Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 423
Filtrar
1.
Blood ; 141(5): 490-502, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36322928

RESUMEN

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by the clonal expansion of myeloid cells, notably megakaryocytes (MKs), and an aberrant cytokine production leading to bone marrow (BM) fibrosis and insufficiency. Current treatment options are limited. TGF-ß1, a profibrotic and immunosuppressive cytokine, is involved in PMF pathogenesis. While all cell types secrete inactive, latent TGF-ß1, only a few activate the cytokine via cell type-specific mechanisms. The cellular source of the active TGF-ß1 implicated in PMF is not known. Transmembrane protein GARP binds and activates latent TGF-ß1 on the surface of regulatory T lymphocytes (Tregs) and MKs or platelets. Here, we found an increased expression of GARP in the BM and spleen of mice with PMF and tested the therapeutic potential of a monoclonal antibody (mAb) that blocks TGF-ß1 activation by GARP-expressing cells. GARP:TGF-ß1 blockade reduced not only fibrosis but also the clonal expansion of transformed cells. Using mice carrying a genetic deletion of Garp in either Tregs or MKs, we found that the therapeutic effects of GARP:TGF-ß1 blockade in PMF imply targeting GARP on Tregs. These therapeutic effects, accompanied by increased IFN-γ signals in the spleen, were lost upon CD8 T-cell depletion. Our results suggest that the selective blockade of TGF-ß1 activation by GARP-expressing Tregs increases a CD8 T-cell-mediated immune reaction that limits transformed cell expansion, providing a novel approach that could be tested to treat patients with myeloproliferative neoplasms.


Asunto(s)
Mielofibrosis Primaria , Factor de Crecimiento Transformador beta1 , Ratones , Animales , Mielofibrosis Primaria/tratamiento farmacológico , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales/metabolismo , Citocinas/metabolismo , Fibrosis , Linfocitos T Reguladores
2.
Haematologica ; 109(7): 2060-2072, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38426279

RESUMEN

BCR::ABL1 negative myeloproliferative neoplasms (MPN) form a distinct group of hematologic malignancies characterized by sustained proliferation of cells from multiple myeloid lineages. With a median survival of 16-35 months in patients with high-risk disease, primary myelofibrosis (PMF) is considered the most aggressive entity amongst all BCR::ABL1 MPN. Additionally, for a significant subset of patients, MPN evolve into secondary acute myeloid leukemia (AML), which has an even poorer prognosis compared to de novo AML. As the exact mechanisms of disease development and progression remain to be elucidated, current therapeutic approaches fail to prevent disease progression or transformation into secondary AML. As each MPN entity is characterized by sustained activation of various immune cells and raised cytokine concentrations within bone marrow (BM) and peripheral blood (PB), MPN may be considered to be typical inflammation-related malignancies. However, the exact role and consequences of increased cytokine concentrations within BM and PB plasma has still not been completely established. Up-regulated cytokines can stimulate cellular proliferation, or contribute to the development of an inflammation-related BM niche resulting in genotoxicity and thereby supporting mutagenesis. The neutrophil chemoattractant CXCL8 is of specific interest as its concentration is increased within PB and BM plasma of patients with PMF. Increased concentration of CXCL8 negatively correlates with overall survival. Furthermore, blockage of the CXCR1/2 axis appears to be able to reduce BM fibrosis and megakaryocyte dysmorphia in murine models. In this review, we summarize available evidence on the role of the CXCL8-CXCR1/2 axis within the pathogenesis of PMF, and discuss potential therapeutic modalities targeting either CXCL8 or its cognate receptors CXCR1/2.


Asunto(s)
Interleucina-8 , Mielofibrosis Primaria , Receptores de Interleucina-8A , Receptores de Interleucina-8B , Humanos , Receptores de Interleucina-8B/metabolismo , Receptores de Interleucina-8B/genética , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/etiología , Mielofibrosis Primaria/mortalidad , Animales , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8A/genética , Interleucina-8/metabolismo , Transducción de Señal
3.
Int J Mol Sci ; 25(12)2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38928358

