Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Blood ; 141(8): 917-929, 2023 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-36356299

RESUMEN

Mutant calreticulin (CALR) proteins resulting from a -1/+2 frameshifting mutation of the CALR exon 9 carry a novel C-terminal amino acid sequence and drive the development of myeloproliferative neoplasms (MPNs). Mutant CALRs were shown to interact with and activate the thrombopoietin receptor (TpoR/MPL) in the same cell. We report that mutant CALR proteins are secreted and can be found in patient plasma at levels up to 160 ng/mL, with a mean of 25.64 ng/mL. Plasma mutant CALR is found in complex with soluble transferrin receptor 1 (sTFR1) that acts as a carrier protein and increases mutant CALR half-life. Recombinant mutant CALR proteins bound and activated the TpoR in cell lines and primary megakaryocytic progenitors from patients with mutated CALR in which they drive thrombopoietin-independent colony formation. Importantly, the CALR-sTFR1 complex remains functional for TpoR activation. By bioluminescence resonance energy transfer assay, we show that mutant CALR proteins produced in 1 cell can specifically interact in trans with the TpoR on a target cell. In comparison with cells that only carry TpoR, cells that carry both TpoR and mutant CALR are hypersensitive to exogenous mutant CALR proteins and respond to levels of mutant CALR proteins similar to those in patient plasma. This is consistent with CALR-mutated cells that expose TpoR carrying immature N-linked sugars at the cell surface. Thus, secreted mutant CALR proteins will act more specifically on the MPN clone. In conclusion, a chaperone, CALR, can turn into a rogue cytokine through somatic mutation of its encoding gene.


Asunto(s)
Trastornos Mieloproliferativos , Neoplasias , Humanos , Citocinas/metabolismo , Calreticulina/genética , Trastornos Mieloproliferativos/genética , Mutación , Factores Inmunológicos , Janus Quinasa 2/genética
2.
Blood ; 135(12): 948-953, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-31978223

RESUMEN

Mutations in the MPL gene encoding the human thrombopoietin receptor (TpoR) drive sporadic and familial essential thrombocythemias (ETs). We identified 2 ET patients harboring double mutations in cis in MPL, namely, L498W-H499C and H499Y-S505N. Using biochemical and signaling assays along with partial saturation mutagenesis, we showed that L498W is an activating mutation potentiated by H499C and that H499C and H499Y enhance the activity of the canonical S505N mutation. L498W and H499C can activate a truncated TpoR mutant, which lacks the extracellular domain, indicating these mutations act on the transmembrane (TM) cytosolic domain. Using a protein complementation assay, we showed that L498W and H499C strongly drive dimerization of TpoR. Activation by tryptophan substitution is exquisitely specific for position 498. Using structure-guided mutagenesis, we identified upstream amino acid W491 as a key residue required for activation by L498W or canonical activating mutations such as S505N and W515K, as well as by eltrombopag. Structural data point to a common dimerization and activation path for TpoR via its TM domain that is shared between the small-molecule agonist eltrombopag and canonical and novel activating TpoR mutations that all depend on W491, a potentially accessible extracellular residue that could become a target for therapeutic intervention.


Asunto(s)
Benzoatos/farmacología , Predisposición Genética a la Enfermedad , Hidrazinas/farmacología , Mutación , Pirazoles/farmacología , Receptores de Trombopoyetina/agonistas , Receptores de Trombopoyetina/genética , Trombocitemia Esencial/genética , Alelos , Sustitución de Aminoácidos , Línea Celular , Estudios de Asociación Genética , Humanos , Fenotipo , Transducción de Señal/efectos de los fármacos , Trombocitemia Esencial/diagnóstico , Trombocitemia Esencial/metabolismo
3.
Proc Natl Acad Sci U S A ; 116(4): 1241-1250, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30626647

