Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 2(21)2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29093266

RESUMEN

Neurogenic heterotopic ossification (NHO) is the formation of ectopic bone generally in muscles surrounding joints following spinal cord or brain injury. We investigated the mechanisms of NHO formation in 64 patients and a mouse model of spinal cord injury-induced NHO. We show that marrow from human NHOs contains hematopoietic stem cell (HSC) niches, in which mesenchymal stromal cells (MSCs) and endothelial cells provide an environment supporting HSC maintenance, proliferation, and differentiation. The transcriptomic signature of MSCs from NHOs shows a neuronal imprinting associated with a molecular network required for HSC support. We demonstrate that oncostatin M (OSM) produced by activated macrophages promotes osteoblastic differentiation and mineralization of human muscle-derived stromal cells surrounding NHOs. The key role of OSM was confirmed using an experimental model of NHO in mice defective for the OSM receptor (OSMR). Our results provide strong evidence that macrophages contribute to NHO formation through the osteogenic action of OSM on muscle cells within an inflammatory context and suggest that OSM/OSMR could be a suitable therapeutic target. Altogether, the evidence of HSCs in ectopic bones growing at the expense of soft tissue in spinal cord/brain-injured patients indicates that inflammation and muscle contribute to HSC regulation by the brain-bone-blood triad.


Asunto(s)
Macrófagos/metabolismo , Oncostatina M/metabolismo , Osificación Heterotópica/inmunología , Osificación Heterotópica/metabolismo , Animales , Antígenos CD34 , Lesiones Encefálicas , Diferenciación Celular , Proliferación Celular , Células Endoteliales , Femenino , Hematopoyesis , Células Madre Hematopoyéticas , Xenoinjertos , Humanos , Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidad beta del Receptor de Oncostatina M , Osificación Heterotópica/patología , Osteogénesis , Médula Espinal , Transcriptoma
2.
Cancer Res ; 75(22): 4753-65, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26404004

RESUMEN

Primary myelofibrosis is a myeloproliferative neoplasm that is a precursor to myeloid leukemia. Dysmegakaryopoiesis and extramedullary hematopoiesis characterize primary myelofibrosis, which is also associated with bone marrow stromal alterations marked by fibrosis, neoangiogenesis, and osteomyelosclerosis. In particular, contributions to primary myelofibrosis from mesenchymal stromal cells (MSC) have been suggested by mouse studies, but evidence in humans remains lacking. In this study, we show that bone marrow MSCs from primary myelofibrosis patients exhibit unique molecular and functional abnormalities distinct from other myeloproliferative neoplasms and these abnormalities are maintained stably ex vivo in the absence of leukemic cells. Primary myelofibrosis-MSC overexpressed heparin-binding cytokines, including proinflammatory TGFß1 and osteogenic BMP-2, as well as glycosaminoglycans such as heparan sulfate and chondroitin sulfate. Transcriptome and functional analyses revealed alterations in MSC differentiation characterized by an increased osteogenic potential and a TGFß1 signaling signature. Accordingly, phospho-Smad2 levels were intrinsically increased in primary myelofibrosis-MSC along with enhanced expression of the master bone regulator RUNX2, while inhibition of the endogenous TGFß1 receptor TGFßR1 impaired osteogenic differentiation in these MSCs. Taken together, our results define the source of a critical osteogenic function in primary myelofibrosis that supports its pathophysiology, suggesting that combined targeting of both the hematopoietic and stromal cell compartments in primary myelofibrosis patients may heighten therapeutic efficacy.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/patología , Osificación Heterotópica/fisiopatología , Mielofibrosis Primaria/fisiopatología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Células Cultivadas , Femenino , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa
3.
Haematologica ; 100(6): 757-67, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25840601

RESUMEN

Primary myelofibrosis is characterized by clonal myeloproliferation, dysmegakaryopoiesis, extramedullary hematopoiesis associated with myelofibrosis and altered stroma in the bone marrow and spleen. The expression of CD9, a tetraspanin known to participate in megakaryopoiesis, platelet formation, cell migration and interaction with stroma, is deregulated in patients with primary myelofibrosis and is correlated with stage of myelofibrosis. We investigated whether CD9 participates in the dysmegakaryopoiesis observed in patients and whether it is involved in the altered interplay between megakaryocytes and stromal cells. We found that CD9 expression was modulated during megakaryocyte differentiation in primary myelofibrosis and that cell surface CD9 engagement by antibody ligation improved the dysmegakaryopoiesis by restoring the balance of MAPK and PI3K signaling. When co-cultured on bone marrow mesenchymal stromal cells from patients, megakaryocytes from patients with primary myelofibrosis displayed modified behaviors in terms of adhesion, cell survival and proliferation as compared to megakaryocytes from healthy donors. These modifications were reversed after antibody ligation of cell surface CD9, suggesting the participation of CD9 in the abnormal interplay between primary myelofibrosis megakaryocytes and stroma. Furthermore, silencing of CD9 reduced CXCL12 and CXCR4 expression in primary myelofibrosis megakaryocytes as well as their CXCL12-dependent migration. Collectively, our results indicate that CD9 plays a role in the dysmegakaryopoiesis that occurs in primary myelofibrosis and affects interactions between megakaryocytes and bone marrow stromal cells. These results strengthen the "bad seed in bad soil" hypothesis that we have previously proposed, in which alterations of reciprocal interactions between hematopoietic and stromal cells participate in the pathogenesis of primary myelofibrosis.