RESUMEN

Myeloproliferative neoplasms (MPNs), namely, polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are clonal stem cell disorders defined by an excessive production of functionally mature and terminally differentiated myeloid cells. MPNs can transform into secondary acute myeloid leukemia (sAML/blast phase MPN) and are linked to alterations in the redox balance, i.e., elevated concentrations of reactive oxygen species and markers of oxidative stress (OS), and changes in antioxidant systems. We evaluated OS in 117 chronic phase MPNs and 21 sAML cases versus controls by measuring total antioxidant capacity (TAC) and 8-hydroxy-2'-deoxy-guanosine (8-OHdG) concentrations. TAC was higher in MPNs than controls (p = 0.03), particularly in ET (p = 0.04) and PMF (p = 0.01). MPL W515L-positive MPNs had higher TAC than controls (p = 0.002) and triple-negative MPNs (p = 0.01). PMF patients who had treatment expressed lower TAC than therapy-free subjects (p = 0.03). 8-OHdG concentrations were similar between controls and MPNs, controls and sAML, and MPNs and sAML. We noted associations between TAC and MPNs (OR = 1.82; p = 0.05), i.e., ET (OR = 2.36; p = 0.03) and PMF (OR = 2.11; p = 0.03), but not sAML. 8-OHdG concentrations were not associated with MPNs (OR = 1.73; p = 0.62) or sAML (OR = 1.89; p = 0.49). In conclusion, we detected redox imbalances in MPNs based on disease subtype, driver mutations, and treatment history.


Asunto(s)
8-Hidroxi-2'-Desoxicoguanosina , Antioxidantes , Trastornos Mieloproliferativos , Humanos , Masculino , Femenino , 8-Hidroxi-2'-Desoxicoguanosina/metabolismo , Persona de Mediana Edad , Anciano , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Antioxidantes/metabolismo , Adulto , Estrés Oxidativo , Anciano de 80 o más Años , Crisis Blástica/metabolismo , Crisis Blástica/genética , Crisis Blástica/patología , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología
4.
J Transl Med ; 21(1): 703, 2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37814319

RESUMEN

Bone marrow fibrosis represents an important structural change in the marrow that interferes with some of its normal functions. The aetiopathogenesis of fibrosis is not well established except in its primary form. The present review consolidates current understanding of marrow fibrosis. We searched PubMed without time restriction using key words: bone marrow and fibrosis as the main stem against the terms: growth factors, cytokines and chemokines, morphology, megakaryocytes and platelets, myeloproliferative disorders, myelodysplastic syndrome, collagen biosynthesis, mesenchymal stem cells, vitamins and minerals and hormones, and mechanism of tissue fibrosis. Tissue marrow fibrosis-related papers were short listed and analysed for the review. It emerged that bone marrow fibrosis is the outcome of complex interactions between growth factors, cytokines, chemokines and hormones together with their facilitators and inhibitors. Fibrogenesis is initiated by mobilisation of special immunophenotypic subsets of mesenchymal stem cells in the marrow that transform into fibroblasts. Fibrogenic stimuli may arise from neoplastic haemopoietic or non-hematopoietic cells, as well as immune cells involved in infections and inflammatory conditions. Autoimmunity is involved in a small subset of patients with marrow fibrosis. Megakaryocytes and platelets are either directly involved or are important intermediaries in stimulating mesenchymal stem cells. MMPs, TIMPs, TGF-ß, PDGRF, and basic FGF and CRCXL4 chemokines are involved in these processes. Genetic and epigenetic changes underlie many of these conditions.


Asunto(s)
Médula Ósea , Mielofibrosis Primaria , Humanos , Médula Ósea/metabolismo , Mielofibrosis Primaria/etiología , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Citocinas/metabolismo , Fibrosis , Quimiocinas/metabolismo , Hormonas
5.
Am J Physiol Cell Physiol ; 322(2): C177-C184, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34910601

RESUMEN

Over the years, numerous studies demonstrated reciprocal communications between processes of bone marrow hematopoiesis and bone remodeling. Megakaryocytes, rare bone marrow cells responsible for platelet production, were demonstrated to be involved in bone homeostasis. Myelofibrosis, characterized by an increase in pleomorphic megakaryocytes in the bone marrow, commonly leads to the development of osteosclerosis. In vivo, an increase in megakaryocyte number was shown to result in osteosclerosis in GATA-1low, Nf-e2-/-, TPOhigh, Mplf/f;PF4cre, Lnk-/-, Mpig6b-/-, Mpig6bfl/fl;Gp1ba-Cr+/KI, and Pt-vWD mouse models. In vitro, megakaryocytes stimulate osteoblast proliferation and have variable effects on osteoclast proliferation and activity through soluble factors and direct cell-cell communications. Intriguingly, new studies revealed that the ability of megakaryocytes to communicate with bone cells is affected by the age and sex of animals. This mini-review summarizes changes seen in bone architecture and bone cell function in mouse models with an elevated number of megakaryocytes and the effects megakaryocytes have on osteoblasts and osteoclasts in vitro, and discusses potential molecular players that can mediate these effects.