RESUMEN

Neutropenia represents an important problem in patients with genetic deficiency in either the glucose-6-phosphate transporter of the endoplasmic reticulum (G6PT/SLC37A4) or G6PC3, an endoplasmic reticulum phosphatase homologous to glucose-6-phosphatase. While affected granulocytes show reduced glucose utilization, the underlying mechanism is unknown and causal therapies are lacking. Using a combination of enzymological, cell-culture, and in vivo approaches, we demonstrate that G6PT and G6PC3 collaborate to destroy 1,5-anhydroglucitol-6-phosphate (1,5AG6P), a close structural analog of glucose-6-phosphate and an inhibitor of low-KM hexokinases, which catalyze the first step in glycolysis in most tissues. We show that 1,5AG6P is made by phosphorylation of 1,5-anhydroglucitol, a compound normally present in human plasma, by side activities of ADP-glucokinase and low-KM hexokinases. Granulocytes from patients deficient in G6PC3 or G6PT accumulate 1,5AG6P to concentrations (∼3 mM) that strongly inhibit hexokinase activity. In a model of G6PC3-deficient mouse neutrophils, physiological concentrations of 1,5-anhydroglucitol caused massive accumulation of 1,5AG6P, a decrease in glucose utilization, and cell death. Treating G6PC3-deficient mice with an inhibitor of the kidney glucose transporter SGLT2 to lower their blood level of 1,5-anhydroglucitol restored a normal neutrophil count, while administration of 1,5-anhydroglucitol had the opposite effect. In conclusion, we show that the neutropenia in patients with G6PC3 or G6PT mutations is a metabolite-repair deficiency, caused by a failure to eliminate the nonclassical metabolite 1,5AG6P.


Asunto(s)
Antiportadores/metabolismo , Glucosa-6-Fosfatasa/metabolismo , Glucosa/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Neutropenia/metabolismo , Fosforilación/fisiología , Animales , Muerte Celular/fisiología , Línea Celular , Retículo Endoplásmico/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/metabolismo , Ratas Wistar
4.
Blood ; 133(25): 2669-2681, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-30902807

RESUMEN

Calreticulin (CALR) +1 frameshift mutations in exon 9 are prevalent in myeloproliferative neoplasms. Mutant CALRs possess a new C-terminal sequence rich in positively charged amino acids, leading to activation of the thrombopoietin receptor (TpoR/MPL). We show that the new sequence endows the mutant CALR with rogue chaperone activity, stabilizing a dimeric state and transporting TpoR and mutants thereof to the cell surface in states that would not pass quality control; this function is absolutely required for oncogenic transformation. Mutant CALRs determine traffic via the secretory pathway of partially immature TpoR, as they protect N117-linked glycans from further processing in the Golgi apparatus. A number of engineered or disease-associated TpoRs such as TpoR/MPL R102P, which causes congenital thrombocytopenia, are rescued for traffic and function by mutant CALRs, which can also overcome endoplasmic reticulum retention signals on TpoR. In addition to requiring N-glycosylation of TpoR, mutant CALRs require a hydrophobic patch located in the extracellular domain of TpoR to induce TpoR thermal stability and initial intracellular activation, whereas full activation requires cell surface localization of TpoR. Thus, mutant CALRs are rogue chaperones for TpoR and traffic-defective TpoR mutants, a function required for the oncogenic effects.


Asunto(s)
Calreticulina/genética , Calreticulina/metabolismo , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Receptores de Trombopoyetina/metabolismo , Animales , Humanos , Ratones , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mutación , Transporte de Proteínas/fisiología
5.
J Pediatr Hematol Oncol ; 43(1): e51-e55, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33122584

RESUMEN

Hypereosinophilia (HE) is rare but often secondary to a nonhematologic disease such as allergic disorders and parasitic infections. HE can also be associated with hematologic malignancies and be the result of a clonal proliferation or reactive to another hematologic condition. Association of HE with acute lymphoblastic leukemia (ALL) is rare in children. We reported a case of a teenager presented with HE secondary to B-ALL who experienced severe cardiac complications with severe absolute eosinophil count. We compared his clinical evolution with other published cases and we reported 2 mutations linked to B-ALL never described before in this context.


Asunto(s)
Cardiomiopatías/patología , Síndrome Hipereosinofílico/patología , Linfoma de Células B/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Adolescente , Cardiomiopatías/etiología , Humanos , Síndrome Hipereosinofílico/etiología , Linfoma de Células B/complicaciones , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Pronóstico
6.
Clin Chem Lab Med ; 58(12): 2141-2150, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33064667

