koblish HK, Oh ST, Challen GA. A Humanized Animal Model Predicts Clonal Evolution and Therapeutic Vulnerabilities in Myeloproliferative Neoplasms. Cancer Discov 2021; 11:3126-3141. [PMID: 34193440 PMCID: PMC8716669 DOI: 10.1158/2159-8290.cd-20-1652] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 06/04/2021] [Accepted: 06/29/2021] [Indexed: 11/16/2022]
Abstract
Myeloproliferative neoplasms (MPN) are chronic blood diseases with significant morbidity and mortality. Although sequencing studies have elucidated the genetic mutations that drive these diseases, MPNs remain largely incurable with a significant proportion of patients progressing to rapidly fatal secondary acute myeloid leukemia (sAML). Therapeutic discovery has been hampered by the inability of genetically engineered mouse models to generate key human pathologies such as bone marrow fibrosis. To circumvent these limitations, here we present a humanized animal model of myelofibrosis (MF) patient-derived xenografts (PDX). These PDXs robustly engrafted patient cells that recapitulated the patient's genetic hierarchy and pathologies such as reticulin fibrosis and propagation of MPN-initiating stem cells. The model can select for engraftment of rare leukemic subclones to identify patients with MF at risk for sAML transformation and can be used as a platform for genetic target validation and therapeutic discovery. We present a novel but generalizable model to study human MPN biology. SIGNIFICANCE Although the genetic events driving MPNs are well defined, therapeutic discovery has been hampered by the inability of murine models to replicate key patient pathologies. Here, we present a PDX system to model human myelofibrosis that reproduces human pathologies and is amenable to genetic and pharmacologic manipulation. This article is highlighted in the In This Issue feature, p. 2945.
Collapse
Affiliation(s)
- Hamza Celik
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Ethan Krug
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Christine R Zhang
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Wentao Han
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Nancy Issa
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Won Kyun Koh
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Hassan Bjeije
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Ostap Kukhar
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Maggie Allen
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Tiandao Li
- Center of Regenerative Medicine, Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri
| | - Daniel A C Fisher
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jared S Fowles
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Terrence N Wong
- Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | | | | | - Stephen T Oh
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Grant A Challen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
232
|
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
|
233
|
Hashimoto Y, Ito T, Gotoh A, Nakamae M, Kimura F, Koike M, Kirito K, Wada H, Usuki K, Tanaka T, Mori T, Wakita S, Saito TI, Kada A, Saito AM, Shimoda K, Sugimoto Y, Kurokawa T, Tomita A, Edahiro Y, Akashi K, Matsumura I, Takenaka K, Komatsu N. Clinical characteristics, prognostic factors, and outcomes of patients with essential thrombocythemia in Japan: the JSH-MPN-R18 study. Int J Hematol 2021; 115:208-221. [PMID: 34727329 DOI: 10.1007/s12185-021-03253-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/23/2021] [Accepted: 10/24/2021] [Indexed: 11/25/2022]
Abstract
We conducted a large-scale, nationwide retrospective study of Japanese patients who were diagnosed with essential thrombocythemia based on the diagnostic criteria in the World Health Organization classification. We investigated clinical characteristics, survival rates, and the incidence of thrombohemorrhagic events as well as risk factors for these events. A total of 1152 patients were analyzed in the present study. Median age at diagnosis was 65 years, the median platelet count was 832 × 109/L, and the positive mutation rates of JAK2V617F, CALR, and MPL were 62.8, 25.1, and 4.1%, respectively. Compared with European and American patients, Japanese patients were more likely to have cardiovascular risk factors and less likely to have systemic symptoms including palpable splenomegaly. Thrombocytosis was identified as a risk factor for hemorrhagic events and prognosis, but not for thrombotic events. The prognostic factors and risk classifications reported in Europe and the United States were generally applicable to Japanese patients. Regarding transformations, secondary myelofibrosis progressed in a time-dependent manner, but progression to acute leukemia was low in "true" ET patients. Skin cancers were less common and gastrointestinal cancers more common as secondary malignancies in Japanese patients, suggesting ethnic differences.
