Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Biomolecules ; 14(4)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38672505

ABSTRACT

Thrombopoietin, the primary regulator of blood platelet production, was postulated to exist in 1958, but was only proven to exist when the cDNA for the hormone was cloned in 1994. Since its initial cloning and characterization, the hormone has revealed many surprises. For example, instead of acting as the postulated differentiation factor for platelet precursors, megakaryocytes, it is the most potent stimulator of megakaryocyte progenitor expansion known. Moreover, it also stimulates the survival, and in combination with stem cell factor leads to the expansion of hematopoietic stem cells. All of these growth-promoting activities have resulted in its clinical use in patients with thrombocytopenia and aplastic anemia, although the clinical development of the native molecule illustrated that "it's not wise to mess with mother nature", as a highly engineered version of the native hormone led to autoantibody formation and severe thrombocytopenia. Finally, another unexpected finding was the role of the thrombopoietin receptor in stem cell biology, including the development of myeloproliferative neoplasms, an important disorder of hematopoietic stem cells. Overall, the past 30 years of clinical and basic research has yielded many important insights, which are reviewed in this paper.


Subject(s)
Blood Platelets , Thrombopoietin , Thrombopoietin/metabolism , Humans , Blood Platelets/metabolism , Animals , Receptors, Thrombopoietin/metabolism , Receptors, Thrombopoietin/genetics , Thrombopoiesis , Thrombocytopenia/metabolism , Megakaryocytes/metabolism , Megakaryocytes/cytology
2.
Front Oncol ; 12: 912060, 2022.
Article in English | MEDLINE | ID: mdl-35814384

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.

3.
Stem Cells ; 40(4): 359-370, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35260895

ABSTRACT

Megakaryocytes (MKs) is an important component of the hematopoietic niche. Abnormal MK hyperplasia is a hallmark feature of myeloproliferative neoplasms (MPNs). The JAK2V617F mutation is present in hematopoietic cells in a majority of patients with MPNs. Using a murine model of MPN in which the human JAK2V617F gene is expressed in the MK lineage, we show that the JAK2V617F-bearing MKs promote hematopoietic stem cell (HSC) aging, manifesting as myeloid-skewed hematopoiesis with an expansion of CD41+ HSCs, a reduced engraftment and self-renewal capacity, and a reduced differentiation capacity. HSCs from 2-year-old mice with JAK2V617F-bearing MKs were more proliferative and less quiescent than HSCs from age-matched control mice. Examination of the marrow hematopoietic niche reveals that the JAK2V617F-bearing MKs not only have decreased direct interactions with hematopoietic stem/progenitor cells during aging but also suppress the vascular niche function during aging. Unbiased RNA expression profiling reveals that HSC aging has a profound effect on MK transcriptomic profiles, while targeted cytokine array shows that the JAK2V617F-bearing MKs can alter the hematopoietic niche through increased levels of pro-inflammatory and anti-angiogenic factors. Therefore, as a hematopoietic niche cell, MKs represent an important connection between the extrinsic and intrinsic mechanisms for HSC aging.


Subject(s)
Myeloproliferative Disorders , Neoplasms , Aging/genetics , Animals , Disease Models, Animal , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Megakaryocytes/metabolism , Mice , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism
5.
Blood Cells Mol Dis ; 90: 102585, 2021 09.
Article in English | MEDLINE | ID: mdl-34139651

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.


Subject(s)
Endothelial Cells/enzymology , Hematologic Neoplasms , Hematopoiesis , Hematopoietic Stem Cells/enzymology , Janus Kinase 2 , Mutation, Missense , Myeloproliferative Disorders , Neoplastic Stem Cells/enzymology , Tumor Microenvironment , Amino Acid Substitution , Animals , Hematologic Neoplasms/enzymology , Hematologic Neoplasms/genetics , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Mice, Transgenic , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/genetics
6.
Front Med (Lausanne) ; 8: 631335, 2021.
Article in English | MEDLINE | ID: mdl-33634153

ABSTRACT

Objective: Examine the possible beneficial effects of early, D-dimer driven anticoagulation in preventing thrombotic complications and improving the overall outcomes of COVID-19 intubated patients. Methods: To address COVID-19 hypercoagulability, we developed a clinical protocol to escalate anticoagulation based on serum D-dimer levels. We retrospectively reviewed all our first 240 intubated patients with COVID-19. Of the 240, 195 were stratified into patients treated based on this protocol (ON-protocol, n = 91) and the control group, patients who received standard thromboprophylaxis (OFF-protocol, n = 104). All patients were admitted to the Stony Brook University Hospital intensive care units (ICUs) between February 7th, 2020 and May 17, 2020 and were otherwise treated in the same manner for all aspects of COVID-19 disease. Results: We found that the overall mortality was significantly lower ON-protocol compared to OFF-protocol (27.47 vs. 58.66%, P < 0.001). Average maximum D-dimer levels were significantly lower in the ON-protocol group (7,553 vs. 12,343 ng/mL), as was serum creatinine (2.2 vs. 2.8 mg/dL). Patients with poorly controlled D-dimer levels had higher rates of kidney dysfunction and mortality. Transfusion requirements and serious bleeding events were similar between groups. To address any possible between-group differences, we performed a propensity-matched analysis of 124 of the subjects (62 matched pairs, ON-protocol and OFF-protocol), which showed similar findings (31 vs. 57% overall mortality in the ON-protocol and OFF-protocol group, respectively). Conclusions: D-dimer-driven anticoagulation appears to be safe in patients with COVID-19 infection and is associated with improved survival. What This Paper Adds: It has been shown that hypercoagulability in patients with severe COVID-19 infection leads to thromboembolic complications and organ dysfunction. Anticoagulation has been variably administered to these patients, but it is unknown whether routine or escalated thromboprophylaxis provides a survival benefit. Our data shows that escalated D-dimer driven anticoagulation is associated with improved organ function and overall survival in intubated COVID-19 ICU patients at our institution. Importantly, we found that timely escalation of this anticoagulation is critical in preventing organ dysfunction and mortality in patients with severe COVID-19 infection.

7.
Ann Biomed Eng ; 49(3): 959-963, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33469819

ABSTRACT

Since the first appearance of the severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) earlier this year, clinicians and researchers alike have been faced with dynamic, daily challenges of recognizing, understanding, and treating the coronavirus disease 2019 (COVID-19) due to SARS-CoV-2. Those who are moderately to severely ill with COVID-19 are likely to develop acute hypoxemic respiratory failure and require administration of supplemental oxygen. Assessing the need to initiate or titrate oxygen therapy is largely dependent on evaluating the patient's existing blood oxygenation status, either by direct arterial blood sampling or by transcutaneous arterial oxygen saturation monitoring, also referred to as pulse oximetry. While the sampling of arterial blood for measurement of dissolved gases provides a direct measurement, it is technically challenging to obtain, is painful to the patient, and can be time and resource intensive. Pulse oximetry allows for non-invasive, real-time, continuous monitoring of the percent of hemoglobin molecules that are saturated with oxygen, and usually closely predicts the arterial oxygen content. As such, it was particularly concerning when patients with severe COVID-19 requiring endotracheal intubation and mechanical ventilation within one of our intensive care units were observed to have significant discordance between their predicted arterial oxygen content via pulse oximetry and their actual measured oxygen content. We offer these preliminary observations along with our speculative causes as a timely, urgent clinical need. In the setting of a COVID-19 intensive care unit, entering a patient room to obtain a fresh arterial blood gas sample not only takes exponentially longer to do given the time required for donning and doffing of personal protective equipment (PPE), it involves the consumption of already sparce PPE, and it increases the risk of viral exposure to the nurse, physician, or respiratory therapist entering the room to obtain the sample. As such, technology similar to pulse oximetry which can be applied to a patients finger, and then continuously monitored from outside the room is essential in preventing a particularly dangerous situation of unrealized hypoxia in this critically-ill patient population. Additionally, it would appear that conventional two-wavelength pulse oximetry may not accurately predict the arterial oxygen content of blood in these patients. This discordance of oxygenation measurements poses a critical concern in the evaluation and management of the acute hypoxemic respiratory failure seen in patients with COVID-19.


Subject(s)
Blood Gas Analysis/methods , COVID-19/blood , COVID-19/therapy , Oxygen/blood , Respiration, Artificial , Humans , Intubation, Intratracheal , Oximetry
8.
Adv Exp Med Biol ; 1273: 135-145, 2020.
Article in English | MEDLINE | ID: mdl-33119879

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.


Subject(s)
Hematopoietic Stem Cells/cytology , Myeloproliferative Disorders/pathology , Neoplasms/pathology , Stem Cell Niche , Tumor Microenvironment , Humans , Janus Kinase 2/genetics , Mutation , Myeloproliferative Disorders/genetics , Neoplasms/genetics
9.
J Thromb Haemost ; 18(12): 3359-3370, 2020 12.
Article in English | MEDLINE | ID: mdl-32920974

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.


Subject(s)
Cardiomyopathies , Myeloproliferative Disorders , Neoplasms , Thrombosis , Animals , Disease Models, Animal , Endothelial Cells , Humans , Janus Kinase 2/genetics , Mice , Mutation , Myeloproliferative Disorders/genetics , Thrombosis/genetics
10.
Ann N Y Acad Sci ; 1466(1): 17-23, 2020 04.
Article in English | MEDLINE | ID: mdl-30767234

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.


Subject(s)
Bone Marrow/pathology , Bone Marrow/physiology , Hematologic Neoplasms/pathology , Stem Cell Niche/physiology , Animals , Cellular Microenvironment/physiology , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Humans , Janus Kinase 2/genetics , Mutation/physiology , Tumor Microenvironment/physiology
11.
Exp Hematol ; 70: 24-30, 2019 02.
Article in English | MEDLINE | ID: mdl-30593829

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.


Subject(s)
Hematologic Neoplasms , Myeloproliferative Disorders , Neoplasms, Experimental , Neoplastic Stem Cells , Stem Cell Niche/genetics , Tumor Microenvironment/genetics , Amino Acid Substitution , Animals , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Mutation, Missense , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
13.
Adv Biol Regul ; 69: 11-15, 2018 08.
Article in English | MEDLINE | ID: mdl-29970351

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.


Subject(s)
Endothelial Cells/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Endothelial Cells/cytology , Hematologic Neoplasms , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism
14.
Stem Cells ; 36(11): 1676-1684, 2018 11.
Article in English | MEDLINE | ID: mdl-30005133

ABSTRACT

The myeloproliferative neoplasms (MPNs) are stem cell disorders characterized by hematopoietic stem/progenitor cell (HSPC) expansion and overproduction of mature blood cells. The acquired kinase mutation JAK2V617F plays a central role in these disorders. The mechanisms responsible for HSPC expansion in MPNs are not fully understood, limiting the effectiveness of current treatments. One hallmark feature of the marrow in patients with MPNs is megakaryocyte (MK) hyperplasia. Previously, we reported that JAK2V617F-bearing MKs cause a murine myeloproliferative syndrome with HSPC expansion. Here we show that JAK2V617F MKs promote MPN stem cell function by inducing HSPC quiescence with increased repopulating capacity. In addition, we demonstrate that thrombopoietin and its receptor MPL are critical for the JAK2V617F-bearing MK-induced myeloproliferation, both by directly affecting the quantity and quality of MKs and by altering the MK-endothelial interaction and vascular niche function. Therefore, targeting HSPC niche-forming MKs and/or their interactions within the vascular niche could provide novel, more effective therapeutic strategies in patients with MPNs. Stem Cells 2018;36:1676-1684.


Subject(s)
Hematopoietic Stem Cells/enzymology , Janus Kinase 2/metabolism , Megakaryocytes/metabolism , Myeloproliferative Disorders/metabolism , Receptors, Thrombopoietin/metabolism , Thrombopoietin/metabolism , Animals , Cell Proliferation/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Janus Kinase 2/genetics , Megakaryocytes/enzymology , Megakaryocytes/pathology , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Signal Transduction , Thrombopoietin/genetics
15.
Haematologica ; 103(7): 1160-1168, 2018 07.
Article in English | MEDLINE | ID: mdl-29567773

ABSTRACT

Myeloproliferative neoplasms are clonal stem cell disorders characterized by hematopoietic stem/progenitor cell expansion. The acquired kinase mutation JAK2V617F plays a central role in these disorders. Abnormalities of the marrow microenvironment are beginning to be recognized as an important factor in the development of myeloproliferative neoplasms. Endothelial cells are an essential component of the hematopoietic vascular niche. Endothelial cells carrying the JAK2V617F mutation can be detected in patients with myeloproliferative neoplasms, suggesting that the mutant vascular niche is involved in the pathogenesis of these disorders. Here, using a transgenic mouse expressing JAK2V617F specifically in all hematopoietic cells (including hematopoietic stem/progenitor cells) and endothelial cells, we show that the JAK2V617F-mutant hematopoietic stem/progenitor cells are relatively protected by the JAK2V617F-bearing vascular niche from an otherwise lethal dose of irradiation during conditioning for stem cell transplantation. Gene expression analysis revealed that chemokine (C-X-C motif) ligand 12, epidermal growth factor, and pleiotrophin are up-regulated in irradiated JAK2V617F-bearing endothelial cells compared to wild-type cells. Our findings suggest that the mutant vascular niche may contribute to the high incidence of disease relapse in patients with myeloproliferative neoplasms following allogeneic stem cell transplantation, the only curative treatment for these disorders.


Subject(s)
Amino Acid Substitution , Hematologic Neoplasms/etiology , Hematologic Neoplasms/metabolism , Hematopoietic Stem Cells/metabolism , Janus Kinase 2/genetics , Mutation , Radiation Injuries/complications , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Dose-Response Relationship, Radiation , Endothelial Cells/metabolism , Gene Expression , Hematologic Neoplasms/pathology , Hematopoiesis/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/radiation effects , Mice , Mice, Transgenic , Radiation Tolerance/genetics , Regeneration , Tumor Microenvironment/genetics , Tumor Microenvironment/radiation effects
16.
Blood Cells Mol Dis ; 62: 42-48, 2016 11.
Article in English | MEDLINE | ID: mdl-27865175

ABSTRACT

The myeloproliferative neoplasms (MPNs) are characterized by hematopoietic stem/progenitor cell (HSPC) expansion and overproduction of blood cells. The acquired mutation JAK2V617F plays a central role in these disorders. Mechanisms responsible for MPN HSPC expansion is not fully understood, limiting the effectiveness of current treatments. Endothelial cells (ECs) carrying the JAK2V617F mutation can be detected in patients with MPNs, suggesting that ECs are involved in the pathogenesis of MPNs. Here we report that JAK2V617F-bearing primary murine ECs have increased cell proliferation and angiogenesis in vitro compared to JAK2WT ECs. While there was no difference between JAK2V617F and JAK2WT HSPC proliferation when co-cultured with JAK2WT EC, the JAK2V617F HSPC displayed a relative growth advantage over the JAK2WT HSPC when co-cultured on JAK2V617F EC. In addition, the thrombopoietin (TPO) receptor MPL is up regulated in JAK2V617F ECs and contributes to the maintenance/expansion of the JAK2V617F clone over JAK2WT clone in vitro. Considering that ECs are an essential component of the hematopoietic niche and most HSPCs reside in the perivascular niche, our studies suggest that the JAK2V617F-bearing ECs form an important component of the MPN vascular niche and contribute to mutant stem/progenitor cell expansion, likely through a critical role of the TPO/MPL signaling axis.


Subject(s)
Cell Proliferation/genetics , Janus Kinase 2/genetics , Myeloproliferative Disorders/genetics , Receptors, Thrombopoietin/physiology , Animals , Coculture Techniques , Endothelial Cells/pathology , Humans , Mice , Mutation, Missense , Myeloproliferative Disorders/pathology , Neoplasms/genetics , Neovascularization, Pathologic , Receptors, Thrombopoietin/metabolism , Stem Cell Niche
17.
Thromb J ; 14(Suppl 1): 40, 2016.
Article in English | MEDLINE | ID: mdl-27766065

ABSTRACT

Thrombopoietin was posited to exist in 1958 and cloned in 1994, and in the ensuing two decades we have learned a great deal about the physiology and pathology of the primary regulator of thrombopoiesis. This paper will review the role of the hormone and its receptor, the product of the c-Mpl proto-oncogene, in health and disease, including many unexpected effects in both normal and neoplastic hematopoiesis. Amongst these unexpected properties are a non-redundant effect on hematopoietic stem cells, a critical role in all three of the acquired, chronic myeloproliferative neoplasms, as well as both gain-of-function and loss-of-function mutations in congenital and acquired states of thrombocytopenia and thrombocythemia.

19.
Exp Hematol ; 44(8): 664-8, 2016 08.
Article in English | MEDLINE | ID: mdl-26773569

ABSTRACT

Although the synergy between erythropoietin and thrombopoietin has previously been pointed out, the clonal demonstration of a human bipotent erythroid/megakaryocytic progenitor (MEP) was first published in Experimental Hematology (Papayannopoulou T, Brice M, Farrer D, Kaushansky K. Exp Hematol. 1996;24:660-669) and later in the same year in Blood (Debili N, Coulombel L, Croisille L, et al. Blood. 1996;88:1284-1296). This demonstration, and the fact that both bipotent and monopotent erythroid or megakaryocytic progenitors co-express markers of both lineages and respond to both lineage-specific transcription factors, has provided a background for the extensive use of MEP assessment by fluorescence-activated cell sorting in many subsequent studies. Beyond this, the demonstration of shared regulatory elements and the presence of single mutations affecting both lineages have inspired further studies to decipher how the shift in transcription factor networks occurs from one lineage to the other. Furthermore, in addition to shared effects, erythropoietin and thrombopoietin have additional independent effects. Most notable for thrombopoietin is its effect on hematopoietic stem cells illustrated by in vitro and in vivo approaches.


Subject(s)
Erythroid Precursor Cells/physiology , Erythropoietin/physiology , Megakaryocyte Progenitor Cells/physiology , Thrombopoietin/physiology , Animals , Biomarkers , Cell Differentiation , Cell Lineage , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythropoietin/pharmacology , Humans , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/drug effects , Phenotype , Thrombopoietin/pharmacology
20.
Semin Hematol ; 52(1): 4-11, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25578413

ABSTRACT

The production of platelets is a complex process that involves hematopoietic stem cells (HSCs), their differentiated progeny, the marrow microenvironment and hematopoietic cytokines. Much has been learned in the 110 years since James Homer Wright postulated that marrow megakaryocytes were responsible for blood platelet production, at a time when platelets were termed the "dust of the blood". In the 1980s a number of in vitro culture systems were developed that could produce megakaryocytes, followed by the identification of several cytokines that could stimulate the process in vitro. However, none of these cytokines produced a substantial thrombocytosis when injected into animals or people, nor were blood levels inversely related to platelet count, the sine qua non of a physiological regulator. A major milestone in our understanding of thrombopoiesis occurred in 1994 when thrombopoietin, the primary regulator of platelet production was cloned and initially characterized. Since that time many of the molecular mechanisms of thrombopoiesis have been identified, including the effects of thrombopoietin on the survival, proliferation, and differentiation of hematopoietic stem and progenitor cells, the development of polyploidy and proplatelet formation, the final fragmentation of megakaryocyte cytoplasm to yield blood platelets, and the regulation of this process. While much progress has been made, several outstanding questions remain, such as the nature of the signals for final platelet formation, the molecular nature of the regulation of marrow stromal thrombopoietin production, and the role of these physiological processes in malignant hematopoiesis.


Subject(s)
Thrombopoiesis , Animals , Bone Marrow , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cytokines/metabolism , Humans , Thrombopoiesis/drug effects , Thrombopoietin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...