RESUMEN

Objectives As severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2) pandemic is increasing its victims on a global scale with recurring outbreaks, it remains of outmost importance to rapidly identify people requiring an intensive care unit (ICU) hospitalization. The aim of this study was to identify Coronavirus Disease 2019 (COVID-19) biomarkers, to investigate their correlation with disease severity and to evaluate their usefulness for follow-up. Methods Fifty patients diagnosed with SARS-Cov-2 were included in March 2020. Clinical and biological data were collected at admission, during hospitalization and one month after discharge. Patients were divided into two severity groups: non-ICU (28) and ICU and/or death (22) to stratify the risk. Results Blood parameters in COVID-19 patients at admission showed increased C-reactive protein (CRP) (100%), ferritin (92%), lactate dehydrogenase (LDH) (80%), white blood cell (WBC) count (26%) with lymphopenia (52%) and eosinopenia (98%). There were significant differences in levels of CRP, ferritin, D-dimers, fibrinogen, lymphocyte count, neutrophil count and neutrophil-to-lymphocyte ratio (NLR) among the two severity groups. Mapping of biomarker's kinetics distinguished early and late parameters. CRP, ferritin, LDH, lymphopenia and eosinopenia were present upon admission with a peak at the first week. Late biomarkers such as anemia, neutrophilia and elevated liver biomarkers appeared after one week with a peak at three weeks of hospitalization. Conclusions We confirmed that high-values of CRP, NLR, D-dimers, ferritin as well as lymphopenia and eosinopenia were consistently found and are good markers for risk stratification. Kinetics of these biomarkers correlate well with COVID-19 severity. Close monitoring of early and late biomarkers is crucial in the management of critical patients to avoid preventable deaths.


Asunto(s)
Infecciones por Coronavirus/diagnóstico , Neumonía Viral/diagnóstico , Adolescente , Adulto , Anciano , Biomarcadores/sangre , Recuento de Células Sanguíneas , COVID-19 , Infecciones por Coronavirus/sangre , Femenino , Estudios de Seguimiento , Hemostasis , Humanos , Masculino , Persona de Mediana Edad , Pandemias , Neumonía Viral/sangre , Pronóstico , Adulto Joven
7.
Blood ; 128(26): 3146-3158, 2016 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-28034873

RESUMEN

The mechanisms behind the hereditary thrombocytosis induced by the thrombopoietin (THPO) receptor MPL P106L mutant remain unknown. A complete trafficking defect to the cell surface has been reported, suggesting either weak constitutive activity or nonconventional THPO-dependent mechanisms. Here, we report that the thrombocytosis phenotype induced by MPL P106L belongs to the paradoxical group, where low MPL levels on platelets and mature megakaryocytes (MKs) lead to high serum THPO levels, whereas weak but not absent MPL cell-surface localization in earlier MK progenitors allows response to THPO by signaling and amplification of the platelet lineage. MK progenitors from patients showed no spontaneous growth and responded to THPO, and MKs expressed MPL on their cell surface at low levels, whereas their platelets did not respond to THPO. Transduction of MPL P106L in CD34+ cells showed that this receptor was more efficiently localized at the cell surface on immature than on mature MKs, explaining a proliferative response to THPO of immature cells and a defect in THPO clearance in mature cells. In a retroviral mouse model performed in Mpl-/- mice, MPL P106L could induce a thrombocytosis phenotype with high circulating THPO levels. Furthermore, we could select THPO-dependent cell lines with more cell-surface MPL P106L localization that was detected by flow cytometry and [125I]-THPO binding. Altogether, these results demonstrate that MPL P106L is a receptor with an incomplete defect in trafficking, which induces a low but not absent localization of the receptor on cell surface and a response to THPO in immature MK cells.


Asunto(s)
Membrana Celular/metabolismo , Mutación/genética , Receptores de Trombopoyetina/genética , Trombocitosis/genética , Trombocitosis/patología , Animales , Línea Celular , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Familia , Femenino , Humanos , Masculino , Megacariocitos/metabolismo , Ratones , Linaje , Transporte de Proteínas , Receptores de Trombopoyetina/metabolismo , Retroviridae/metabolismo , Transducción Genética
8.
Blood ; 127(10): 1325-35, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26668133