Asunto(s)
Megacariocitos/metabolismo , Mielofibrosis Primaria/metabolismo , Células del Estroma/metabolismo , Tetraspanina 29/fisiología , Trombopoyesis/fisiología , Técnicas de Cocultivo , Humanos , Megacariocitos/patología , Mielofibrosis Primaria/patología , Células del Estroma/patología
4.
J Pathol ; 236(2): 229-40, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25712044

RESUMEN

Neurological heterotopic ossification (NHO) is the abnormal formation of bone in soft tissues as a consequence of spinal cord or traumatic brain injury. NHO causes pain, ankyloses, vascular and nerve compression and delays rehabilitation in this high-morbidity patient group. The pathological mechanisms leading to NHO remain unknown and consequently there are no therapeutic options to prevent or reduce NHO. Genetically modified mouse models of rare genetic forms of heterotopic ossification (HO) exist, but their relevance to NHO is questionable. Consequently, we developed the first model of spinal cord injury (SCI)-induced NHO in genetically unmodified mice. Formation of NHO, measured by micro-computed tomography, required the combination of both SCI and localized muscular inflammation. Our NHO model faithfully reproduced many clinical features of NHO in SCI patients and both human and mouse NHO tissues contained macrophages. Muscle-derived mesenchymal progenitors underwent osteoblast differentiation in vitro in response to serum from NHO mice without additional exogenous osteogenic stimuli. Substance P was identified as a candidate NHO systemic neuropeptide, as it was significantly elevated in the serum of NHO patients. However, antagonism of substance P receptor in our NHO model only modestly reduced the volume of NHO. In contrast, ablation of phagocytic macrophages with clodronate-loaded liposomes reduced the size of NHO by 90%, supporting the conclusion that NHO is highly dependent on inflammation and phagocytic macrophages in soft tissues. Overall, we have developed the first clinically relevant model of NHO and demonstrated that a combined insult of neurological injury and soft tissue inflammation drives NHO pathophysiology.


Asunto(s)
Macrófagos/fisiología , Miositis/etiología , Osificación Heterotópica/etiología , Traumatismos de la Médula Espinal/complicaciones , Animales , Cardiotoxinas/farmacología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones Endogámicos C57BL , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Paraplejía/complicaciones , Células Madre/fisiología
5.
Blood ; 123(2): 191-202, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24277075

RESUMEN

In addition to its well-known effect on migration and homing of hematopoietic stem/progenitor cells (HSPCs), CXCL12 chemokine also exhibits a cell cycle and survival-promoting factor for human CD34(+) HSPCs. CXCR4 was suggested to be responsible for CXCL12-induced biological effects until the recent discovery of its second receptor, CXCR7. Until now, the participation of CXCR7 in CXCL12-induced HSPC cycling and survival is unknown. We show here that CXCL12 was capable of binding CXCR7 despite its scarce expression at CD34(+) cell surface. Blocking CXCR7 inhibited CXCL12-induced Akt activation as well as the percentage of CD34(+) cells in cycle, colony formation, and survival, demonstrating its participation in CXCL12-induced functional effects in HSPCs. At steady state, CXCR7 and ß-arrestin2 co-localized near the plasma membrane of CD34(+) cells. After CXCL12 treatment, ß-arrestin2 translocated to the nucleus, and this required both CXCR7 and CXCR4. Silencing ß-arrestin expression decreased CXCL12-induced Akt activation in CD34(+) cells. Our results demonstrate for the first time the role of CXCR7, complementary to that played by CXCR4, in the control of HSPC cycling, survival, and colony formation induced by CXCL12. We also provide evidence for the involvement of ß-arrestins as signaling hubs downstream of both CXCL12 receptors in primary human HSPCs.