Collapse
Affiliation(s)
- Yoshinori Hashimoto
- Department of Hematology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
- Laboratory for the Development of Therapies Against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomoki Ito
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Akihiko Gotoh
- Department of Hematology, Tokyo Medical University, Tokyo, Japan
| | - Mika Nakamae
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Fumihiko Kimura
- Division of Hematology, Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Michiaki Koike
- Department of Hematology, Juntendo University Shizuoka Hospital, Shizuoka, Japan
| | - Keita Kirito
- Department of Hematology and Oncology, University of Yamanashi, Yamanashi, Japan
| | - Hideho Wada
- Department of Hematology, Kawasaki Medical School, Okayama, Japan
| | - Kensuke Usuki
- Department of Hematology, NTT Medical Center Tokyo, Tokyo, Japan
| | - Takayuki Tanaka
- Department of Hematology, Tottori Prefectural Central Hospital, Tottori, Japan
| | - Takehiko Mori
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Hematology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoshi Wakita
- Division of Hematology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Toshiki I Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Aichi, Japan
| | - Akiko Kada
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Aichi, Japan
| | - Akiko M Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Aichi, Japan
| | - Kazuya Shimoda
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Yuka Sugimoto
- Department of Community Hematology, Mie University, Mie, Japan
| | - Toshiro Kurokawa
- Department of Hematology, Toyama Red Cross Hospital, Toyama, Japan
| | - Akihiro Tomita
- Department of Hematology, Fujita Health University School of Medicine, Aichi, Japan
| | - Yoko Edahiro
- Department of Hematology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
- Laboratory for the Development of Therapies Against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Itaru Matsumura
- Division of Hematology and Rheumatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Katsuto Takenaka
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Norio Komatsu
- Department of Hematology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Laboratory for the Development of Therapies Against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- PharmaEssentia Japan KK, Tokyo, Japan.
| |
Collapse
|
234
|
Abstract
PURPOSE OF THE REVIEW This review highlights recent advancements in understanding the regulation of platelet numbers, focusing on mechanisms by which carbohydrates (glycans) link platelet removal with platelet production in the bone marrow in health and disease. RECENT FINDINGS This review is focused on the role of carbohydrates, specifically sialic acid moieties, as a central mediator of platelet clearance. We discuss recently identified novel mechanisms of carbohydrate-mediated platelet removal and carbohydrate-binding receptors that mediate platelet removal. SUMMARY The platelet production rate by megakaryocytes and removal kinetics controls the circulating platelet count. Alterations in either process can lead to thrombocytopenia (low platelet count) or thrombocytosis (high platelet count) are associated with the risk of bleeding or overt thrombus formation and serious complications. Thus, regulation of a steady-state platelet count is vital in preventing adverse events. There are few mechanisms delineated that shed light on carbohydrates' role in the complex and massive platelet removal process. This review focuses on carbohydrate-related mechanisms that contribute to the control of platelet numbers.
Collapse
Affiliation(s)
- Leonardo Rivadeneyra
- Translational Glycomics Center, Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Herve Falet
- Translational Glycomics Center, Versiti Blood Research Institute, Milwaukee, WI, United States
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Karin M. Hoffmeister
- Translational Glycomics Center, Versiti Blood Research Institute, Milwaukee, WI, United States
- Departments of Medicine and Biochemistry, Medical College of Wisconsin, Milwaukee WI, United States
| |
Collapse
|
235
|
Loscocco GG, Guglielmelli P, Gangat N, Rossi E, Mannarelli C, Betti S, Maccari C, Ramundo F, Jadoon Y, Gesullo F, Ceglie S, Paoli C, Pardanani A, De Stefano V, Tefferi A, Vannucchi AM. Clinical and molecular predictors of fibrotic progression in essential thrombocythemia: A multicenter study involving 1607 patients. Am J Hematol 2021; 96:1472-1480. [PMID: 34424575 PMCID: PMC9293196 DOI: 10.1002/ajh.26332] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/14/2021] [Accepted: 08/17/2021] [Indexed: 12/31/2022]
Abstract
The current retrospective study involving a total of 1607 patients was designed to identify clinical and molecular variables that were predictive of inferior myelofibrosis‐free survival (MFS) in WHO‐defined essential thrombocythemia (ET), utilizing three independent patient cohorts: University of Florence, Italy (n = 718); Mayo Clinic, USA (n = 479) and Policlinico Gemelli, Catholic University, Rome, Italy (n = 410). The Florence patient cohort was first examined to identify independent risk factors for MFS, which included age > 60 years (HR 2.5, 95% CI 1.3–4.9), male sex (2.1, 1.2–3.9), palpable splenomegaly (2.1, 1.2–3.9), CALR 1/1‐like or MPL mutation (3.4, 1.9–6.1) and JAK2V617F variant allele frequency > 35% (4.2, 1.6–10.8). Subsequently, an operational molecular risk category was developed and validated in the other two cohorts from Mayo Clinic and Rome: “high molecular risk” category included patients with JAK2V617F VAF >35%, CALR type 1/1‐like or MPL mutations; all other driver mutation profiles were assigned to “low molecular risk” category. The former, compared to the latter molecular risk category, displayed significantly higher risk of fibrotic transformation: Florence cohort with respective fibrotic transformation risk rates of 8% vs. 1.2% at 10 years and 33% vs. 8% at 20 years (p < 0.001; HR 6.1; 95% CI 3.2–11.7); Mayo Cohort, 16% vs. 7% at 10 years and 44% vs. 25% at 20 years (p < 0.001; HR 2.5; 95% CI 1.6–4.1); and Rome cohort 7.8% vs. 4.6% at 10 years and 31.2% vs. 7.1% at 20 years (p = 0.007, HR 2.7; 95% CI 1.3–5.8). The present study provides practically useful risk signals for fibrotic transformation in ET and facilitates identification of patients who require close monitoring and appropriate counseling.