RESUMEN

Mutations in the calreticulin gene (CALR) represented by deletions and insertions in exon 9 inducing a -1/+2 frameshift are associated with a significant fraction of myeloproliferative neoplasms (MPNs). The mechanisms by which CALR mutants induce MPN are unknown. Here, we show by transcriptional, proliferation, biochemical, and primary cell assays that the pathogenic CALR mutants specifically activate the thrombopoietin receptor (TpoR/MPL). No activation is detected with a battery of type I and II cytokine receptors, except granulocyte colony-stimulating factor receptor, which supported only transient and weak activation. CALR mutants induce ligand-independent activation of JAK2/STAT/phosphatydylinositol-3'-kinase (PI3-K) and mitogen-activated protein (MAP) kinase pathways via TpoR, and autonomous growth in Ba/F3 cells. In these transformed cells, no synergy is observed between JAK2 and PI3-K inhibitors in inhibiting cytokine-independent proliferation, thus showing a major difference from JAK2V617F cells where such synergy is strong. TpoR activation was dependent on its extracellular domain and its N-glycosylation, especially at N117. The glycan binding site and the novel C-terminal tail of the mutant CALR proteins were required for TpoR activation. A soluble form of TpoR was able to prevent activation of full-length TpoR provided that it was N-glycosylated. By confocal microscopy and subcellular fractionation, CALR mutants exhibit different intracellular localization from that of wild-type CALR. Finally, knocking down either MPL/TpoR or JAK2 in megakaryocytic progenitors from patients carrying CALR mutations inhibited cytokine-independent megakaryocytic colony formation. Taken together, our study provides a novel signaling paradigm, whereby a mutated chaperone constitutively activates cytokine receptor signaling.


Asunto(s)
Calreticulina/metabolismo , Neoplasias Hematológicas/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas Musculares/metabolismo , Mutación , Trastornos Mieloproliferativos/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Trombopoyetina/metabolismo , Animales , Calreticulina/genética , Línea Celular Tumoral , Glicosilación , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Proteínas con Dominio LIM/genética , Ratones , Proteínas Musculares/genética , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Proteínas de Neoplasias/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/genética , Transporte de Proteínas/genética , Receptores de Trombopoyetina/genética , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal/genética
9.
Blood ; 127(3): 333-42, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26450985

RESUMEN

Mutations in signaling molecules of the cytokine receptor axis play a central role in myeloproliferative neoplasm (MPN) pathogenesis. Polycythemia vera is mainly related to JAK2 mutations, whereas a wider mutational spectrum is detected in essential thrombocythemia (ET) with mutations in JAK2, the thrombopoietin (TPO) receptor (MPL), and the calreticulin (CALR) genes. Here, we studied the mutational profile of 17 ET patients negative for JAK2V617F, MPLW515K/L, and CALR mutations, using whole-exome sequencing and next-generation sequencing (NGS) targeted on JAK2 and MPL. We found several signaling mutations including JAK2V617F at very low allele frequency, 1 homozygous SH2B3 mutation, 1 MPLS505N, 1 MPLW515R, and 2 MPLS204P mutations. In the remaining patients, 4 presented a clonal and 7 a polyclonal hematopoiesis, suggesting that certain triple-negative ETs are not MPNs. NGS on 26 additional triple-negative ETs detected only 1 MPLY591N mutation. Functional studies on MPLS204P and MPLY591N revealed that they are weak gain-of-function mutants increasing MPL signaling and conferring either TPO hypersensitivity or independence to expressing cells, but with a low efficiency. Further studies should be performed to precisely determine the frequency of MPLS204 and MPLY591 mutants in a bigger cohort of MPN.


Asunto(s)
Mutación , Receptores de Trombopoyetina/genética , Trombocitemia Esencial/genética , Sustitución de Aminoácidos , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Codón , Estudios de Cohortes , Hibridación Genómica Comparativa , Citocinas/farmacología , Análisis Mutacional de ADN , Exoma , Genotipo , Granulocitos/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Janus Quinasa 2/genética , Transporte de Proteínas , Receptores de Trombopoyetina/metabolismo , Trombocitemia Esencial/metabolismo
10.
J Biol Chem ; 291(6): 2974-87, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26627830

RESUMEN

Ligand binding to the extracellular domain of the thrombopoietin receptor (TpoR) imparts a specific orientation on the transmembrane (TM) and intracellular domains of the receptors that is required for physiologic activation via receptor dimerization. To map the inactive and active dimeric orientations of the TM helices, we performed asparagine (Asn)-scanning mutagenesis of the TM domains of the murine and human TpoR. Substitution of Asn at only one position (S505N) activated the human receptor, whereas Asn substitutions at several positions activated the murine receptor. Second site mutational studies indicate that His(499) near the N terminus of the TM domain is responsible for protecting the human receptor from activation by Asn mutations. Structural studies reveal that the sequence preceding His(499) is helical in the murine receptor but non-helical in peptides corresponding to the TM domain of the inactive human receptor. The activating S505N mutation and the small molecule agonist eltrombopag both induce helix in this region of the TM domain and are associated with dimerization and activation of the human receptor. Thus, His(499) regulates the activation of human TpoR and provides additional protection against activating mutations, such as oncogenic Asn mutations in the TM domain.