Asunto(s)
Arrestinas/metabolismo , Ciclo Celular , Quimiocina CXCL12/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores CXCR/metabolismo , Antígenos CD34/metabolismo , Supervivencia Celular , Ensayo de Unidades Formadoras de Colonias , Activación Enzimática , Humanos , Espacio Intracelular/metabolismo , Fosforilación , Unión Proteica , Transporte de Proteínas , beta-Arrestinas
6.
Haematologica ; 96(8): 1213-20, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21546497

RESUMEN

Umbilical cord blood transplantation has been increasingly used over the past years for both malignant and non-malignant hematologic and other diseases as an alternative to mismatched-related or matched-unrelated bone marrow or peripheral blood hematopoietic stem cell transplantation. A disadvantage of cord blood is its low cell content which limits cord blood transplantation to generally low weight recipients, such as children. Various alternatives have been used to overcome this limitation, including co-infusion of two partially HLA-matched cord blood units. According to Eurocord Registry data, this strategy has been applied in approximately 993 adult patients with hematologic diseases since the first double umbilical cord blood transplantation in 1999. In fact, since 2005, the number of adult patients receiving double umbilical cord blood transplantation has surpassed the number of adults transplanted with single cord blood units. The engraftment rate is comparable for both single and double umbilical cord blood transplantation, although the latter is accompanied by a higher incidence of grade II acute graft-versus-host disease and lower leukemia relapse for patients in first complete remission. In the majority of patients undergoing double umbilical cord blood transplantation, transient chimerism, due to the presence of cells from both donor units early post transplant, is replaced by sustained dominance of one unit from which long-term hematopoiesis is derived. Although the biology and the factors that determine unit dominance have not been clarified, the implication of immune-mediated mechanisms has been reported. Preliminary data have demonstrated the safety of double umbilical cord blood transplantation. Ongoing clinical trials and prolonged follow up of the patients will clarify the immunology and determine the efficacy of this approach. We present here a brief overview of the clinical experience on double umbilical cord blood transplantation and its underlying biology.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical , Animales , Quimerismo , Trasplante de Células Madre de Sangre del Cordón Umbilical/efectos adversos , Enfermedad Injerto contra Huésped/etiología , Humanos , Infecciones/etiología , Acondicionamiento Pretrasplante , Trasplantes
7.
Cancer Res ; 71(8): 2901-15, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21487043

RESUMEN

Primary myelofibrosis (PMF) is characterized by increased number of hematopoietic progenitors and a dysmegakaryopoiesis which supports the stromal reaction defining this disease. We showed that increased ligand (FL) levels in plasma, hematopoietic progenitors, and stromal cells from PMF patients were associated with upregulation of the cognate Flt3 receptor on megakaryocytic (MK) cells. This connection prompted us to study a functional role for the FL/Flt3 couple in PMF dysmegakaryopoiesis, as a route to reveal insights into pathobiology and therapy in this disease. Analysis of PMF CD34(+) and MK cell transcriptomes revealed deregulation of the mitogen-activated protein kinase (MAPK) pathway along with Flt3 expression. In PMF patients, a higher proportion of circulating Flt3(+)CD34(+)CD41(+) cells exhibited an increased MAPK effector phosphorylation independently of Jak2(V617F) mutation. Activation of FL/Flt3 axis in PMF MK cell cultures, in response to FL, induced activation of the p38-MAPK cascade, which is known to be involved in inflammation, also increasing expression of its target genes (NFATC4, p53, AP-1, IL-8). Inhibiting Flt3 or MAPK or especially p38 by chemical, antibody, or silencing strategies restored megakaryopoiesis and reduced phosphorylation of Flt3 and p38 pathway effectors, confirming the involvement of Flt3 in PMF dysmegakaryopoiesis via p38 activation. In addition, in contrast to healthy donors, MK cells derived from PMF CD34(+) cells exhibited an FL-induced migration that could be reversed by p38 inhibition. Taken together, our results implicate the FL/Flt3 ligand-receptor complex in PMF dysmegakaryopoiesis through persistent p38-MAPK activation, with implications for therapeutic prospects to correct altered megakaryopoiesis in an inflammatory context.