Asunto(s)
Comunicación Celular/fisiología , Modelos Animales de Enfermedad , Megacariocitos/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Mielofibrosis Primaria/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Humanos , Megacariocitos/patología , Ratones , Ratones Noqueados , Osteoblastos/patología , Osteoclastos/patología , Mielofibrosis Primaria/patología
6.
Blood ; 136(18): 2051-2064, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-32726410

RESUMEN

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm (MPN) that leads to progressive bone marrow (BM) fibrosis. Although the cellular mutations involved in the pathogenesis of PMF have been extensively investigated, the sequential events that drive stromal activation and fibrosis by hematopoietic-stromal cross-talk remain elusive. Using an unbiased approach and validation in patients with MPN, we determined that the differential spatial expression of the chemokine CXCL4/platelet factor-4 marks the progression of fibrosis. We show that the absence of hematopoietic CXCL4 ameliorates the MPN phenotype, reduces stromal cell activation and BM fibrosis, and decreases the activation of profibrotic pathways in megakaryocytes, inflammation in fibrosis-driving cells, and JAK/STAT activation in both megakaryocytes and stromal cells in 3 murine PMF models. Our data indicate that higher CXCL4 expression in MPN has profibrotic effects and is a mediator of the characteristic inflammation. Therefore, targeting CXCL4 might be a promising strategy to reduce inflammation in PMF.


Asunto(s)
Médula Ósea/patología , Fibrosis/patología , Inflamación/patología , Trastornos Mieloproliferativos/complicaciones , Factor Plaquetario 4/metabolismo , Mielofibrosis Primaria/patología , Animales , Médula Ósea/inmunología , Médula Ósea/metabolismo , Proliferación Celular , Progresión de la Enfermedad , Fibrosis/etiología , Fibrosis/inmunología , Fibrosis/metabolismo , Humanos , Inflamación/etiología , Inflamación/inmunología , Inflamación/metabolismo , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Masculino , Megacariocitos , Ratones , Ratones Noqueados , Mutación , Factor Plaquetario 4/genética , Mielofibrosis Primaria/etiología , Mielofibrosis Primaria/inmunología , Mielofibrosis Primaria/metabolismo
7.
Future Oncol ; 18(20): 2559-2571, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35603634

RESUMEN

Myelofibrosis is a myeloproliferative neoplasm characterized by splenomegaly, debilitating constitutional symptoms and bone marrow failure. Disease-related anemia is common and associated with an inferior quality of life and survival. Unfortunately, few therapies exist to improve hemoglobin in myelofibrosis patients. Momelotinib is a JAK1/JAK2 inhibitor that also antagonizes ACVR1, leading to downregulation of hepcidin expression and increased availability of iron for erythropoiesis. In clinical testing, momelotinib has demonstrated a unique ability to improve hemoglobin and reduce transfusion burden in myelofibrosis patients with baseline anemia, while producing reductions in spleen size and symptom burden. This review explores the preclinical rationale, clinical trial data and future role of momelotinib in the evolving therapeutic landscape of myelofibrosis.


Patients with myelofibrosis (MF), a blood cancer, experience many symptoms including tiredness, night sweats and an increased spleen size. They also may experience low red blood cell counts (anemia) and require blood transfusions. MF is normally treated with medications called JAK inhibitors, but they worsen anemia. Momelotinib is a new JAK inhibitor that may be able to improve anemia. This is a review article that covers the available information on momelotinib and describes how this new drug may be incorporated into the future treatment of MF.


Asunto(s)
Anemia , Inhibidores de las Cinasas Janus , Mielofibrosis Primaria , Anemia/tratamiento farmacológico , Anemia/etiología , Benzamidas/uso terapéutico , Humanos , Janus Quinasa 2/genética , Nitrilos/uso terapéutico , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/tratamiento farmacológico , Mielofibrosis Primaria/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas , Calidad de Vida
8.
Curr Opin Hematol ; 28(5): 364-371, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34232140

RESUMEN

PURPOSE OF REVIEW: Bone marrow fibrosis is the progressive replacement of blood-forming cells by reticulin fibres, caused by the acquisition of somatic mutations in hematopoietic stem cells. The molecular and cellular mechanisms that drive the progression of bone marrow fibrosis remain unknown, yet chronic inflammation appears to be a conserved feature in most patients suffering from myeloproliferative neoplasms. RECENT FINDINGS: Here, we review recent literature pertaining to the role of inflammation in driving bone marrow fibrosis, and its effect on the various hematopoietic and nonhematopoietic cell populations. SUMMARY: Recent evidence suggests that the pathogenesis of MPN is primarily driven by the hematopoietic stem and progenitor cells, together with their mutated progeny, which in turn results in chronic inflammation that disrupts the bone marrow niche and perpetuates a disease-permissive environment. Emerging data suggests that specifically targeting stromal inflammation in combination with JAK inhibition may be the way forward to better treat MPNs, and bone marrow fibrosis specifically.


Asunto(s)
Médula Ósea , Neoplasias Hematológicas , Células Madre Hematopoyéticas , Mielofibrosis Primaria , Médula Ósea/metabolismo , Médula Ósea/patología , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Mielofibrosis Primaria/tratamiento farmacológico , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología
9.
Blood ; 133(21): 2320-2324, 2019 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-30745304

RESUMEN

Bone marrow (BM) sclerosis is commonly found in patients with late-stage myelofibrosis (MF). Because osteoclasts (OCs) and osteoblasts play a key role in bone remodeling, and MF monocytes, the OC precursors, are derived from the neoplastic clone, we wondered whether decreased OC numbers or impairment in their osteolytic function affects the development of osteosclerosis. Analysis of BM biopsies from 50 MF patients showed increased numbers of multinucleated tartrate-resistant acid phosphatase (TRAP)/cathepsin K+ OCs expressing phosphorylated Janus kinase 2 (JAK2). Randomly microdissected TRAP+ OCs from 16 MF patients harbored JAK2 or calreticulin (CALR) mutations, confirming MF OCs are clonal. To study OC function, CD14+ monocytes from MF patients and healthy individuals were cultured and differentiated into OCs. Unlike normal OCs, MF OCs appeared small and round, with few protrusions, and carried the mutations and chromosomal abnormalities of neoplastic clones. In addition, MF OCs lacked F-actin-rich ring-like structures and had fewer nuclei and reduced colocalization signals, compatible with decreased fusion events, and their mineral resorption capacity was significantly reduced, indicating impaired osteolytic function. Taken together, our data suggest that, although the numbers of MF OCs are increased, their impaired osteolytic activity distorts bone remodeling and contributes to the induction of osteosclerosis.


Asunto(s)
Remodelación Ósea , Osteoclastos , Osteólisis , Mielofibrosis Primaria , Calreticulina/metabolismo , Catepsina K/genética , Catepsina K/metabolismo , Femenino , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Masculino , Mutación , Osteoclastos/metabolismo , Osteoclastos/patología , Osteólisis/genética , Osteólisis/metabolismo , Osteólisis/patología , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Fosfatasa Ácida Tartratorresistente/genética , Fosfatasa Ácida Tartratorresistente/metabolismo
10.
Ann Hematol ; 100(1): 105-116, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33089365

RESUMEN

Mesenchymal stromal cells (MSCs) represent an essential component of the bone marrow (BM) niche and display disease-specific alterations in several myeloid malignancies. The aim of this work was to study possible MSC abnormalities in Philadelphia-negative myeloproliferative neoplasms (MPNs) in relationship to the degree of BM fibrosis. MSCs were isolated from BM of 6 healthy donors (HD) and of 23 MPN patients, classified in 3 groups according to the diagnosis and the grade of BM fibrosis: polycythemia vera and essential thrombocythemia (PV/ET), low fibrosis myelofibrosis (LF-MF), and high fibrosis MF (HF-MF). MSC cultures were established from 21 of 23 MPN patients. MPN-derived MSCs did not exhibit any functional impairment in their adipogenic/osteogenic/chondrogenic differentiation potential and displayed a phenotype similar to HD-derived MSCs but with a decreased expression of CD146. All MPN-MSC lines were negative for the patient-specific hematopoietic clone mutations (JAK2, MPL, CALR). MSCs derived from HF-MF patients displayed a reduced clonogenic potential and a lower growth kinetic compared to MSCs from HD, LF-MF, and PV/ET patients. mRNA levels of hematopoiesis regulatory molecules were unaffected in MSCs from HF-MF compared to HD. Finally, in vitro ActivinA secretion by MSCs was increased in HF-MF compared to LF-MF patients, in association with a lower hemoglobin value. Increased ActivinA immunolabeling on stromal cells and erythroid precursors was also observed in HF-MF BM biopsies. In conclusion, higher grade of BM fibrosis is associated with functional impairment of MSCs and the increased secretion of ActivinA may represent a suitable target for anemia treatment in MF patients.