Asunto(s)
Proteínas Proto-Oncogénicas/metabolismo , Receptores de Trombopoyetina/metabolismo , Animales , Asparagina/genética , Asparagina/metabolismo , Línea Celular , Histidina/genética , Histidina/metabolismo , Humanos , Ratones , Mutación Missense , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/genética , Receptores de Trombopoyetina/genética
11.
Proc Natl Acad Sci U S A ; 110(7): 2540-5, 2013 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-23359689

RESUMEN

Dimerization of single-pass membrane receptors is essential for activation. In the human thrombopoietin receptor (TpoR), a unique amphipathic RWQFP motif separates the transmembrane (TM) and intracellular domains. Using a combination of mutagenesis, spectroscopy, and biochemical assays, we show that W515 of this motif impairs dimerization of the upstream TpoR TM helix. TpoR is unusual in that a specific residue is required for this inhibitory function, which prevents receptor self-activation. Mutations as diverse as W515K and W515L cause oncogenic activation of TpoR and lead to human myeloproliferative neoplasms. Two lines of evidence support a general mechanism in which W515 at the intracellular juxtamembrane boundary inhibits dimerization of the TpoR TM helix by increasing the helix tilt angle relative to the membrane bilayer normal, which prevents the formation of stabilizing TM dimer contacts. First, measurements using polarized infrared spectroscopy show that the isolated TM domain of the active W515K mutant has a helix tilt angle closer to the bilayer normal than that of the wild-type receptor. Second, we identify second-site R514W and Q516W mutations that reverse dimerization and tilt angle changes induced by the W515K and W515L mutations. The second-site mutations prevent constitutive activation of TpoR W515K/L, while preserving ligand-induced signaling. The ability of tryptophan to influence the angle and dimerization of the TM helix in wild-type TpoR and in the second-site revertants is likely associated with its strong preference to be buried in the headgroup region of membrane bilayers.


Asunto(s)
Modelos Moleculares , Conformación Proteica , Receptores de Trombopoyetina/química , Receptores de Trombopoyetina/metabolismo , Triptófano/metabolismo , Secuencias de Aminoácidos/genética , Animales , Secuencia de Bases , Western Blotting , Línea Celular , Clonación Molecular , Dimerización , Citometría de Flujo , Prueba de Complementación Genética , Humanos , Luciferasas , Espectroscopía de Resonancia Magnética , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Receptores de Trombopoyetina/genética , Análisis de Secuencia de ADN , Espectrofotometría Infrarroja , Ultracentrifugación
12.
EMBO J ; 30(21): 4398-413, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21892137

RESUMEN

Ligand binding to the thrombopoietin receptor is thought to stabilize an active receptor dimer that regulates megakaryocyte differentiation and platelet formation, as well as haematopoietic stem cell renewal. By fusing a dimeric coiled coil in all seven possible orientations to the thrombopoietin receptor transmembrane (TM)-cytoplasmic domains, we show that specific biological effects and in vivo phenotypes are imparted by distinct dimeric orientations, which can be visualized by cysteine mutagenesis and crosslinking. Using functional assays and computational searches, we identify one orientation that represents the inactive dimeric state and another similar to a physiologically activated receptor. Several other dimeric orientations are identified that induce proliferation and in vivo myeloproliferative and myelodysplastic disorders, indicating the receptor can signal from several dimeric interfaces. The set of dimeric thrombopoietin receptors with different TM orientations may offer new insights into the activation of distinct signalling pathways by a single receptor and suggests that subtle differences in cytokine receptor dimerization provide a new layer of signalling regulation that is relevant for disease.