Asunto(s)
Megacariocitos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mielofibrosis Primaria/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Antígenos CD34/biosíntesis , Activación Enzimática , Células Madre Hematopoyéticas/enzimología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Leucocitos Mononucleares/enzimología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Sistema de Señalización de MAP Quinasas , Megacariocitos/enzimología , Megacariocitos/patología , Proteínas de la Membrana/sangre , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/farmacología , Fosforilación , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/enzimología , Células del Estroma/enzimología , Células del Estroma/metabolismo , Células del Estroma/patología , Tirosina Quinasa 3 Similar a fms/biosíntesis
8.
Stem Cells ; 27(10): 2552-62, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19650038

RESUMEN

Identification of prevalent specific markers is crucial to stem/progenitor cell purification. Determinants such as the surface antigens CD34 and CD38 are traditionally used to analyze and purify hematopoietic stem/progenitor cells (HSCs/HPCs). However, the variable expression of these membrane antigens poses some limitations to their use in HSC/HPC purification. Techniques based on drug/stain efflux through the ATP-binding cassette (ABC)G2 pump (side population [SP] phenotype) or on detection of aldehyde dehydrogenase (ALDH) activity have been independently developed and distinguish the SP and ALDH(Bright) (ALDH(Br)) cell subsets for their phenotype and proliferative capability. In this study, we developed a multiparametric flow cytometric method associating both SP and ALDH activities on human lineage negative (Lin(-)) bone marrow cells and sorted different cell fractions according to their SP/ALDH activity level. We find that Lin(-)CD34(+)CD38(Low/-) cells are found throughout the spectrum of ALDH expression and are enriched especially in ALDH(Br) cells when associated with SP functionality (SP/ALDH(Br) fraction). Furthermore, the SP marker identified G(0) cells in all ALDH fractions, allowing us to sort quiescent cells regardless of ALDH activity. Moreover, we show that, within the Lin(-)CD34(+)CD38(-)ALDH(Br) population, the SP marker identifies cells with higher primitive characteristics, in terms of stemness-related gene expression and in vitro and in vivo proliferative potential, than the Lin(-)CD34(+) CD38(-)ALDH(Br) main population cells. In conclusion, our study shows that the coexpression of SP and ALDH markers refines the Lin(-)CD34(+)CD38(-) hematopoietic compartment and identifies an SP/ALDH(Br) cell subset enriched in quiescent primitive HSCs/HPCs.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Antígenos de Superficie/metabolismo , Separación Celular/métodos , Citometría de Flujo/métodos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , ADP-Ribosil Ciclasa 1/análisis , ADP-Ribosil Ciclasa 1/metabolismo , Aldehído Deshidrogenasa/análisis , Animales , Antígenos CD34/análisis , Antígenos CD34/metabolismo , Antígenos de Superficie/análisis , Biomarcadores/análisis , Biomarcadores/metabolismo , Ciclo Celular/fisiología , Desdiferenciación Celular/genética , Linaje de la Célula , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Ratones , Nucleotidiltransferasas/análisis , Nucleotidiltransferasas/metabolismo
9.
Stem Cells ; 26(12): 3150-61, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18757300

RESUMEN

Cell cycle regulation plays a fundamental role in stem cell biology. A balance between quiescence and proliferation of hematopoietic stem cells in interaction with the microenvironment is critical for sustaining long-term hematopoiesis and for protection against stress. We analyzed the molecular mechanisms by which stromal cell-derived factor-1 (SDF-1) exhibited a cell cycle-promoting effect and interacted with transforming growth factor-beta (TGF-beta), which has negative effects on cell cycle orchestration of human hematopoietic CD34(+) progenitor cells. We demonstrated that a low concentration of SDF-1 modulated the expression of key cell cycle regulators such as cyclins, cyclin-dependent kinase inhibitors, and TGF-beta target genes, confirming its cell cycle-promoting effect. We showed that a cross-talk between SDF-1- and TGF-beta-related signaling pathways involving phosphatidylinositol 3-kinase (PI3K)/Akt phosphorylation participated in the control of CD34(+) cell cycling. We demonstrated a pivotal role of two downstream effectors of the PI3K/Akt pathway, FoxO3a and mammalian target of rapamycin, as connectors in the SDF-1-/TGF-beta-induced control of the cycling/quiescence switch and proposed a model integrating a dialogue between the two molecules in cell cycle progression. Our data shed new light on the signaling pathways involved in SDF-1 cell cycle-promoting activity and suggest that the balance between SDF-1- and TGF-beta-activated pathways is critical for the regulation of hematopoietic progenitor cell cycle status.