Collapse
Affiliation(s)
- Giuseppe G. Loscocco
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| | - Paola Guglielmelli
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| | - Naseema Gangat
- Divisions of Hematology Mayo Clinic Rochester Minnesota USA
| | - Elena Rossi
- Section of Hematology, Department of Radiological and Hematological Sciences Catholic University Rome Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS Rome Italy
| | - Carmela Mannarelli
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| | - Silvia Betti
- Fondazione Policlinico Universitario A. Gemelli IRCCS Rome Italy
| | - Chiara Maccari
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| | - Francesco Ramundo
- Section of Hematology, Department of Radiological and Hematological Sciences Catholic University Rome Italy
| | - Yamna Jadoon
- Divisions of Hematology Mayo Clinic Rochester Minnesota USA
| | - Francesca Gesullo
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| | - Sara Ceglie
- Section of Hematology, Department of Radiological and Hematological Sciences Catholic University Rome Italy
| | - Chiara Paoli
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| | | | - Valerio De Stefano
- Section of Hematology, Department of Radiological and Hematological Sciences Catholic University Rome Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS Rome Italy
| | - Ayalew Tefferi
- Divisions of Hematology Mayo Clinic Rochester Minnesota USA
| | - Alessandro M. Vannucchi
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| |
Collapse
|
236
|
Thomas S, Krishnan A. Platelet Heterogeneity in Myeloproliferative Neoplasms. Arterioscler Thromb Vasc Biol 2021; 41:2661-2670. [PMID: 34615371 PMCID: PMC8551046 DOI: 10.1161/atvbaha.121.316373] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022]
Abstract
Myeloproliferative neoplasms (MPNs) are a group of malignant disorders of the bone marrow where a dysregulated balance between proliferation and differentiation gives rise to abnormal numbers of mature blood cells. MPNs encompass a spectrum of disease entities with progressively more severe clinical features, including complications with thrombosis and hemostasis and an increased propensity for transformation to acute myeloid leukemia. There is an unmet clinical need for markers of disease progression. Our understanding of the precise mechanisms that influence pathogenesis and disease progression has been limited by access to disease-specific cells as biosources. Here, we review the landscape of MPN pathology and present blood platelets as potential candidates for disease-specific understanding. We conclude with our recent work discovering progressive platelet heterogeneity by subtype in a large clinical cohort of patients with MPN.
Collapse
Affiliation(s)
- Sally Thomas
- Department of Oncology and Metabolism, University of Sheffield and Department of Haematology, Royal Hallamshire Hospital, United Kingdom (S.T.)
| | - Anandi Krishnan
- Department of Pathology, Stanford University School of Medicine, CA (A.K.)
| |
Collapse
|
237
|
Cross NCP, Godfrey AL, Cargo C, Garg M, Mead AJ. The use of genetic tests to diagnose and manage patients with myeloproliferative and myeloproliferative/myelodysplastic neoplasms, and related disorders. Br J Haematol 2021; 195:338-351. [PMID: 34409596 DOI: 10.1111/bjh.17766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 07/28/2021] [Indexed: 12/11/2022]
Affiliation(s)
- Nicholas C P Cross
- Wessex Regional Genetics Laboratory, Salisbury District Hospital, Salisbury, UK
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Anna L Godfrey
- Haematopathology & Oncology Diagnostics Service, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Catherine Cargo
- Haematological Malignancy Diagnostic Service, Leeds Cancer Centre, St James's University Hospital, Leeds, UK
| | - Mamta Garg
- Leicester Royal Infirmary, Infirmary Square, Leicester, UK
| | - Adam J Mead
- MRC Molecular Haematology Unit, NIHR Oxford Biomedical Research Centre, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
238
|
Penna D. New Horizons in Myeloproliferative Neoplasms Treatment: A Review of Current and Future Therapeutic Options. Medicina (B Aires) 2021; 57:medicina57111181. [PMID: 34833399 PMCID: PMC8619471 DOI: 10.3390/medicina57111181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 11/29/2022] Open
Abstract
Philadelphia-negative myeloproliferative neoplasms (MPN) are aggressive diseases characterized by clonal proliferation of myeloid stem cells. The clonal process leads to excessive red cells production, platelets production, and bone marrow fibrosis. According to the phenotype, MPN can be classified as polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). MPN patients have shortened survival due to the increased risk of thrombosis, hemorrhages, and transformation to acute myeloid leukemia (AML). Prognosis is variable, with a shorter life expectancy in myelofibrosis. Currently, drug therapy can reduce symptoms, splenomegaly, and risk of thrombosis. Still, some patients can be resistant or intolerant to the treatment. At the same time, allogeneic stem cell transplant (ASCT) is the only treatment modality with the potential to cure the disease. Nevertheless, the ASCT is reserved for high-risk leukemic progression patients due to the risk of treatment-related death and comorbidity. Therefore, there is a need for new drugs that can eradicate clonal hematopoiesis and prevent progression to more aggressive myeloid neoplasms. Thanks to the better understanding of the disease’s molecular pathogenesis, many new potentially disease-modifying drugs have been developed and are currently in clinical trials. This review explores the most promising new drugs currently in clinical trials.
Collapse
Affiliation(s)
- Domenico Penna
- Hematology Unit, Azienda Unità Sanitaria Locale—IRCCS, 42123 Reggio Emilia, Italy; ; Tel.: +39-522-296-623
- Ph.D. Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 42121 Modena, Italy
| |
Collapse
|
239
|
Easwar A, Siddon AJ. Genetic Landscape of Myeloproliferative Neoplasms with an Emphasis on Molecular Diagnostic Laboratory Testing. Life (Basel) 2021; 11:1158. [PMID: 34833034 PMCID: PMC8625510 DOI: 10.3390/life11111158] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/18/2021] [Accepted: 10/27/2021] [Indexed: 12/27/2022] Open
Abstract
Chronic myeloproliferative neoplasms (MPNs) are hematopoietic stem cell neoplasms with driver events including the BCR-ABL1 translocation leading to a diagnosis of chronic myeloid leukemia (CML), or somatic mutations in JAK2, CALR, or MPL resulting in Philadelphia-chromosome-negative MPNs with constitutive activation of the JAK-STAT signaling pathway. In the Philadelphia-chromosome-negative MPNs, modern sequencing panels have identified a vast molecular landscape including additional mutations in genes involved in splicing, signal transduction, DNA methylation, and chromatin modification such as ASXL1, SF3B1, SRSF2, and U2AF1. These additional mutations often influence prognosis in MPNs and therefore are increasingly important for risk stratification. This review focuses on the molecular alterations within the WHO classification of MPNs and laboratory testing used for diagnosis.
Collapse
Affiliation(s)
- Arti Easwar
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06510, USA;
| | - Alexa J. Siddon
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06510, USA;
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
240
|
Sant'Antonio E, Borsani O, Camerini C, Botta C, Santoro M, Allegra A, Siragusa S. Philadelphia chromosome-negative myeloproliferative neoplasms in younger adults: A critical discussion of unmet medical needs, with a focus on pregnancy. Blood Rev 2021; 52:100903. [PMID: 34742614 DOI: 10.1016/j.blre.2021.100903] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/23/2021] [Accepted: 10/27/2021] [Indexed: 11/02/2022]
Abstract
Myeloproliferative neoplasms (MPN) are traditionally regarded as a disease of older adults, though a not negligible fraction of cases occurs at a younger age, including women of childbearing potential. MPN in younger patients, indeed, offer several challenges for the clinical hematologist, that goes from difficulties in reaching a timely and accurate diagnosis to a peculiar thrombotic risk, with a relatively high incidence of thromboses in unusual sites (as the splanchnic veins or the cerebral ones). Moreover, the issue of pregnancy is recently gaining more attention as maternal age is rising and molecular screening are widely implemented, leading to a better recognition of these cases, both before and during pregnancy. In the present work we aim at discussing four clinical topic that we identified as areas of uncertainty or true unmet medical needs in the management of younger patients with MPN, with a particular focus on the topic of pregnancy. For each of these topics, we critically reviewed the available evidence that support treatment decisions, though acknowledging that recommendations in this field are mostly based on expert opinion or derived from guidelines of other clinical conditions that share with MPN a high vascular risk, as antiphospholipid syndrome. Taking into consideration both the lack of evidence-based data and the clinical heterogeneity of MPN, we support an individualized strategy of counseling and management for both young patients and for expectant mother with MPN.
Collapse
Affiliation(s)
| | - Oscar Borsani
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Chiara Camerini
- Division of Hematology, Azienda USL Toscana Nord Ovest, Ospedale San Luca, Lucca, Italy
| | - Cirino Botta
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Marco Santoro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Messina, Italy
| | - Sergio Siragusa
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| |
Collapse
|
241
|
Hu X, Feng C, Zhou Y, Harrison A, Chen M. DeepTrio: a ternary prediction system for protein-protein interaction using mask multiple parallel convolutional neural networks. Bioinformatics 2021; 38:694-702. [PMID: 34694333 PMCID: PMC8756175 DOI: 10.1093/bioinformatics/btab737] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/05/2021] [Accepted: 10/20/2021] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION Protein-protein interaction (PPI), as a relative property, is determined by two binding proteins, which brings a great challenge to design an expert model with an unbiased learning architecture and a superior generalization performance. Additionally, few efforts have been made to allow PPI predictors to discriminate between relative properties and intrinsic properties. RESULTS We present a sequence-based approach, DeepTrio, for PPI prediction using mask multiple parallel convolutional neural networks. Experimental evaluations show that DeepTrio achieves a better performance over several state-of-the-art methods in terms of various quality metrics. Besides, DeepTrio is extended to provide additional insights into the contribution of each input neuron to the prediction results. AVAILABILITY AND IMPLEMENTATION We provide an online application at http://bis.zju.edu.cn/deeptrio. The DeepTrio models and training data are deposited at https://github.com/huxiaoti/deeptrio.git. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Xiaotian Hu
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Cong Feng
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yincong Zhou
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Andrew Harrison
- Department of Mathematical Sciences, University of Essex, Colchester CO4 3SQ, UK
| | - Ming Chen
- To whom correspondence should be addressed.
| |
Collapse
|
242
|
From Metcalf to myeloproliferative neoplasms - a personal journey. Exp Hematol 2021; 105:2-9. [PMID: 34706253 DOI: 10.1016/j.exphem.2021.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 11/20/2022]
Abstract
The human myeloproliferative neoplasms constitute a biologically fascinating group of chronic myeloid malignancies. This perspective outlines how a postdoctoral fellowship working in Don Metcalf's unit proved a formative and immensely enjoyable experience for my family and me. It laid the foundation for a subsequent body of work over three decades that revealed the genetic basis of these diseases, defined how these genetic alterations subvert normal hematopoiesis, altered clinical practice, and provided insights of broad biological relevance for cancer and cytokine signaling.
Collapse
|
243
|
Campanelli R, Massa M, Rosti V, Barosi G. New Markers of Disease Progression in Myelofibrosis. Cancers (Basel) 2021; 13:5324. [PMID: 34771488 PMCID: PMC8582535 DOI: 10.3390/cancers13215324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/30/2022] Open
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm due to the clonal proliferation of a hematopoietic stem cell. The vast majority of patients harbor a somatic gain of function mutation either of JAK2 or MPL or CALR genes in their hematopoietic cells, resulting in the activation of the JAK/STAT pathway. Patients display variable clinical and laboratoristic features, including anemia, thrombocytopenia, splenomegaly, thrombotic complications, systemic symptoms, and curtailed survival due to infections, thrombo-hemorrhagic events, or progression to leukemic transformation. New drugs have been developed in the last decade for the treatment of PMF-associated symptoms; however, the only curative option is currently represented by allogeneic hematopoietic cell transplantation, which can only be offered to a small percentage of patients. Disease prognosis is based at diagnosis on the classical International Prognostic Scoring System (IPSS) and Dynamic-IPSS (during disease course), which comprehend clinical parameters; recently, new prognostic scoring systems, including genetic and molecular parameters, have been proposed as meaningful tools for a better patient stratification. Moreover, new biological markers predicting clinical evolution and patient survival have been associated with the disease. This review summarizes basic concepts of PMF pathogenesis, clinics, and therapy, focusing on classical prognostic scoring systems and new biological markers of the disease.
Collapse
Affiliation(s)
- Rita Campanelli
- Center for the Study of Myelofibrosis, General Medicine 2—Center for Systemic Amyloidosis and High-Complexity Diseases, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy; (V.R.); (G.B.)
| | - Margherita Massa
- General Medicine 2—Center for Systemic Amyloidosis and High-Complexity Diseases, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy;
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, General Medicine 2—Center for Systemic Amyloidosis and High-Complexity Diseases, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy; (V.R.); (G.B.)
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis, General Medicine 2—Center for Systemic Amyloidosis and High-Complexity Diseases, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy; (V.R.); (G.B.)
| |
Collapse
|
244
|
Schischlik F. Transcriptional configurations of myeloproliferative neoplasms. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 366:25-39. [PMID: 35153005 DOI: 10.1016/bs.ircmb.2021.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Myeloproliferative neoplasms (MPNs) is an umbrella term for several heterogenous diseases, which are characterized by their stem cell origin, clonal hematopoiesis and increase of blood cells of the myeloid lineage. The focus will be on BCR-ABL1 negative MPNs, polycythemia vera (PV), primary myelofibrosis (PMF), essential thrombocythemia (ET). Seminal findings in the field of MPN were driven by genomic analysis, focusing on dissecting genomic changes MPN patients. This led to identification of major MPN driver genes, JAK2, MPL and CALR. Transcriptomic analysis promises to bridge the gap between genetic and phenotypic characterization of each patient's tumor and with the advent of single cell sequencing even for each MPN cancer cell. This review will focus on efforts to mine the bulk transcriptome of MPN patients, including analysis of fusion genes and splicing alterations which can be addressed with RNA-seq technologies. Furthermore, this paper aims to review recent endeavors to elucidate tumor heterogeneity in MPN hematopoietic stem and progenitor cells using single cell technologies. Finally, it will highlight current shortcoming and future applications to advance the field in MPN biology and improve patient diagnostics using RNA-based assays.
Collapse
Affiliation(s)
- Fiorella Schischlik
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States.
| |
Collapse
|
245
|
Goel H, Rahul E, Gupta I, Chopra A, Ranjan A, Gupta AK, Meena JP, Viswanathan GK, Bakhshi S, Misra A, Hussain S, Kumar R, Singh A, Rath GK, Sharma A, Mittan S, Tanwar P. Molecular and genomic landscapes in secondary & therapy related acute myeloid leukemia. AMERICAN JOURNAL OF BLOOD RESEARCH 2021; 11:472-497. [PMID: 34824881 PMCID: PMC8610791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 09/04/2021] [Indexed: 06/13/2023]
Abstract
Acute myeloid leukemia (AML) is a complex, aggressive myeloid neoplasm characterized by frequent somatic mutations that influence different functional categories' genes, resulting in maturational arrest and clonal expansion. AML can arise de novo (dn-AML) or can be secondary AML (s-AML) refers to a leukemic process which may arise from an antecedent hematologic disorder (AHD-AML), mostly from a myelodysplastic syndrome (MDS) or myeloproliferative neoplasm (MPN) or can be the result of an antecedent cytotoxic chemotherapy or radiation therapy (therapy-related AML, t-AML). Clinical and biological features in secondary and therapy-related AML are distinct from de novo AML. Secondary and therapy-related AML occurs mainly in the elderly population and responds worse to therapy with higher relapse rates due to resistance to cytotoxic chemotherapy. Over the last decade, advances in molecular genetics have disclosed the sub-clonal architecture of secondary and therapy-related AML. Recent investigations have revealed that cytogenetic abnormalities and underlying genetic aberrations (mutations) are likely to be significant factors dictating prognosis and critical impacts on treatment outcome. Secondary and therapy-related AML have a poorer outcome with adverse cytogenetic abnormalities and higher recurrences of unfavorable mutations compared to de novo AML. In this review, we present an overview of the clinical features of secondary and therapy-related AML and address the function of genetic mutations implicated in the pathogenesis of secondary leukemia. Detailed knowledge of the pathogenetic mechanisms gives an overview of new prognostic markers, including targetable mutations that will presumably lead to the designing and developing novel molecular targeted therapies for secondary and therapy-related AML. Despite significant advances in knowing the genetic aspect of secondary and therapy-related AML, its influence on the disease's pathophysiology, standard treatment prospects have not significantly evolved during the past three decades. Thus, we conclude this review by summarizing the modern and developing treatment strategies in secondary and therapy-related acute myeloid leukemia.
Collapse
Affiliation(s)
- Harsh Goel
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| | - Ekta Rahul
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| | - Ishan Gupta
- All India Institute of Medical SciencesNew Delhi 110029, India
| | - Anita Chopra
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| | - Amar Ranjan
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| | - Aditya Kumar Gupta
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Jagdish Prasad Meena
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Ganesh Kumar Viswanathan
- Department of Hematology, All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Aroonima Misra
- National Institute of Pathology, ICMRNew Delhi 110029, India
| | - Showket Hussain
- Division Of Molecular Oncology, National Institute of Cancer Prevention & Research I-7, Sector-39Noida 201301, India
| | - Ritesh Kumar
- Department of Radiation Oncology, Rudgers Cancer Institute of New JerseyNJ 07103, United States
| | - Archana Singh
- Department of Pathology, College of Medical Sciences, Rajasthan University of Health SciencesJaipur 302033, India
| | - GK Rath
- Department of Radiotherapy, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Sandeep Mittan
- Department of Cardiology, Ichan School of Medicine, Mount Sinai Hospital1468 Madison Avenue, New York 10028, United States
| | - Pranay Tanwar
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| |
Collapse
|
246
|
Stivala S, Meyer SC. Recent Advances in Molecular Diagnostics and Targeted Therapy of Myeloproliferative Neoplasms. Cancers (Basel) 2021; 13:cancers13205035. [PMID: 34680185 PMCID: PMC8534234 DOI: 10.3390/cancers13205035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/24/2021] [Accepted: 10/04/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Myeloproliferative neoplasms (MPN) are clonal hematologic malignancies with dysregulated myeloid blood cell production driven by JAK2, calreticulin, and MPL gene mutations. Technological advances have revealed a heterogeneous genomic landscape with additional mutations mainly in epigenetic regulators and splicing factors, which are of diagnostic and prognostic value and may inform treatment decisions. Thus, genetic testing has become an integral part of the state-of-the-art work-up for MPN. The finding that JAK2, CALR, and MPL mutations activate JAK2 signaling has promoted the development of targeted JAK2 inhibitor therapies. However, their disease-modifying potential remains limited and investigations of additional molecular vulnerabilities in MPN are imperative to advance the development of new therapeutic options. Here, we summarize the current insights into the genetic basis of MPN, its use as diagnostic and prognostic tool in clinical settings, and recent advances in targeted therapies for MPN. Abstract Somatic mutations in JAK2, calreticulin, and MPL genes drive myeloproliferative neoplasms (MPN), and recent technological advances have revealed a heterogeneous genomic landscape with additional mutations in MPN. These mainly affect genes involved in epigenetic regulation and splicing and are of diagnostic and prognostic value, predicting the risk of progression and informing decisions on therapeutic management. Thus, genetic testing has become an integral part of the current state-of-the-art laboratory work-up for MPN patients and has been implemented in current guidelines for disease classification, tools for prognostic risk assessment, and recommendations for therapy. The finding that JAK2, CALR, and MPL driver mutations activate JAK2 signaling has provided a rational basis for the development of targeted JAK2 inhibitor therapies and has fueled their translation into clinical practice. However, the disease-modifying potential of JAK2 inhibitors remains limited and is further impeded by loss of therapeutic responses in a substantial proportion of patients over time. Therefore, the investigation of additional molecular vulnerabilities involved in MPN pathogenesis is imperative to advance the development of new therapeutic options. Combination of novel compounds with JAK2 inhibitors are of specific interest to enhance therapeutic efficacy of molecularly targeted treatment approaches. Here, we summarize the current insights into the genetic basis of MPN, its use as a diagnostic and prognostic tool in clinical settings, and the most recent advances in targeted therapies for MPN.
Collapse
Affiliation(s)
- Simona Stivala
- Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland;
| | - Sara C. Meyer
- Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland;
- Division of Hematology, University Hospital Basel, 4031 Basel, Switzerland
- Correspondence: ; Tel.: +41-61-556-5965; Fax: +41-61-265-4568
| |
Collapse
|
247
|
Watanabe A, Mizoguchi I, Hasegawa H, Katahira Y, Inoue S, Sakamoto E, Furusaka Y, Sekine A, Miyakawa S, Murakami F, Xu M, Yoneto T, Yoshimoto T. A Chaperone-Like Role for EBI3 in Collaboration With Calnexin Under Inflammatory Conditions. Front Immunol 2021; 12:757669. [PMID: 34603342 PMCID: PMC8484754 DOI: 10.3389/fimmu.2021.757669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/02/2021] [Indexed: 01/31/2023] Open
Abstract
The interleukin-6 (IL-6)/IL-12 family of cytokines plays critical roles in the induction and regulation of innate and adaptive immune responses. Among the various cytokines, only this family has the unique characteristic of being composed of two distinct subunits, α- and β-subunits, which form a heterodimer with subunits that occur in other cytokines as well. Recently, we found a novel intracellular role for one of the α-subunits, Epstein-Barr virus-induced gene 3 (EBI3), in promoting the proper folding of target proteins and augmenting its expression at the protein level by binding to its target protein and a well-characterized lectin chaperone, calnexin, presumably through enhancing chaperone activity. Because calnexin is ubiquitously and constitutively expressed but EBI3 expression is inducible, these results could open an avenue to establish a new paradigm in which EBI3 plays an important role in further increasing the expression of target molecules at the protein level in collaboration with calnexin under inflammatory conditions. This theory well accounts for the heterodimer formation of EBI3 with p28, and probably with p35 and p19 to produce IL-27, IL-35, and IL-39, respectively. In line with this concept, another β-subunit, p40, plays a critical role in the assembly-induced proper folding of p35 and p19 to produce IL-12 and IL-23, respectively. Thus, chaperone-like activities in proper folding and maturation, which allow the secretion of biologically active heterodimeric cytokines, have recently been highlighted. This review summarizes the current understanding of chaperone-like activities of EBI3 to form heterodimers and other associations together with their possible biological implications.
Collapse
Affiliation(s)
- Aruma Watanabe
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Izuru Mizoguchi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Hideaki Hasegawa
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Yasuhiro Katahira
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Shinya Inoue
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Eri Sakamoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Yuma Furusaka
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Ami Sekine
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Satomi Miyakawa
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Fumihiro Murakami
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Mingli Xu
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Toshihiko Yoneto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Takayuki Yoshimoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
248
|
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
|
249
|
Coltro G, Loscocco GG, Vannucchi AM. Classical Philadelphia-negative myeloproliferative neoplasms (MPNs): A continuum of different disease entities. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 365:1-69. [PMID: 34756241 DOI: 10.1016/bs.ircmb.2021.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Classical Philadelphia-negative myeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell-derived disorders characterized by uncontrolled proliferation of differentiated myeloid cells and close pathobiologic and clinical features. According to the 2016 World Health Organization (WHO) classification, MPNs include polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). The 2016 revision aimed in particular at strengthening the distinction between masked PV and JAK2-mutated ET, and between prefibrotic/early (pre-PMF) and overt PMF. Clinical manifestations in MPNs include constitutional symptoms, microvascular disorders, thrombosis and bleeding, splenomegaly secondary to extramedullary hematopoiesis, cytopenia-related symptoms, and progression to overt MF and acute leukemia. A dysregulation of the JAK/STAT pathway is the unifying mechanistic hallmark of MPNs, and is guided by somatic mutations in driver genes including JAK2, CALR and MPL. Additional mutations in myeloid neoplasm-associated genes have been also identified, with established prognostic relevance, particularly in PMF. Prognostication of MPN patients relies on disease-specific clinical models. The increasing knowledge of MPN biology led to the development of integrated clinical and molecular prognostic scores that allow a more refined stratification. Recently, the therapeutic landscape of MPNs has been revolutionized by the introduction of potent, selective JAK inhibitors (ruxolitinib, fedratinib), that proved effective in controlling disease-related symptoms and splenomegaly, yet leaving unmet critical needs, owing the lack of disease-modifying activity. In this review, we will deal with molecular, clinical, and therapeutic aspects of the three classical MPNs aiming at highlighting either shared characteristics, that overall define a continuum within a single disease family, and uniqueness, at the same time.
Collapse
Affiliation(s)
- Giacomo Coltro
- CRIMM, Center for Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giuseppe G Loscocco
- CRIMM, Center for Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandro M Vannucchi
- CRIMM, Center for Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| |
Collapse
|
250
|
Towards Understanding the Pathogenicity of DROSHA Mutations in Oncohematology. Cells 2021; 10:cells10092357. [PMID: 34572006 PMCID: PMC8471307 DOI: 10.3390/cells10092357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/01/2021] [Accepted: 09/06/2021] [Indexed: 11/16/2022] Open
Abstract
Myelodysplastic syndrome (MDS) refers to a heterogeneous group of closely related clonal hematopoietic disorders, which are characterized by accumulation of somatic mutations. The acquired mutation burden is suggested to define the pathway and consequent phenotype of the pathology. Recent studies have called attention to the role of miRNA biogenesis genes in MDS progression; in particular, the mutational pressure of the DROSHA gene was determined. Therefore, this highlights the importance of studying the impact of all collected missense mutations found within the DROSHA gene in oncohematology that might affect the functionality of the protein. In this study, the selected mutations were extensively examined by computational screening, and the most deleterious were subjected to a further molecular dynamic simulation in order to uncover the molecular mechanism of the structural damage to the protein altering its biological function. The most significant effect was found for variants I625K, L1047S, and H1170D, presumably affecting the endonuclease activity of DROSHA. Such alterations arisen during MDS progression should be taken into consideration as evoking certain clinical traits in the malignifying clonal evolution.
Collapse
|