Asunto(s)
Multimerización de Proteína/fisiología , Receptores de Trombopoyetina/química , Receptores de Trombopoyetina/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Dominios y Motivos de Interacción de Proteínas/fisiología , Mapas de Interacción de Proteínas , Multimerización de Proteína/genética , Receptores de Trombopoyetina/genética , Receptores de Trombopoyetina/fisiología , Transducción de Señal/fisiología , Estereoisomerismo
14.
Blood ; 119(20): 4625-35, 2012 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-22378845

RESUMEN

The constitutively active JAK2 V617F mutant is the major determinant of human myeloproliferative neoplasms (MPNs). We show that coexpression of murine JAK2 V617F and the murine thrombopoietin (Tpo) receptor (TpoR, c-MPL) in hematopoietic cell lines or heterozygous knock-in of JAK2 V617F in mice leads to down-modulation of TpoR levels. Enhanced TpoR ubiquitinylation, proteasomal degradation, reduced recycling, and maturation are induced by the constitutive JAK2 V617F activity. These effects can be prevented in cell lines by JAK2 and proteasome inhibitors. Restoration of TpoR levels by inhibitors could be detected in platelets from JAK2 inhibitor-treated myelofibrosis patients that express the JAK2 V617F mutant, and in platelets from JAK2 V617F knock-in mice that were treated in vivo with JAK2 or proteasome inhibitors. In addition, we show that Tpo can induce both proliferative and antiproliferative effects via TpoR at low and high JAK2 activation levels, respectively, or on expression of JAK2 V617F. The antiproliferative signaling and receptor down-modulation by JAK2 V617F were dependent on signaling via TpoR cytosolic tyrosine 626. We propose that selection against TpoR antiproliferative signaling occurs by TpoR down-modulation and that restoration of down-modulated TpoR levels could become a biomarker for the treatment of MPNs.


Asunto(s)
Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/fisiología , Inhibidores de Proteasoma , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Trombopoyetina/genética , Sustitución de Aminoácidos/genética , Sustitución de Aminoácidos/fisiología , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Células HEK293 , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense/fisiología , Fenilalanina/genética , Receptor Cross-Talk/efectos de los fármacos , Receptor Cross-Talk/fisiología , Receptores de Trombopoyetina/metabolismo , Transducción de Señal/efectos de los fármacos , Valina/genética
15.
J Cell Mol Med ; 17(11): 1397-409, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24251790

RESUMEN

Current JAK2 inhibitors used for myeloproliferative neoplasms (MPN) treatment are not specific enough to selectively suppress aberrant JAK2 signalling and preserve physiological JAK2 signalling. We tested whether combining a JAK2 inhibitor with a series of serine threonine kinase inhibitors, targeting nine signalling pathways and already used in clinical trials, synergized in inhibiting growth of haematopoietic cells expressing mutant and wild-type forms of JAK2 (V617F) or thrombopoietin receptor (W515L). Out of 15 kinase inhibitors, the ZSTK474 phosphatydylinositol-3'-kinase (PI3K) inhibitor molecule showed strong synergic inhibition by Chou and Talalay analysis with JAK2 and JAK2/JAK1 inhibitors. Other pan-class I, but not gamma or delta specific PI3K inhibitors, also synergized with JAK2 inhibitors. Synergy was not observed in Bcr-Abl transformed cells. The best JAK2/JAK1 and PI3K inhibitor combination pair (ruxolitinib and GDC0941) reduces spleen weight in nude mice inoculated with Ba/F3 cells expressing TpoR and JAK2 V617F. It also exerted strong inhibitory effects on erythropoietin-independent erythroid colonies from MPN patients and JAK2 V617F knock-in mice, where at certain doses, a preferential inhibition of JAK2 V617F mutated progenitors was detected. Our data support the use of a combination of JAK2 and pan-class I PI3K inhibitors in the treatment of MPNs.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Janus Quinasa 2/antagonistas & inhibidores , Trastornos Mieloproliferativos/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Técnicas de Sustitución del Gen , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/enzimología , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Ratones , Ratones Desnudos , Ratones Transgénicos , Mutación Missense , Trastornos Mieloproliferativos/enzimología , Trasplante de Neoplasias , Nitrilos , Fosfatidilinositol 3-Quinasas/metabolismo , Pirazoles/administración & dosificación , Pirimidinas , Pirrolidinas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Sulfonamidas/administración & dosificación , Triazinas/administración & dosificación , Células Tumorales Cultivadas
16.
Elife ; 122023 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-37338955