Asunto(s)
Activinas/metabolismo , Médula Ósea/metabolismo , Células Madre Mesenquimatosas/metabolismo , Trastornos Mieloproliferativos/metabolismo , Mielofibrosis Primaria/metabolismo , Adulto , Anciano , Médula Ósea/patología , Diferenciación Celular/fisiología , Células Cultivadas , Estudios de Cohortes , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/patología , Persona de Mediana Edad , Trastornos Mieloproliferativos/patología , Policitemia Vera/metabolismo , Policitemia Vera/patología , Mielofibrosis Primaria/patología , Trombocitemia Esencial/metabolismo , Trombocitemia Esencial/patología
11.
J Cell Mol Med ; 24(19): 11100-11110, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32889753

RESUMEN

Primary myelofibrosis (PMF) is a Ph-negative myeloproliferative neoplasm (MPN), characterized by advanced bone marrow fibrosis and extramedullary haematopoiesis. The bone marrow fibrosis results from excessive proliferation of fibroblasts that are influenced by several cytokines in the microenvironment, of which transforming growth factor-ß (TGF-ß) is the most important. Micromechanics related to the niche has not yet been elucidated. In this study, we hypothesized that mechanical stress modulates TGF-ß signalling leading to further activation and subsequent proliferation and invasion of bone marrow fibroblasts, thus showing the important role of micromechanics in the development and progression of PMF, both in the bone marrow and in extramedullary sites. Using three PMF-derived fibroblast cell lines and transforming growth factor-ß receptor (TGFBR) 1 and 2 knock-down PMF-derived fibroblasts, we showed that mechanical stress does stimulate the collagen synthesis by the fibroblasts in patients with myelofibrosis, through the TGFBR1, which however seems to be activated through alternative pathways, other than TGFBR2.


Asunto(s)
Progresión de la Enfermedad , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/fisiopatología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Fenómenos Biomecánicos , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/diagnóstico por imagen , Ratones Desnudos , Modelos Biológicos , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/patología , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Estrés Mecánico
12.
Br J Haematol ; 191(2): 152-170, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32196650

RESUMEN

Myelofibrosis is an enigmatic myeloproliferative neoplasm, despite noteworthy strides in understanding its genetic underpinnings. Driver mutations involving JAK2, CALR or MPL in 90% of patients mediate constitutive JAK-STAT signaling which, in concert with epigenetic alterations (ASXL1, DNMT3A, SRSF2, EZH2, IDH1/2 mutations), play a fundamental role in disease pathogenesis. Aberrant immature megakaryocytes are a quintessential feature, exhibiting reduced GATA1 protein expression and secreting a plethora of pro-inflammatory cytokines (IL-1 ß, TGF-ß), growth factors (b-FGF, PDGF, VEGF) in addition to extra cellular matrix components (fibronectin, laminin, collagens). The ensuing disrupted interactions amongst the megakaryocytes, osteoblasts, endothelium, stromal cells and myofibroblasts within the bone marrow culminate in the development of fibrosis and osteosclerosis. Presently, prognostic assessment tools for primary myelofibrosis (PMF) are centered on genetics, with incorporation of cytogenetic and molecular information into the mutation-enhanced (MIPSS 70-plus version 2.0) and genetically-inspired (GIPSS) prognostic scoring systems. Both models illustrate substantial clinical heterogeneity in PMF and serve as the crux for risk-adapted therapeutic decisions. A major challenge remains the dearth of disease-modifying drugs, whereas allogeneic transplant offers the chance of long-term remission for some patients. Our review serves to synopsise current appreciation of the pathogenesis of myelofibrosis together with emerging management strategies.


Asunto(s)
Regulación de la Expresión Génica , Mutación , Mielofibrosis Primaria , Humanos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Mielofibrosis Primaria/terapia
13.
Blood Cells Mol Dis ; 81: 102396, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31837568