Asunto(s)
Quimiocina CXCL12/metabolismo , Factores de Transcripción Forkhead/metabolismo , Proteínas Quinasas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Antígenos CD34/biosíntesis , Ciclo Celular , Proteína Forkhead Box O3 , Hematopoyesis , Células Madre Hematopoyéticas/citología , Humanos , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Madre/citología , Serina-Treonina Quinasas TOR
10.
Blood ; 107(5): 2098-100, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16293597

RESUMEN

Most patients with polycythemia vera and half with idiopathic myelofibrosis and essential thrombocythemia have an acquired V617F mutation in JAK2. Using sensitive polymerase chain reaction (PCR)-based methods, we genotyped 152 patients with idiopathic myelofibrosis to establish whether there were differences in presentation and outcome between those with and those without the mutation. Patients positive for V617F had higher neutrophil and white cell counts (P = .02) than did patients negative for V617F, but other diagnostic features were comparable between the 2 groups. Patients positive for V617F were less likely to require blood transfusion during follow-up (P = .03). Despite this, patients positive for V617F had poorer overall survival, even after correction for confounding factors (P = .01).


Asunto(s)
Sustitución de Aminoácidos , Mutación Puntual , Policitemia Vera/genética , Mielofibrosis Primaria/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Trombocitemia Esencial/genética , Anciano , Transfusión Sanguínea , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Janus Quinasa 2 , Masculino , Persona de Mediana Edad , Policitemia Vera/mortalidad , Policitemia Vera/terapia , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/mortalidad , Mielofibrosis Primaria/terapia , Trombocitemia Esencial/complicaciones , Trombocitemia Esencial/mortalidad , Trombocitemia Esencial/terapia
11.
J Leukoc Biol ; 77(5): 634-43, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15673546

RESUMEN

Human CD34+ hematopoietic progenitors (HP) are mainly resident in adult bone marrow (BM). However, their recent revelation in nonhematopoietic tissues implies their circulation through peripheral blood (PB). The intimate mechanisms of this physiological process are not yet understood. Our results showed that steady-state CD34+ HP exhibit a differential phenotypic profile according to their BM versus PB localization. We demonstrated that this phenotype could be modulated by incubation in the presence of their counterpart mononuclear cells (MNC) through cell interactions and cytokine production. Such a modulation mainly concerns migration-mediated cytokine and chemokine receptors as well as some adhesion molecules and partly results from MNC specificity. These phenotypic profiles are associated with distinct cell-cycle position, cloning efficiency, and migration capacity of CD34+ cells from the different anatomical sources. We therefore propose a definition for a circulating versus resident CD34+ cell profile, which mostly depends on their cellular environment. We suggest that blood would represent a supply of cells for which phenotypic and functional characteristics would be a prerequisite for their bio-availability.


Asunto(s)
Sangre , Médula Ósea , Comunicación Celular , Diferenciación Celular/fisiología , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Antígenos CD34 , Células Cultivadas , Quimiotaxis de Leucocito/fisiología , Ensayo de Unidades Formadoras de Colonias , Humanos , Inmunofenotipificación
12.
Blood ; 105(2): 464-73, 2005 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15454487

RESUMEN

Myeloproliferation, myelofibrosis, and neoangiogenesis are the 3 major intrinsic pathophysiologic features of myeloid metaplasia with myelofibrosis (MMM). The myeloproliferation is characterized by an increased number of circulating CD34+ progenitors with the prominent amplification of dystrophic megakaryocytic (MK) cells and myeloid metaplasia in the spleen and liver. The various biologic activities of interleukin 8 (IL-8) in hematopoietic progenitor proliferation and mobilization as well as in neoangiogenesis prompted us to analyze its potential role in MMM. We showed that the level of IL-8 chemokine is significantly increased in the serum of patients and that various hematopoietic cells, including platelets, participate in its production. In vitro inhibition of autocrine IL-8 expressed by CD34+ cells with either a neutralizing or an antisense anti-IL-8 treatment increases the proliferation of MMM CD34(+)-derived cells and stimulates their MK differentiation. Moreover, addition of neutralizing anti-IL-8 receptor (CXC chemokine receptor 1 [CXCR1] or 2 [CXCR2]) antibodies to MMM CD34+ cells cultured under MK liquid culture conditions increases the proliferation and differentiation of MMM CD41+ MK cells and restores their polyploidization. Our results suggest that IL-8 and its receptors participate in the altered MK growth that features MMM and open new therapeutic prospects for this still incurable disease.


Asunto(s)
Interleucina-8/sangre , Megacariocitos/citología , Mielofibrosis Primaria/fisiopatología , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/metabolismo , Anciano , Anticuerpos/farmacología , Antígenos CD34/metabolismo , Plaquetas/fisiología , Diferenciación Celular/inmunología , División Celular/inmunología , Expresión Génica/inmunología , Humanos , Interleucina-8/genética , Megacariocitos/fisiología , Persona de Mediana Edad , Pruebas de Neutralización , Ploidias , Mielofibrosis Primaria/metabolismo , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/inmunología , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...