RESUMEN

Dimerization of the thrombopoietin receptor (TpoR) is necessary for receptor activation and downstream signaling through activated Janus kinase 2. We have shown previously that different orientations of the transmembrane (TM) helices within a receptor dimer can lead to different signaling outputs. Here we addressed the structural basis of activation for receptor mutations S505N and W515K that induce myeloproliferative neoplasms. We show using in vivo bone marrow reconstitution experiments that ligand-independent activation of TpoR by TM asparagine (Asn) substitutions is proportional to the proximity of the Asn mutation to the intracellular membrane surface. Solid-state NMR experiments on TM peptides indicate a progressive loss of helical structure in the juxtamembrane (JM) R/KWQFP motif with proximity of Asn substitutions to the cytosolic boundary. Mutational studies in the TpoR cytosolic JM region show that loss of the helical structure in the JM motif by itself can induce activation, but only when localized to a maximum of six amino acids downstream of W515, the helicity of the remaining region until Box 1 being required for receptor function. The constitutive activation of TpoR mutants S505N and W515K can be inhibited by rotation of TM helices within the TpoR dimer, which also restores helicity around W515. Together, these data allow us to develop a general model for activation of TpoR and explain the critical role of the JM W515 residue in the regulation of the activity of the receptor.


Asunto(s)
Receptores de Trombopoyetina , Transducción de Señal , Receptores de Trombopoyetina/genética , Receptores de Trombopoyetina/metabolismo , Línea Celular , Mutación , Estructura Secundaria de Proteína , Transducción de Señal/genética
17.
Int J Lab Hematol ; 43(3): 450-457, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33185328

RESUMEN

BACKGROUND: Evaluation of an individual's thrombin-generating capacity enables a global assessment of the coagulation cascade and is therefore thought to better reflect the clotting function of blood. However, the lack of standardization still hampers the use in routine clinical practice. METHODS: Nineteen healthy subjects were sampled once a week for 5 consecutive weeks. Thrombin generation assay (TGA) was performed in duplicate by calibrated automated thrombogram (CAT) on platelet poor plasma with and without thrombomodulin. After exclusion of outliers, a nested analysis of variance (ANOVA) was performed to evaluate the biological variability (BV) results. Analytical variation (CVA ), within-individual variation (CVI ), between-individual variation (CVG ), index of individuality (II), and reference change value (RCV) were calculated. RESULTS: All parameters taken together, the CVA, CVI , and CVG without TM, ranged from 2.8% to 6.5%, from 4.1% to 13.3% and from 10.4% to 28.4%, respectively. For TG with TM, CVI and CVG were higher and ranged from 5.0% to 18.1% and from 14.9% to 35.3%, respectively. For endogenous thrombin potential (ETP), a CVI of 4.1% and CVG of 10.4% were obtained without addition of thrombomodulin (TM). With addition of TM, both CVI and CVG were higher: 14.0% and 34.8%, respectively. The II was low and the RCV ranged from 17.2% to 50.4%. CONCLUSION: CAT parameters are highly individualized and population-based reference values could be called into question. The assessment of BV and RCV for thrombin generation assays could optimize interpretation of serial patient results and guide setting of analytical specification goals.


Asunto(s)
Pruebas de Coagulación Sanguínea , Trombina/análisis , Adulto , Pruebas de Coagulación Sanguínea/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Plasma/metabolismo , Valores de Referencia , Trombina/metabolismo , Trombomodulina/metabolismo , Adulto Joven
18.
J Virol Methods ; 295: 114197, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34033854

RESUMEN

OBJECTIVES: The SARS-CoV-2 pandemic has created an unprecedented need for rapid large-scale diagnostic testing to prompt clinical and public health interventions. Currently, several quantitative reverse-transcription polymerase chain reaction (RT-qPCR) assays recommended by the World Health Organization are being used by clinical and public health laboratories and typically target regions of the RNA-dependent RNA polymerase (RdRp), envelope (E) and nucleocapsid (N) coding region. However, it is currently unclear if results from different tests are comparable. This study aimed to clarify the clinical performances of the primer/probe sets designed by US CDC and Charité/Berlin to help clinical laboratories in assay selection for SARS-CoV-2 routine detection. METHODS: We compared the clinical performances of the recommended primer/probe sets using one hundred nasopharyngeal swab specimens from patients who were clinically diagnosed with COVID-19. An additional 30 "pre-intervention screening" samples from patients who were not suspected of COVID-19 were also included in this study. We also performed sequence alignment between 31064 European SARS-CoV-2 and variants of concern genomes and the recommended primer/probe sets. RESULTS: The present study demonstrates substantial differences in SARS-CoV-2 RNA detection sensitivity among the primer/probe sets recommended by the World Health Organization especially for low-level viral loads. The alignment of thousands of SARS-CoV-2 sequences reveals that the genetic diversity remains relatively low at the primer/probe binding sites. However, multiple nucleotide mismatches might contribute to false negatives. CONCLUSION: An understanding of the limitations depending on the targeted genes and primer/probe sets may influence the selection of molecular detection assays by clinical laboratories.