RESUMEN

Aurora-kinase-A (AURKA), BORA and Polo-like-kinase-1 (PLK1) are regulating cell-cycle control and promotion of mitosis entry. AURKA contributes to Janus-kinase-2 (JAK2) activation and increased AURKA protein levels were reported in CD34+ and CD41+ cells of myeloproliferative neoplasm patients, leading to aneuploidy and aberrant megakaryopoiesis. We aimed to investigate AURKA, BORA and PLK1 mRNA expression in unfractionated bone-marrow aspirates of 43 patients with myelofibrosis (28 primary-/PMF, 15 secondary-myelofibrosis/SMF) and 12 controls and to assess their clinical correlations. AURKA expression did not significantly differ between myelofibrosis and controls (P = 0.466). Higher AURKA expression was significantly associated with higher absolute monocyte-count (P = 0.024) and shorter overall survival (HR = 3.77; P = 0.012). Patients with both PMF and SMF had lower BORA expression than controls (P = 0.009). Higher BORA expression was significantly associated with absence of constitutional symptoms (P = 0.049), absence of circulatory blasts (P = 0.047), higher monocyte- (P = 0.040) and higher eosinophil-counts (P = 0.016) and had neutral effect on survival (P > 0.05). PLK1 expression did not significantly differ between myelofibrosis and controls (P = 0.103). Higher PLK1 expression was significantly associated with higher white-blood-cell-count (P = 0.042) and inferior overall survival (HR = 5.87; P = 0.003). In conclusion, AURKA, BORA and PLK1 are involved in pathogenesis of myelofibrosis and may affect survival. Future studies investigating these interesting associations are warranted.


Asunto(s)
Aurora Quinasa A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Mielofibrosis Primaria/mortalidad , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mielofibrosis Primaria/metabolismo , Pronóstico , Tasa de Supervivencia , Quinasa Tipo Polo 1
14.
Blood Cells Mol Dis ; 82: 102420, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32179412

RESUMEN

BACKGROUND: We aimed to investigate the association of bone marrow mast cell numbers (MCN) and the degree of reticulin fibrosis in patients with chronic myeloproliferative neoplasms (MPN). METHODS: This was a case-control study that recruited 47 patients who were diagnosed with bcr-abl negative MPN. Thirty patients with lymphoma served as controls. JAK2 mutation was studied and all subjects underwent bone marrow biopsy at the time of diagnosis. Mast and CD34+ cells were counted. Marrow reticulin fiber was graded. RESULTS: Thirty-four patients had essential thrombocythemia (ET), 8 patients had primary myelofibrosis (PMF) and 5 patients had polycythemia vera (PV). Fourteen MPN patients had JAK2, whereas the controls had not. MCN was higher in patients than controls (p = 0.001). There was no significant difference regarding CD34. Reticulin fibrosis was present in 57.4% of MPN patients, whereas there was any in controls. PMF patients had more CD34 + cells than PV and ET. PMF patients had more reticulin fibers compared with other subgroups (p < 0.001). MCN, but not CD34+ cell counts, was significantly higher in JAK2(+) patients than JAK2(-) patients. CONCLUSION: MCN and reticulin fibrosis were significantly increased in MPN patients. JAK2 positivity had significantly increased MCN compared to patients without JAK2. JAK2 was associated with increased reticulin fibrosis.


Asunto(s)
Neoplasias Hematológicas , Mastocitos , Trastornos Mieloproliferativos , Proteínas de Neoplasias , Mielofibrosis Primaria , Reticulina , Anciano , Enfermedad Crónica , Femenino , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Humanos , Janus Quinasa 2 , Masculino , Mastocitos/metabolismo , Mastocitos/patología , Persona de Mediana Edad , Mutación , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Reticulina/genética , Reticulina/metabolismo
15.
Blood ; 132(19): 2053-2066, 2018 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-30213875

RESUMEN

Although the pathogenesis of primary myelofibrosis (PMF) and other myeloproliferative neoplasms (MPNs) is linked to constitutive activation of the JAK-STAT pathway, JAK inhibitors have neither curative nor MPN-stem cell-eradicating potential, indicating that other targetable mechanisms are contributing to the pathophysiology of MPNs. We previously demonstrated that Abelson interactor 1 (Abi-1), a negative regulator of Abelson kinase 1, functions as a tumor suppressor. Here we present data showing that bone marrow-specific deletion of Abi1 in a novel mouse model leads to development of an MPN-like phenotype resembling human PMF. Abi1 loss resulted in a significant increase in the activity of the Src family kinases (SFKs), STAT3, and NF-κB signaling. We also observed impairment of hematopoietic stem cell self-renewal and fitness, as evidenced in noncompetitive and competitive bone marrow transplant experiments. CD34+ hematopoietic progenitors and granulocytes from patients with PMF showed decreased levels of ABI1 transcript as well as increased activity of SFKs, STAT3, and NF-κB. In aggregate, our data link the loss of Abi-1 function to hyperactive SFKs/STAT3/NF-κB signaling and suggest that this signaling axis may represent a regulatory module involved in the molecular pathophysiology of PMF.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Médula Ósea/patología , Proteínas del Citoesqueleto/genética , Eliminación de Gen , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/patología , Animales , Médula Ósea/metabolismo , Autorrenovación de las Células , Células Cultivadas , Regulación hacia Abajo , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Mielofibrosis Primaria/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo
16.
Blood ; 131(19): 2111-2119, 2018 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-29572380

RESUMEN

Myofibroblasts are fibrosis-driving cells and are well characterized in solid organ fibrosis, but their role and cellular origin in bone marrow fibrosis remains obscure. Recent work has demonstrated that Gli1+ and LepR+ mesenchymal stromal cells (MSCs) are progenitors of fibrosis-causing myofibroblasts in the bone marrow. Genetic ablation of Gli1+ MSCs or pharmacologic targeting of hedgehog (Hh)-Gli signaling ameliorated fibrosis in mouse models of myelofibrosis (MF). Moreover, pharmacologic or genetic intervention in platelet-derived growth factor receptor α (Pdgfrα) signaling in Lepr+ stromal cells suppressed their expansion and ameliorated MF. Improved understanding of cellular and molecular mechanisms in the hematopoietic stem cell niche that govern the transition of MSCs to myofibroblasts and myofibroblast expansion in MF has led to new paradigms in the pathogenesis and treatment of MF. Here, we highlight the central role of malignant hematopoietic clone-derived megakaryocytes in reprogramming the hematopoietic stem cell niche in MF with potential detrimental consequences for hematopoietic reconstitution after allogenic stem cell transplantation, so far the only therapeutic approach in MF considered to be curative. We and others have reported that targeting Hh-Gli signaling is a therapeutic strategy in solid organ fibrosis. Data indicate that targeting Gli proteins directly inhibits Gli1+ cell proliferation and myofibroblast differentiation, which results in reduced fibrosis severity and improved organ function. Although canonical Hh inhibition (eg, smoothened [Smo] inhibition) failed to improve pulmonary fibrosis, kidney fibrosis, or MF, the direct inhibition of Gli proteins ameliorated fibrosis. Therefore, targeting Gli proteins directly might be an interesting and novel therapeutic approach in MF.


Asunto(s)
Miofibroblastos/metabolismo , Mielofibrosis Primaria/etiología , Mielofibrosis Primaria/metabolismo , Biomarcadores , Células Cultivadas , Reprogramación Celular , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Mielofibrosis Primaria/patología , Mielofibrosis Primaria/terapia , Transducción de Señal/efectos de los fármacos , Nicho de Células Madre , Células del Estroma/metabolismo , Transcriptoma , Proteína con Dedos de Zinc GLI1/metabolismo
17.
J Bone Miner Metab ; 38(2): 145-150, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31897749

RESUMEN

The bone marrow (BM) is located inside the bone. Now, it appears that bone tissue functionally communicates with the BM hematopoietic system. Osteoblast lineage cells serve as a part of the microenvironment for immature hematopoietic (stem/progenitor) cells. In contrast, mature hematopoietic cells such as neutrophils and macrophages play a critical role to regulate osteoblast activity. A progressive distortion of this precise inter-organ communication between hematopoietic and skeletal systems may lead to hematologic disorders. Recent studies have revealed that vitamin D receptor is a pivotal bridging molecule for this network and for the pathogenesis of myelofibrosis.


Asunto(s)
Huesos/fisiología , Hematología , Animales , Huesos/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas , Humanos , Mielofibrosis Primaria/metabolismo , Receptores de Calcitriol/metabolismo
18.
Pediatr Blood Cancer ; 67(5): e28232, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32134181

RESUMEN

OBJECTIVES: Pediatric myelofibrosis is a rare entity with the largest reported series of 19 cases. We describe here the clinicopathological spectrum and outcomes of 15 cases of pediatric myelofibrosis. METHODS: Case files of myelofibrosis of patients less than 18 years were retrieved from January 2016 to January 2019, and patients with idiopathic myelofibrosis after exhaustive work-up were studied. Their clinicopathological profiles were studied and then followed up for resolution and malignant transformation. RESULTS: Of the 15 cases of idiopathic myelofibrosis, transfusion-dependent anemia (14/15) was most common presentation. Only one patient showed leukoerythroblastosis with dacryocytes. Myeloid hyperplasia was seen in 13 of 15 patients and megakaryocytic hyperplasia in 10 patients. Dysmegakaryopoiesis was seen in 8 of 15 patients, and only three had small loose megakaryocytic clustering. None showed hyperchromatic megakaryocytes, intrasinusoidal hematopoiesis, or osteosclerosis. One patient with trisomy 8 tested positive for JAK2V617F. Bone marrow biopsy was hypercellular in 13, and 8 had world health organization (WHO) MF-3 fibrosis. None of the patients developed malignancy, one had spontaneous resolution, and one patient required allogenic stem cell transplant. CONCLUSIONS: Pediatric myelofibrosis is a distinct entity from primary myelofibrosis in adults and merits mention in the WHO manual as a distinct entity.


Asunto(s)
Transformación Celular Neoplásica , Janus Quinasa 2 , Mutación Missense , Proteínas de Neoplasias , Trombopoyesis , Adolescente , Adulto , Sustitución de Aminoácidos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Niño , Preescolar , Femenino , Humanos , Lactante , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Masculino , Megacariocitos/metabolismo , Megacariocitos/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Osteosclerosis/genética , Osteosclerosis/metabolismo , Osteosclerosis/patología , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Estudios Retrospectivos
19.
Proc Natl Acad Sci U S A ; 114(18): 4757-4762, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28424250

RESUMEN

Fibrotic diseases are not well-understood. They represent a number of different diseases that are characterized by the development of severe organ fibrosis without any obvious cause, such as the devastating diseases idiopathic pulmonary fibrosis (IPF) and scleroderma. These diseases have a poor prognosis comparable with endstage cancer and are uncurable. Given the phenotypic differences, it was assumed that the different fibrotic diseases also have different pathomechanisms. Here, we demonstrate that many endstage fibrotic diseases, including IPF; scleroderma; myelofibrosis; kidney-, pancreas-, and heart-fibrosis; and nonalcoholic steatohepatosis converge in the activation of the AP1 transcription factor c-JUN in the pathologic fibroblasts. Expression of the related AP1 transcription factor FRA2 was restricted to pulmonary artery hypertension. Induction of c-Jun in mice was sufficient to induce severe fibrosis in multiple organs and steatohepatosis, which was dependent on sustained c-Jun expression. Single cell mass cytometry revealed that c-Jun activates multiple signaling pathways in mice, including pAkt and CD47, which were also induced in human disease. αCD47 antibody treatment and VEGF or PI3K inhibition reversed various organ c-Jun-mediated fibroses in vivo. These data suggest that c-JUN is a central molecular mediator of most fibrotic conditions.


Asunto(s)
Fibrosis Pulmonar Idiopática , Mielofibrosis Primaria , Proteínas Proto-Oncogénicas c-jun , Esclerodermia Sistémica , Factor de Transcripción AP-1 , Antígeno 2 Relacionado con Fos/genética , Antígeno 2 Relacionado con Fos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/metabolismo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo
20.
Int J Mol Sci ; 21(8)2020 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-32331228

RESUMEN

Primary myelofibrosis (PMF) is a rare myeloproliferative neoplasm characterized by stem-cell-derived clonal over-proliferation of mature myeloid lineages, bone marrow fibrosis, osteosclerosis, defective erythropoiesis, and pro-inflammatory cytokine over-expression. The aim of the present study was to highlight possible differences in the transcriptome among CD34+ cells from peripheral blood (PB) of PMF patients. Therefore, we merged two microarray datasets of healthy control subjects and PMF (34 JAK2V617F MUTATED and 28 JAK2 wild-type). The GO analysis of upregulated genes revealed enrichment for JAK2/STAT1 pathway gene set in PB CD34+ cells of PMF patients with and without the JAK2V617F mutation comparing to the healthy control subjects, and in particular a significant upregulation of immunoproteasome (IP)-belonging genes as PSMB8, PSMB9, and PSMB10. A more detailed investigation of the IFN-gamma (IFNG) pathway also revealed that IFNG, IRF1, and IFNGR2 were significantly upregulated in PB CD34+ cells of PMF patients carrying the mutation for JAK2V617F compared to JAK2 wild-type PMF patients. Finally, we showed an upregulation of HLA-class I genes in PB CD34+ cells from PMF JAK2V617F mutated patients compared to JAK2 wild-type and healthy controls. In conclusion, our results demonstrate that IPs and IFNG pathways could be involved in PMF disease and in particular in patients carrying the JAK2V617F mutation.


Asunto(s)
Inmunomodulación/genética , Janus Quinasa 2/genética , Mutación , Mielofibrosis Primaria/genética , Complejo de la Endopetidasa Proteasomal/genética , Alelos , Antígenos/metabolismo , Antígenos CD34/metabolismo , Células Cultivadas , Biología Computacional/métodos , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Estudios de Asociación Genética , Humanos , Modelos Biológicos , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/inmunología , Mielofibrosis Primaria/metabolismo , Pronóstico , Complejo de la Endopetidasa Proteasomal/metabolismo , Curva ROC , Transducción de Señal
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda