1
|
Zhang Y, Zhao Y, Liu Y, Zhang M, Zhang J. New advances in the role of JAK2 V617F mutation in myeloproliferative neoplasms. Cancer 2024; 130:4229-4240. [PMID: 39277798 DOI: 10.1002/cncr.35559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/05/2024] [Accepted: 08/26/2024] [Indexed: 09/17/2024]
Abstract
The JAK2 V617F mutation is the most common driver gene in myeloproliferative neoplasm (MPN), which means that the JAK/STAT signaling pathway is persistently activated independent of cytokines, and plays an important part in the onset and development of MPN. The JAK inhibitors, although widely used in the clinical practice, are unable to eradicate MPN. Therefore, the unavoidable long-term treatment poses a serious burden for patients with MPN. It is established that the JAK2 V617F mutation, in addition its role in the JAK/STAT pathway, can promote cell proliferation, differentiation, anti-apoptosis, DNA damage accumulation, and other key biologic processes through multiple pathways. Other than that, the JAK2 V617F mutation affects the cardiovascular system through multiple mechanisms. Although JAK inhibitors cannot eradicate MPN cells, the combined use of JAK inhibitors and other drugs may have surprising effects. This requires an in-depth understanding of the mechanism of action of the JAK2 V617F mutation. In this review, the authors explored the role of the JAK2 V617F mutation in MPN from multiple aspects, including the mechanisms of non-JAK/STAT pathways, the regulation of cellular methylation, the induction of cellular DNA damage accumulation, and effects on the cardiovascular system, with the objective of providing valuable insights into multidrug combination therapy for MPN.
Collapse
Affiliation(s)
- Yongchao Zhang
- Department of Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yue Zhao
- Department of Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yusi Liu
- Department of Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Minyu Zhang
- Department of Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jihong Zhang
- Department of Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
2
|
Zhan H, Kaushansky K. Megakaryocytes as the Regulator of the Hematopoietic Vascular Niche. Front Oncol 2022; 12:912060. [PMID: 35814384 PMCID: PMC9258777 DOI: 10.3389/fonc.2022.912060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Megakaryocytes (MKs) are important components of the hematopoietic niche. Compared to the non-hematopoietic niche cells, MKs serving as part of the hematopoietic niche provides a mechanism for feedback regulation of hematopoietic stem cells (HSCs), in which HSC progeny (MKs) can modulate HSC adaptation to hematopoietic demands during both steady-state and stress hematopoiesis. MKs are often located adjacent to marrow sinusoids. Considering that most HSCs reside close to a marrow vascular sinusoid, as do MKs, the interactions between MKs and vascular endothelial cells are positioned to play important roles in modulating HSC function, and by extrapolation, might be dysregulated in various disease states. In this review, we discuss the interactions between MKs and the vascular niche in both normal and neoplastic hematopoiesis.
Collapse
Affiliation(s)
- Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, United States
- Medical Service, Northport Veterans Affairs (VA) Medical Center, Northport, NY, United States
- *Correspondence: Huichun Zhan,
| | - Kenneth Kaushansky
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, United States
| |
Collapse
|
3
|
Lei Z, Hu X, Wu Y, Fu L, Lai S, Lin J, Li X, Lv Y. The Role and Mechanism of the Vascular Endothelial Niche in Diseases: A Review. Front Physiol 2022; 13:863265. [PMID: 35574466 PMCID: PMC9092213 DOI: 10.3389/fphys.2022.863265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/13/2022] [Indexed: 12/15/2022] Open
Abstract
Vascular endothelial cells, forming the inner wall of the blood vessels, participate in the body’s pathological and physiological processes of immunity, tumors, and infection. In response to an external stimulus or internal pathological changes, vascular endothelial cells can reshape their microenvironment, forming a “niche”. Current research on the vascular endothelial niche is a rapidly growing field in vascular biology. Endothelial niches not only respond to stimulation by external information but are also decisive factors that act on neighboring tissues and circulating cells. Intervention through the vascular niche is meaningful for improving the treatment of several diseases. This review aimed to summarize reported diseases affected by endothelial niches and signal molecular alterations or release within endothelial niches. We look forward to contributing knowledge to increase the understanding the signaling and mechanisms of the vascular endothelial niche in multiple diseases.
Collapse
Affiliation(s)
- Zhiqiang Lei
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Xiang Hu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yaoqi Wu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Longsheng Fu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Songqing Lai
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Lin
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaobing Li
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yanni Lv
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China.,Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
4
|
Ye B, Sheng Y, Zhang M, Hu Y, Huang H. Early detection and intervention of clonal hematopoiesis for preventing hematological malignancies. Cancer Lett 2022; 538:215691. [DOI: 10.1016/j.canlet.2022.215691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/03/2022] [Accepted: 04/17/2022] [Indexed: 12/17/2022]
|
5
|
Owen-Woods C, Kusumbe A. Fundamentals of bone vasculature: Specialization, interactions and functions. Semin Cell Dev Biol 2022; 123:36-47. [PMID: 34281770 DOI: 10.1016/j.semcdb.2021.06.025] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023]
Abstract
Angiogenesis, hematopoiesis and osteogenesis are fundamental processes mediating complex and essential biological functions. In the bone marrow, endothelial cells (ECs) are a principal mediator of regulatory signals that govern hematopoietic and mesenchymal stem cells. EC and osteoblast interactions and niche functions of ECs are fundamental in maintaining bone health and coordinating repair and regeneration following injury. These cellular interactions are subject to dysregulation and deterioration under stress, aging, chronic disease states and malignancy. Thus, the prospect of manipulating the bone vasculature has tremendous potential to advance therapeutic interventions for the management of bone diseases. This review discusses the current state of vascular-skeletal tissue interactions focusing on osteoblast and hematopoietic stem cells interaction with ECs.
Collapse
Affiliation(s)
- Charlotte Owen-Woods
- Tissue and Tumor Microenvironments Group, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Anjali Kusumbe
- Tissue and Tumor Microenvironments Group, NDORMS, University of Oxford, Oxford OX3 7FY, UK.
| |
Collapse
|
6
|
Migliaccio AR. A Novel Megakaryocyte Subpopulation Poised to Exert the Function of HSC Niche as Possible Driver of Myelofibrosis. Cells 2021; 10:3302. [PMID: 34943811 PMCID: PMC8699046 DOI: 10.3390/cells10123302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022] Open
Abstract
Careful morphological investigations, coupled with experimental hematology studies in animal models and in in vitro human cultures, have identified that platelets are released in the circulation by mature megakaryocytes generated by hematopoietic stem cells by giving rise to lineage-restricted progenitor cells and then to morphologically recognizable megakaryocyte precursors, which undergo a process of terminal maturation. Advances in single cell profilings are revolutionizing the process of megakaryocytopoiesis as we have known it up to now. They identify that, in addition to megakaryocytes responsible for producing platelets, hematopoietic stem cells may generate megakaryocytes, which exert either immune functions in the lung or niche functions in organs that undergo tissue repair. Furthermore, it has been discovered that, in addition to hematopoietic stem cells, during ontogeny, and possibly in adult life, megakaryocytes may be generated by a subclass of specialized endothelial precursors. These concepts shed new light on the etiology of myelofibrosis, the most severe of the Philadelphia negative myeloproliferative neoplasms, and possibly other disorders. This perspective will summarize these novel concepts in thrombopoiesis and discuss how they provide a framework to reconciliate some of the puzzling data published so far on the etiology of myelofibrosis and their implications for the therapy of this disease.
Collapse
Affiliation(s)
- Anna Rita Migliaccio
- Department of Medicine and Surgery, Campus Bio-Medico University, 00128 Rome, Italy; or amigliaccio.altius.org
- Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
| |
Collapse
|
7
|
Zhang H, Yeware A, Lee S, Zhan H. A Murine Model With JAK2V617F Expression in Both Hematopoietic Cells and Vascular Endothelial Cells Recapitulates the Key Features of Human Myeloproliferative Neoplasm. Front Oncol 2021; 11:753465. [PMID: 34765558 PMCID: PMC8576565 DOI: 10.3389/fonc.2021.753465] [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: 08/04/2021] [Accepted: 10/07/2021] [Indexed: 11/13/2022] Open
Abstract
The myeloproliferative neoplasms (MPNs) are characterized by an expansion of the neoplastic hematopoietic stem/progenitor cells (HSPC) and an increased risk of cardiovascular complications. The acquired kinase mutation JAK2V617F is present in hematopoietic cells in a majority of patients with MPNs. Vascular endothelial cells (ECs) carrying the JAK2V617F mutation can also be detected in patients with MPNs. In this study, we show that a murine model with both JAK2V617F-bearing hematopoietic cells and JAK2V617F-bearing vascular ECs recapitulated all the key features of the human MPN disease, which include disease transformation from essential thrombocythemia to myelofibrosis, extramedullary splenic hematopoiesis, and spontaneous cardiovascular complications. We also found that, during aging and MPN disease progression, there was a loss of both HSPC number and HSPC function in the marrow while the neoplastic hematopoiesis was relatively maintained in the spleen, mimicking the advanced phases of human MPN disease. Different vascular niche of the marrow and spleen could contribute to the different JAK2V617F mutant stem cell functions we have observed in this JAK2V617F-positive murine model. These results indicate that the spleen is functionally important for the JAK2V617F mutant neoplastic hematopoiesis during aging and MPN disease progression. Compared to other MPN murine models reported so far, our studies demonstrate that JAK2V617F-bearing vascular ECs play an important role in both the hematologic and cardiovascular abnormalities of MPN.
Collapse
Affiliation(s)
- Haotian Zhang
- Graduate Program in Molecular & Cellular Biology, Stony Brook University, Stony Brook, NY, United States
| | - Amar Yeware
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, United States
| | - Sandy Lee
- Graduate Program in Molecular & Cellular Pharmacology, Stony Brook University, Stony Brook, NY, United States
| | - Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, United States.,Medical Service, Northport VA Medical Center, Northport, NY, United States
| |
Collapse
|
8
|
Bone marrow microenvironment of MPN cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021. [PMID: 34756245 DOI: 10.1016/bs.ircmb.2021.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
In this chapter, we will discuss the current knowledge concerning the alterations of the cellular components in the bone marrow niche in Myeloproliferative Neoplasms (MPNs), highlighting the central role of the megakaryocytes in MPN progression, and the extracellular matrix components characterizing the fibrotic bone marrow.
Collapse
|
9
|
Zhang H, Castiglione M, Zheng L, Zhan H. Cell competition between wild-type and JAK2V617F mutant cells prevents disease relapse after stem cell transplantation in a murine model of myeloproliferative neoplasm. Exp Hematol Oncol 2021; 10:47. [PMID: 34666822 PMCID: PMC8524893 DOI: 10.1186/s40164-021-00241-2] [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: 09/25/2021] [Accepted: 10/07/2021] [Indexed: 11/10/2022] Open
Abstract
Disease relapse after allogeneic stem cell transplantation is a major cause of treatment-related morbidity and mortality in patients with myeloproliferative neoplasms (MPNs). The cellular and molecular mechanisms for MPN relapse are not well understood. Here, we established a murine model of MPN relapse, in which ~ 60% of the MPN recipient mice develop disease relapse after receiving stem cell transplantation with wild-type marrow donor. Using this model, we find that impaired wild-type cell function is associated with MPN disease relapse. We also show that competition between wild-type and JAK2V617F mutant cells can modulate the immune cell composition and PD-L1 expression induced by the JAK2V617F oncogene. These results suggest that cell competition between wild-type donor cells and JAK2V617F mutant recipient cells can prevent MPN disease relapse after stem cell transplantation.
Collapse
Affiliation(s)
- Haotian Zhang
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA
| | - Melissa Castiglione
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, USA.,Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lei Zheng
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, USA. .,Medical Service, Northport VA Medical Center, Northport, NY, USA. .,Division of Hematology-Oncology, Department of Medicine, Stony Brook School of Medicine, HSC T15, Room 053, Stony Brook, NY, 11794, USA. .,Northport VA Medical Center, Building 62, Room 124, 79 Middleville Road, Northport, NY, 11768, USA.
| |
Collapse
|
10
|
Castiglione M, Zhang H, Kaushansky K, Zhan H. Cell competition between wild-type and JAK2V617F mutant cells in a murine model of a myeloproliferative neoplasm. Exp Hematol 2021; 100:52-62. [PMID: 34153382 PMCID: PMC9911310 DOI: 10.1016/j.exphem.2021.06.003] [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: 05/27/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 10/21/2022]
Abstract
The myeloproliferative neoplasms (MPNs) are characterized by overproduction of mature blood cells and increased risk of transformation to frank leukemia. The acquired kinase mutation JAK2V617F plays a central role in a majority of patients with these diseases. As MPNs are clonal stem cell disorders (i.e. arise from a single stem cell which eventually expands), the hematopoietic stem/progenitor cell (HSPC) compartment in MPNs is heterogeneous with the presence of both JAK2 wild-type and JAK2V617F mutant cells. Mechanisms responsible for the mutant stem cell expansion in MPNs are not fully understood. Utilizing in vitro co-culture assays and in vivo competitive transplantation assays, we show that the presence of wild-type cells alters both the gene expression profile and cellular function of JAK2V617F mutant HSPCs and inhibits the expansion of co-existing JAK2V617F mutant cells in a normal microenvironment. In contrast, we found that a microenvironment bearing the mutant kinase promotes JAK2V617F mutant HSPC expansion over wild-type cells due in part to altered CXCL12/CXCR4 signaling. Further understanding of the molecular mechanisms controlling the competitive interactions between normal and JAK2V617F mutant cells, and how these mechanisms break down during MPN disease progression hold great potential for advances in treating patients with these diseases.
Collapse
Affiliation(s)
| | - Haotian Zhang
- Graduate Program in Molecular & Cellular Biology, Stony Brook University, Stony Brook, NY
| | - Kenneth Kaushansky
- Office of the Sr. Vice President, Health Sciences, Stony Brook School of Medicine, Stony Brook, NY
| | - Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY; Medical Service, Northport VA Medical Center, Northport, NY.
| |
Collapse
|
11
|
Mazzeo C, Quan M, Wong H, Castiglione M, Kaushansky K, Zhan H. JAK2V617F mutant endothelial cells promote neoplastic hematopoiesis in a mixed vascular microenvironment. Blood Cells Mol Dis 2021; 90:102585. [PMID: 34139651 DOI: 10.1016/j.bcmd.2021.102585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/24/2021] [Accepted: 05/24/2021] [Indexed: 11/19/2022]
Abstract
The chronic myeloproliferative neoplasms (MPNs) are clonal stem cell disorders. The hematopoietic stem/progenitor cell (HSPC) compartment in patients with MPNs is heterogeneous with the presence of both wild-type and JAK2V617F mutant cells. Mechanisms responsible for mutant stem cell expansion in MPNs are not fully understood. Vascular endothelial cells (ECs) are an essential component of the hematopoietic microenvironment. ECs carrying the JAK2V617F mutation can be detected in patients with MPNs. Utilizing an ex vivo EC-HSPC co-culture system with mixed wild-type and JAK2V617F mutant ECs, we show that even small numbers of JAK2V617F mutant ECs can promote the expansion of JAK2V617F mutant HSPCs in preference to wild-type HSPCs during irradiation or cytotoxic chemotherapy, the two treatments commonly used in the conditioning regimen for stem cell transplantation, the only curative treatment for patients with MPNs. Mechanistically, we found that both cell-cell interactions and secreted factors are important for JAK2V617F mutant EC-mediated neoplastic hematopoiesis. Further understanding of how the JAK2V617F mutation alters vascular niche function will help identify new strategies to not only control neoplastic cell expansion but also prevent disease relapse in patients with MPNs.
Collapse
Affiliation(s)
| | - Moqing Quan
- Massachusetts General Hospital, Boston, MA, USA
| | - Helen Wong
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY, USA
| | | | - Kenneth Kaushansky
- Office of the Sr. Vice President, Health Sciences, Stony Brook School of Medicine, Stony Brook, NY, USA
| | - Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, USA; Medical Service, Northport VA Medical Center, Northport, NY, USA.
| |
Collapse
|
12
|
Guijarro-Hernández A, Vizmanos JL. A Broad Overview of Signaling in Ph-Negative Classic Myeloproliferative Neoplasms. Cancers (Basel) 2021; 13:cancers13050984. [PMID: 33652860 PMCID: PMC7956519 DOI: 10.3390/cancers13050984] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary There is growing evidence that Ph-negative myeloproliferative neoplasms are disorders in which multiple signaling pathways are significantly disturbed. The heterogeneous phenotypes observed among patients have highlighted the importance of having a comprehensive knowledge of the molecular mechanisms behind these diseases. This review aims to show a broad overview of the signaling involved in myeloproliferative neoplasms (MPNs) and other processes that can modify them, which could be helpful to better understand these diseases and develop more effective targeted treatments. Abstract Ph-negative myeloproliferative neoplasms (polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF)) are infrequent blood cancers characterized by signaling aberrations. Shortly after the discovery of the somatic mutations in JAK2, MPL, and CALR that cause these diseases, researchers extensively studied the aberrant functions of their mutant products. In all three cases, the main pathogenic mechanism appears to be the constitutive activation of JAK2/STAT signaling and JAK2-related pathways (MAPK/ERK, PI3K/AKT). However, some other non-canonical aberrant mechanisms derived from mutant JAK2 and CALR have also been described. Moreover, additional somatic mutations have been identified in other genes that affect epigenetic regulation, tumor suppression, transcription regulation, splicing and other signaling pathways, leading to the modification of some disease features and adding a layer of complexity to their molecular pathogenesis. All of these factors have highlighted the wide variety of cellular processes and pathways involved in the pathogenesis of MPNs. This review presents an overview of the complex signaling behind these diseases which could explain, at least in part, their phenotypic heterogeneity.
Collapse
Affiliation(s)
- Ana Guijarro-Hernández
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain;
| | - José Luis Vizmanos
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain;
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Correspondence:
| |
Collapse
|
13
|
Zhan H, Kaushansky K. The Hematopoietic Microenvironment in Myeloproliferative Neoplasms: The Interplay Between Nature (Stem Cells) and Nurture (the Niche). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1273:135-145. [PMID: 33119879 DOI: 10.1007/978-3-030-49270-0_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Hematopoietic stem cells (HSCs) rely on instructive cues from the marrow microenvironment for their maintenance and function. Accumulating evidence indicates that the survival and proliferation of hematopoietic neoplasms are dependent not only on cell-intrinsic, genetic mutations, and other molecular alterations present within neoplastic stem cells, but also on the ability of the surrounding microenvironmental cells to nurture and promote the malignancy. It is anticipated that a better understanding of the molecular and cellular events responsible for these microenvironmental features of neoplastic hematopoiesis will lead to improved treatment for patients. This review will focus on the myeloproliferative neoplasms (MPNs), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), in which an acquired signaling kinase mutation (JAK2V617F) plays a central, pathogenetic role in 50-100% of patients with these disorders. Evidence is presented that the development of an MPN requires both an abnormal, mutation-bearing (i.e., neoplastic) HSC and an abnormal, mutation-bearing microenvironment.
Collapse
Affiliation(s)
- Huichun Zhan
- Division of Hematology-Oncology, Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, USA. .,Northport VA Medical Center, Northport, NY, USA.
| | | |
Collapse
|
14
|
Castiglione M, Jiang Y, Mazzeo C, Lee S, Chen J, Kaushansky K, Yin W, Lin RZ, Zheng H, Zhan H. Endothelial JAK2V617F mutation leads to thrombosis, vasculopathy, and cardiomyopathy in a murine model of myeloproliferative neoplasm. J Thromb Haemost 2020; 18:3359-3370. [PMID: 32920974 PMCID: PMC7756295 DOI: 10.1111/jth.15095] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/05/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Cardiovascular complications are the leading cause of morbidity and mortality in patients with myeloproliferative neoplasms (MPNs). The acquired kinase mutation JAK2V617F plays a central role in these disorders. Mechanisms responsible for cardiovascular dysfunction in MPNs are not fully understood, limiting the effectiveness of current treatment. Vascular endothelial cells (ECs) carrying the JAK2V617F mutation can be detected in patients with MPNs. The goal of this study was to test the hypothesis that the JAK2V617F mutation alters endothelial function to promote cardiovascular complications in patients with MPNs. APPROACH AND RESULTS We employed murine models of MPN in which the JAK2V617F mutation is expressed in specific cell lineages. When JAK2V617F is expressed in both blood cells and vascular ECs, the mice developed MPN and spontaneous, age-related dilated cardiomyopathy with an increased risk of sudden death as well as a prothrombotic and vasculopathy phenotype on histology evaluation. In contrast, despite having significantly higher leukocyte and platelet counts than controls, mice with JAK2V617F-mutant blood cells alone did not demonstrate any cardiac dysfunction, suggesting that JAK2V617F-mutant ECs are required for this cardiovascular disease phenotype. Furthermore, we demonstrated that the JAK2V617F mutation promotes a pro-adhesive, pro-inflammatory, and vasculopathy EC phenotype, and mutant ECs respond to flow shear differently than wild-type ECs. CONCLUSIONS These findings suggest that the JAK2V617F mutation can alter vascular endothelial function to promote cardiovascular complications in MPNs. Therefore, targeting the MPN vasculature represents a promising new therapeutic strategy for patients with MPNs.
Collapse
Affiliation(s)
| | - Ya‐Ping Jiang
- Department of Physiology and BiophysicsInstitute of Molecular CardiologyStony Brook UniversityStony BrookNYUSA
| | | | - Sandy Lee
- Department of Molecular and Cellular PharmacologyStony Brook UniversityStony BrookNYUSA
| | - Juei‐Suei Chen
- Department of MedicineStony Brook School of MedicineStony BrookNYUSA
| | - Kenneth Kaushansky
- Office of the Sr. Vice PresidentHealth SciencesStony Brook MedicineStony BrookNYUSA
| | - Wei Yin
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNYUSA
| | - Richard Z. Lin
- Department of Physiology and BiophysicsInstitute of Molecular CardiologyStony Brook UniversityStony BrookNYUSA
- Medical ServiceNorthport VA Medical CenterNorthportNYUSA
| | - Haoyi Zheng
- Cardiac ImagingThe Heart CenterSaint Francis HospitalRoslynNYUSA
| | - Huichun Zhan
- Department of MedicineStony Brook School of MedicineStony BrookNYUSA
- Medical ServiceNorthport VA Medical CenterNorthportNYUSA
| |
Collapse
|
15
|
Testa U, Pelosi E, Castelli G. Endothelial Progenitors in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1263:85-115. [PMID: 32588325 DOI: 10.1007/978-3-030-44518-8_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor vascularization refers to the formation of new blood vessels within a tumor and is considered one of the hallmarks of cancer. Tumor vessels supply the tumor with oxygen and nutrients, required to sustain tumor growth and progression, and provide a gateway for tumor metastasis through the blood or lymphatic vasculature. Blood vessels display an angiocrine capacity of supporting the survival and proliferation of tumor cells through the production of growth factors and cytokines. Although tumor vasculature plays an essential role in sustaining tumor growth, it represents at the same time an essential way to deliver drugs and immune cells to the tumor. However, tumor vasculature exhibits many morphological and functional abnormalities, thus resulting in the formation of hypoxic areas within tumors, believed to represent a mechanism to maintain tumor cells in an invasive state.Tumors are vascularized through a variety of modalities, mainly represented by angiogenesis, where VEGF and other members of the VEGF family play a key role. This has represented the basis for the development of anti-VEGF blocking agents and their use in cancer therapy: however, these agents failed to induce significant therapeutic effects.Much less is known about the cellular origin of vessel network in tumors. Various cell types may contribute to tumor vasculature in different tumors or in the same tumor, such as mature endothelial cells, endothelial progenitor cells (EPCs), or the same tumor cells through a process of transdifferentiation. Early studies have suggested a role for bone marrow-derived EPCs; these cells do not are true EPCs but myeloid progenitors differentiating into monocytic cells, exerting a proangiogenic effect through a paracrine mechanism. More recent studies have shown the existence of tissue-resident endothelial vascular progenitors (EVPs) present at the level of vessel endothelium and their possible involvement as cells of origin of tumor vasculature.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy.
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
16
|
Role of microvascular endothelial cells on proliferation, migration and adhesion of hematopoietic stem cells. Biosci Rep 2020; 40:222324. [PMID: 32154555 PMCID: PMC7087325 DOI: 10.1042/bsr20192104] [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: 07/04/2019] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 11/30/2022] Open
Abstract
Background: The present study investigated the effects of microvascular endothelial cells (MECs) on the chemotaxis, adhesion and proliferation of bone marrow hematopoietic stem cells (HSCs) ex vivo. Methods and Results: MECs were collected from the lung tissue of C57BL/6 mice, and HSCs were isolated with immunomagnetic beads from bone marrow of GFP mice. MECs and HSCs were co-cultured with or without having direct cell–cell contact in Transwell device for the measurement of chemotaxis and adhesion of MECs to HSCs. Experimental results indicate that the penetration rate of HSCs from the Transwell upper chamber to lower chamber in ‘co-culture’ group was significantly higher than that of ‘HSC single culture’ group. Also, the HSCs in co-culture group were all adherent at 24 h, and the co-culture group with direct cell–cell contact had highest proliferation rate. The HSC number was positively correlated with vascular endothelial growth factor (VEGF) and stromal cell-derived factor-1 (SDF-1) levels in supernatants of the culture. Conclusions: Our study reports that MECs enhance the chemotaxis, adhesion and proliferation of HSCs, which might be related to cytokines SDF-1 and VEGF secreted by MECs, and thus MECs enhance the HSC proliferation through cell–cell contact. The present study revealed the effect of MECs on HSCs, and provided a basis and direction for effective expansion of HSCs ex vivo.
Collapse
|
17
|
Kaushansky K, Zhan H. The marrow stem cell niche in normal and malignant hematopoiesis. Ann N Y Acad Sci 2019; 1466:17-23. [PMID: 30767234 DOI: 10.1111/nyas.14028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/09/2019] [Accepted: 01/23/2019] [Indexed: 02/06/2023]
Abstract
The hematopoietic niche is composed of endothelial cells, mesenchymal stromal cells of several types, and megakaryocytes, and functions to support the survival, proliferation, and differentiation of normal hematopoietic stem cells (HSCs). An abundance of evidence from a range of hematological malignancies supports the concept that the niche also participates in the pathogenesis of malignant hematopoiesis, differentially supporting malignant stem or progenitor cells over that of normal blood cell development. In 2005, patients with myeloproliferative neoplasms were reported to harbor an acquired, activating, missense V617F mutation of the cytokine-signaling Janus kinase (JAK)-2, JAK2V617F , present in virtually all patients with polycythemia vera and half of patients with essential thrombocythemia and primary myelofibrosis. Using both in vitro and in vivo methods, several investigators have shown that in addition to driving cytokine-independent proliferation in HSCs, JAK2V617F contributes to these neoplasms by altering the hematopoietic niche. The role of both endothelial cells and megakaryocytes bearing JAK2V617F will be presented, which involves altering cytokine production within the niche, resulting in their differential support of mutant kinase-bearing stem cells over their normal counterparts, and imparting relative radiation resistance to stem cells. The clinical correlates of these findings will be discussed, as will their therapeutic implications.
Collapse
Affiliation(s)
| | - Huichun Zhan
- Stony Brook University School of Medicine, Stony Brook, New York
| |
Collapse
|
18
|
Zhan H, Kaushansky K. Functional interdependence of hematopoietic stem cells and their niche in oncogene promotion of myeloproliferative neoplasms: the 159th biomedical version of "it takes two to tango". Exp Hematol 2018; 70:24-30. [PMID: 30593829 DOI: 10.1016/j.exphem.2018.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/13/2018] [Accepted: 12/20/2018] [Indexed: 01/24/2023]
Abstract
The role of stem cells in normal and neoplastic hematopoiesis is well established. However, neither normal nor neoplastic hematopoietic stem cells (HSCs) develop in isolation and accumulating evidence indicates that a critical developmental role is played by the perivascular "niche." The cellular, humoral, and cell surface contacts that provide the proper environment for HSC survival, proliferation, and differentiation are becoming increasingly better understood. A number of studies have established that endothelial cells (ECs), several types of perivascular stromal cells, and megakaryocytes (MKs) provide several cell surface and secreted molecules required for HSC development. Accumulating evidence also indicates that the normal stem cell niche is altered in patients with hematological neoplasms and that the "neoplastic niche" plays an important role in promoting malignant and suppressing normal blood cell development in such patients. To explore this concept in the myeloproliferative neoplasms (MPNs), we employed a murine model to determine the effects of Jak2V617F, an oncogene found in a majority of such patients, in marrow ECs and MKs and their effect on promoting neoplastic and suppressing normal hematopoiesis. We found that Jak2V617F has profound effects on both cell types, which together are critical for the growth advantage and radioresistance shown by Jak2V617F-bearing HSCs. Such findings should provide new approaches to the treatment of patients with MPNs.
Collapse
Affiliation(s)
- Huichun Zhan
- School of Medicine, Stony Brook University, Stony Brook, New York
| | | |
Collapse
|
19
|
O'Sullivan J, Mead AJ. Heterogeneity in myeloproliferative neoplasms: Causes and consequences. Adv Biol Regul 2018; 71:55-68. [PMID: 30528537 DOI: 10.1016/j.jbior.2018.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 01/09/2023]
Abstract
Myeloproliferative neoplasms (MPNs) are haematopoietic stem cell-derived clonal disorders characterised by proliferation of some or all myeloid lineages, depending on the subtype. MPNs are classically categorized into three disease subgroups; essential thrombocythaemia (ET), polycythaemia vera (PV) and primary myelofibrosis (PMF). The majority (>85%) of patients carry a disease-initiating or driver mutation, the most prevalent occurring in the janus kinase 2 gene (JAK2 V617F), followed by calreticulin (CALR) and myeloproliferative leukaemia virus (MPL) genes. Although these diseases are characterised by shared clinical, pathological and molecular features, one of the most challenging aspects of these disorders is the diverse clinical features which occur in each disease type, with marked variability in risks of disease complications and progression to leukaemia. A remarkable aspect of MPN biology is that the JAK2 V617F mutation, often occurring in the absence of additional mutations, generates a spectrum of phenotypes from asymptomatic ET through to aggressive MF, associated with a poor outcome. The mechanisms promoting MPN heterogeneity remain incompletely understood, but contributing factors are broad and include patient characteristics (gender, age, comorbidities and environmental exposures), additional somatic mutations, target disease-initiating cell, bone marrow microenvironment and germline genetic associations. In this review, we will address these in detail and discuss their role in heterogeneity of MPN disease phenotypes. Tailoring patient management according to the multiple different factors that influence disease phenotype may prove to be the most effective approach to modify the natural history of the disease and ultimately improve outcomes for patients.
Collapse
Affiliation(s)
- Jennifer O'Sullivan
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, United Kingdom.
| | - Adam J Mead
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, United Kingdom; NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
20
|
Kaushansky K, Zhan H. The regulation of normal and neoplastic hematopoiesis is dependent on microenvironmental cells. Adv Biol Regul 2018; 69:11-15. [PMID: 29970351 DOI: 10.1016/j.jbior.2018.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 06/20/2018] [Accepted: 06/26/2018] [Indexed: 01/10/2023]
Abstract
Each day the adult human produces 4 × 1011 red blood cells, 1 × 1011 white blood cells and 1 × 1011 platelets, levels of production which can increase 10-20 fold in times of heightened demand. Hematopoiesis, or the formation of the ten different types of blood and marrow cells, is a complex process involving hematopoietic stem cells (HSCs), cytokine growth factors and cell surface adhesion molecules, and both specific and ubiquitous transcription factors. The marrow micro-environmental niche is defined as the site at which HSCs reside and are nurtured, receiving the signals that lead to their survival, replication and/or differentiation. Using microscopic, biochemical and molecular methods many different cells and the signals responsible for niche function have been identified. Early studies suggested two distinct anatomical sites for the niche, perivascular and periosteal, but the preponderance of evidence now favors the former. Within the "vascular niche" much evidence exists for important contributions by vascular endothelial cells (ECs), CXCL12-abundant reticular (CAR) cells and mesenchymal stromal cells, through their elaboration of chemokines, cytokines and cell surface adhesion molecules. In a series of studies we have found, and will present the evidence that megakaryocytes (MKs), the precursors of blood platelets, must be added to this list. In addition to normal blood cell development, numerous studies have implicated the perivascular niche as contributing to the pathogenesis of a variety of hematological malignancies. Our laboratory focuses on the Ph (Crane et al., 2017)-negative myeloproliferative neoplasms (MPNs), polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF). These diseases are characterized by clonal expansion of HSCs and one or more mature blood cell types, hypermetabolism, a propensity to disorders of hemostasis (thrombosis > bleeding) and in some, evolution to acute leukemia. While a variety of therapies can control the abnormal expansion of the progeny of the malignant HSC, the only curative therapy is myeloablation with conditioning therapy or immunological means, followed by allogeneic stem cell transplantation (SCT), a procedure that is often inadequate due to relapse of the malignant clone. While the three disorders were postulated by Dameshek in the 1950s to be related to one another, proof came in 2005 when an acquired mutation in the signaling kinase Janus kinase 2 (Jak2V617F) was identified in virtually all patients with PV, and ∼50% of patients with ET and PMF. Since that time a number of other mutations have been identified that account for the "Jak2V617F negative" MPNs, including the thrombopoietin receptor, c-MPL, other mutations of Jak2, calreticulin and a variety of epigenetic modifier genes (e.g. TET2). Using a cell-specific Cre recombinase and SCT techniques we can introduce Jak2V617F into murine megakaryocytes and platelets, hematopoietic stem cells, and endothelial cells, alone or in combination, in order to probe the role of the mutant kinase in various cells on several aspects of the MPNs. Using these tools we have found that the expression of Jak2V617F in HSCs and ECs drives a MPN characterized by neutrophilia, thrombocytosis and splenomegaly, eventually evolving into myelosclerosis. Somewhat surprisingly, we found that Jak2V617F-bearing ECs were required for many features of the MPN, such as enhancing the growth of Jak2V617F-bearing HSCs over that of wild type HSCs, its characteristic radioresistance, and a hemostatic defect. Altogether, our studies suggest that the malignant vascular niche is a critical element in the pathogenesis of MPNs, and a more thorough understanding of the molecular basis for these findings could lead to improved treatment for patients with these disorders.
Collapse
Affiliation(s)
| | - Huichun Zhan
- Stony Brook University School of Medicine, Stony Brook, NY, USA
| |
Collapse
|