Asunto(s)
Cartilla de ADN/genética , Genoma Viral/genética , SARS-CoV-2/aislamiento & purificación , COVID-19/diagnóstico , COVID-19/virología , Prueba de Ácido Nucleico para COVID-19 , Coronavirus/genética , Humanos , ARN Viral/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , SARS-CoV-2/genética , Sensibilidad y Especificidad , Alineación de Secuencia , Carga Viral , Proteínas Virales/genética
19.
QJM ; 2020 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-31943120

RESUMEN

BACKGROUND: Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening disorder characterized by hyperimmune response. The mortality is high despite progress being made in the diagnosis and treatment of the disease. AIM: This review aimed to update knowledge on adult HLH pathophysiology, identifiy the numerous causes, and help clinicians make early diagnosis and initiate treatment. DESIGN AND METHODS: Using Embase, we searched relevant articles published from January 1, 2010 to October 31, 2019, with the MESH term « hemophagocytic lymphohistiocytosis; macrophagic activation syndrome, adult ¼. RESULTS: The mean age at presentation is about 50 years, with a male predominance. The most frequent disease associations are haematological diseases, viral or bacterial infections, and autoimmune diseases. The pathophysiologic mechanism is probably the combination of inherited genetic mutations and extrinsic triggers. The mortality rate is 26.5% to 74.8%. H-score is more efficient than HLH-2004 criteria to identify HLH, with diagnostic sensitivity and specificity 90% and 79%, respectively.18F-FDG PET/CT is potentially useful for detecting underlying disease and the extent of secondary HLH. Disease-specific treatment should be given as soon as possible. Treatment with corticosteroids combined or not with etoposide is the mainstay of treatment. Monoclonal antibodies and JAK pathway inhibitors show promise of being effective. CONCLUSION: In adult HLH, infectious diseases, autoimmune disease and malignancy should be suspected so that disease-specific treatment can be given promptly. Treatment with corticosteroids combined or not with etoposide is the mainstay of treatment, but new therapies show promise of being effective.

20.
Aging (Albany NY) ; 12(20): 19911-19922, 2020 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-33104521

RESUMEN

Telomeres are non-coding DNA sequences that protect chromosome ends and shorten with age. Short telomere length (TL) is associated with chronic diseases and immunosenescence. The main risk factor for mortality of coronavirus disease 2019 (COVID-19) is older age, but outcome is very heterogeneous among individuals of the same age group. Therefore, we hypothesized that TL influences COVID-19-related outcomes. In a prospective study, we measured TL by Flow-FISH in 70 hospitalized COVID-19 patients and compared TL distribution with our reference cohort of 491 healthy volunteers. We also correlated TL with baseline clinical and biological parameters. We stained autopsy lung tissue from six non-survivor COVID-19 patients to detect senescence-associated ß-galactosidase activity, a marker of cellular aging. We found a significantly higher proportion of patients with short telomeres (<10th percentile) in the COVID-19 patients as compared to the reference cohort (P<0.001). Short telomeres were associated with a higher risk of critical disease, defined as admission to intensive care unit (ICU) or death without ICU. TL was negatively correlated with C-reactive protein and neutrophil-to-lymphocyte ratio. Finally, lung tissue from patients with very short telomeres exhibit signs of senescence in structural and immune cells. Our results suggest that TL influences the severity of the disease.


Asunto(s)
Infecciones por Coronavirus , Pandemias , Neumonía Viral , Homeostasis del Telómero , Telómero , Adulto , Anciano , Anciano de 80 o más Años , COVID-19 , Senescencia Celular , Femenino , Humanos , Pulmón/patología , Masculino , Persona de Mediana Edad , Estudios Prospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA