Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Int J Mol Sci ; 24(12)2023 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-37373206

RESUMEN

For targeted protein panels, the ability to specifically assay post-translational modifications (PTMs) in a quantitative, sensitive, and straightforward manner would substantially advance biological and pharmacological studies. The present study highlights the effectiveness of the Affi-BAMS™ epitope-directed affinity bead capture/MALDI MS platform for quantitatively defining complex PTM marks of H3 and H4 histones. Using H3 and H4 histone peptides and isotopically labelled derivatives, this affinity bead and MALDI MS platform achieves a range of >3 orders of magnitude with a technical precision CV of <5%. Using nuclear cellular lysates, Affi-BAMS PTM-peptide capture resolves heterogeneous histone N-terminal PTMs with as little as 100 µg of starting material. In an HDAC inhibitor and MCF7 cell line model, the ability to monitor dynamic histone H3 acetylation and methylation events is further demonstrated (including SILAC quantification). Affi-BAMS (and its capacity for the multiplexing of samples and target PTM-proteins) thus provides a uniquely efficient and effective approach for analyzing dynamic epigenetic histone marks, which is critical for the regulation of chromatin structure and gene expression.


Asunto(s)
Histonas , Proteómica , Histonas/metabolismo , Espectrometría de Masas en Tándem , Procesamiento Proteico-Postraduccional , Código de Histonas , Péptidos/metabolismo , Acetilación
2.
Int J Mol Sci ; 21(6)2020 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-32188029

RESUMEN

The ability to quantitatively probe diverse panels of proteins and their post-translational modifications (PTMs) across multiple samples would aid a broad spectrum of biological, biochemical and pharmacological studies. We report a novel, microarray analytical technology that combines immuno-affinity capture with Matrix Assisted Laser Desorption Ionization Mass Spectrometry (MALDI MS), which is capable of supporting highly multiplexed, targeted proteomic assays. Termed "Affinity-Bead Assisted Mass Spectrometry" (Affi-BAMS), this LC-free technology enables development of highly specific and customizable assay panels for simultaneous profiling of multiple proteins and PTMs. While affinity beads have been used previously in combination with MS, the Affi-BAMS workflow uses enrichment on a single bead that contains one type of antibody, generally capturing a single analyte (protein or PTM) while having enough binding capacity to enable quantification within approximately 3 orders of magnitude. The multiplexing capability is achieved by combining Affi-BAMS beads with different protein specificities. To enable screening of bead-captured analytes by MS, we further developed a novel method of performing spatially localized elution of targets from individual beads arrayed on a microscope slide. The resulting arrays of micro spots contain highly concentrated analytes localized within 0.5 mm diameter spots that can be directly measured using MALDI MS. While both intact proteins and protein fragments can be monitored by Affi-BAMS, we initially focused on applying this technology for bottom-up proteomics to enable screening of hundreds of samples per day by combining the robust magnetic bead-based workflow with the high throughput nature of MALDI MS acquisition. To demonstrate the variety of applications and robustness of Affi-BAMS, several studies are presented that focus on the response of 4EBP1, RPS6, ERK1/ERK2, mTOR, Histone H3 and C-MET to stimuli including rapamycin, H2O2, EPO, SU11274, Staurosporine and Vorinostat.


Asunto(s)
Análisis por Micromatrices/métodos , Proteómica/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Cromatina , Humanos , Peróxido de Hidrógeno , Isótopos , Péptido Hidrolasas/química , Mutación Puntual , Procesamiento Proteico-Postraduccional , Proteínas/química , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Transducción de Señal
3.
Blood ; 125(23): 3536-41, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25887776

RESUMEN

As essential mediators of red cell production, erythropoietin (EPO) and its cell surface receptor (EPO receptor [EPOR]) have been intensely studied. Early investigations defined basic mechanisms for hypoxia-inducible factor induction of EPO expression, and within erythroid progenitors EPOR engagement of canonical Janus kinase 2/signal transducer and activator of transcription 5 (JAK2/STAT5), rat sarcoma/mitogen-activated protein kinase/extracellular signal-regulated kinase (RAS/MEK/ERK), and phosphatidylinositol 3-kinase (PI3K) pathways. Contemporary genetic, bioinformatic, and proteomic approaches continue to uncover new clinically relevant modulators of EPO and EPOR expression, and EPO's biological effects. This Spotlight review highlights such factors and their emerging roles during erythropoiesis and anemia.


Asunto(s)
Células Precursoras Eritroides/metabolismo , Eritropoyesis/fisiología , Eritropoyetina/biosíntesis , Regulación de la Expresión Génica/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Receptores de Eritropoyetina/biosíntesis , Anemia/genética , Anemia/metabolismo , Animales , Células Precursoras Eritroides/citología , Eritropoyetina/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Receptores de Eritropoyetina/genética , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
5.
Stem Cells ; 32(9): 2550-6, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24964278

RESUMEN

Prime regulation over hematopoietic progenitor cell (HPC) production is exerted by hematopoietins (HPs) and their Janus kinase-coupled receptors (HP-Rs). For HP/HP-R studies, one central challenge in determining specific effects involves the delineation of nonredundant signal transduction factors and their lineage restricted actions. Via loss-of-function studies, we define roles for an HP-regulated Serpina3g/Spi2A intracellular serpin during granulomyelocytic, B-cell, and hematopoietic stem cell (HSC) formation. In granulomyelocytic progenitors, granulocyte macrophage colony stimulating factor (GMCSF) strongly induced Serpina3g expression with Stat5 dependency. Spi2A-knockout (KO) led to 20-fold decreased CFU-GM formation, limited GMCSF-dependent granulocyte formation, and compromised neutrophil survival upon tumor necrosis factor alpha (TNF-α) exposure. In B-cell progenitors, Serpina3g was an interleukin-7 (IL7) target. Spi2A-KO elevated CFU-preB greater than sixfold and altered B-cell formation in competitive bone marrow transplant (BMT), and CpG challenge experiments. In HSCs, Serpina3g/Spi2A expression was also elevated. Spi2A-KO compromised LT-HSC proliferation (as well as lineage(neg) Sca1(pos) Kit(pos) (LSK) cell lysosomal integrity), and skewed LSK recovery post 5-FU. Spi2A therefore functions to modulate HP-regulated immune cell and HSC formation post-5-FU challenge.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Serpinas/metabolismo , Animales , Ratones
6.
Blood ; 119(23): 5522-31, 2012 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-22508938

RESUMEN

Sprouty proteins are established modifiers of receptor tyrosine kinase (RTK) signaling and play important roles in vasculogenesis, bone morphogenesis, and renal uteric branching. Little is understood, however, concerning possible roles for these molecular adaptors during hematopoiesis. Within erythroid lineage, Spry1 was observed to be selectively and highly expressed at CFU-e to erythroblast stages. In analyses of possible functional roles, an Mx1-Cre approach was applied to conditionally delete Spry1. At steady state, Spry1 deletion selectively perturbed erythroid development and led to reticulocytosis plus heightened splenic erythropoiesis. When challenged by hemolysis, Spry1-null mice exhibited worsened anemia and delayed recovery. During short-term marrow transplantation, Spry1-null donor marrow also failed to efficiently rescue the erythron. In each anemia model, however, hyperexpansion of erythroid progenitors was observed. Spry function depends on phosphorylation of a conserved N-terminal PY motif. Through an LC-MS/MS approach, Spry1 was discovered to be regulated via the erythropoietin receptor (EPOR), with marked EPO-induced Spry1-PY53 phosphorylation observed. When EPOR signaling pathways were analyzed within Spry1-deficient erythroid progenitors, hyperactivation of not only Erk1,2 but also Jak2 was observed. Studies implicate Spry1 as a novel regulator of erythropoiesis during anemia, transducer of EPOR signals, and candidate suppressor of Jak2 activity.


Asunto(s)
Eritropoyesis , Eritropoyetina/metabolismo , Janus Quinasa 2/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Receptores de Eritropoyetina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Anemia/genética , Anemia/metabolismo , Animales , Trasplante de Médula Ósea , Células Cultivadas , Activación Enzimática , Eritroblastos/citología , Eritroblastos/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfoproteínas/genética , Reticulocitos/citología , Reticulocitos/metabolismo
7.
Blood ; 116(24): 5334-46, 2010 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-20810925

RESUMEN

Investigations of bone marrow (BM) erythroblast development are important for clinical concerns but are hindered by progenitor cell and tissue availability. We therefore sought to more specifically define dynamics, and key regulators, of the formation of developing BM erythroid cell cohorts. A unique Kit(-)CD71(high)Ter119(-) "stage E2" proerythroblast pool first is described, which (unlike its Kit(+) "stage E1" progenitors, or maturing Ter119(+) "stage E3" progeny) proved to selectively expand ∼ 7-fold on erythropoietin challenge. During short-term BM transplantation, stage E2 proerythroblasts additionally proved to be a predominantly expanded progenitor pool within spleen. This E1→E2→E3 erythroid series reproducibly formed ex vivo, enabling further characterizations. Expansion, in part, involved E1 cell hyperproliferation together with rapid E2 conversion plus E2 stage restricted BCL2 expression. Possible erythropoietin/erythropoietin receptor proerythroblast stage specific events were further investigated in mice expressing minimal erythropoietin receptor alleles. For a hypomorphic erythropoietin receptor-HM allele, major defects in erythroblast development occurred selectively at stage E2. In addition, stage E2 cells proved to interact productively with primary BM stromal cells in ways that enhanced both survival and late-stage development. Overall, findings reveal a novel transitional proerythroblast compartment that deploys unique expansion devices.


Asunto(s)
Anemia , Proliferación Celular , Eritroblastos/citología , Células Precursoras Eritroides/citología , Eritropoyetina/farmacología , Alelos , Animales , Trasplante de Médula Ósea , Comunicación Celular/fisiología , Eritropoyesis , Ratones , Ratones Endogámicos C57BL , Receptores de Eritropoyetina/genética , Bazo/citología , Células del Estroma
9.
Blood ; 113(20): 4955-62, 2009 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-19264917

RESUMEN

Anemia as associated with numerous clinical conditions can be debilitating, but frequently can be treated via administration of epoetin-alfa, darbepoietin-alfa, or methoxy-PEG epoetin-beta. Despite the complexity of EPO-EPO receptor interactions, the development of interesting EPO mimetic peptides (EMPs) also has been possible. CNTO 530 is one such novel MIMETIBODY Fc-domain dimeric EMP fusion protein. In a mouse model, single-dose CNTO 530 (unlike epoetin-alfa or darbepoietin-alfa) bolstered red cell production for up to 1 month. In 5-fluorouracil and carboplatin-paclitaxel models, CNTO 530 also protected against anemia with unique efficiency. These actions were not fully accounted for by half-life estimates, and CNTO 530 signaling events therefore were studied. Within primary bone marrow erythroblasts, kinetics of STAT5, ERK, and AKT activation were similar for CNTO 530 and epoetin-alfa. p70S6K activation by CNTO 530, however, was selectively sustained. In vivo, CNTO 530 uniquely stimulated the enhanced formation of PODXL(high)CD71(high) (pro)erythroblasts at frequencies multifold above epoetin-alfa or darbepoietin-alfa. CNTO 530 moreover supported the sustained expansion of a bone marrow-resident Kit(neg)CD71(high)Ter119(neg) progenitor pool. Based on these distinct erythropoietic and EPOR signaling properties, CNTO 530 holds excellent promise as a new EPO mimetic.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Eritroblastos/efectos de los fármacos , Eritropoyetina/análogos & derivados , Hematínicos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Anemia/patología , Animales , Células de la Médula Ósea/fisiología , Recuento de Células , Células Cultivadas , Evaluación Preclínica de Medicamentos , Eritroblastos/fisiología , Eritropoyesis/efectos de los fármacos , Eritropoyetina/química , Femenino , Hematínicos/química , Ratones , Ratones Endogámicos C57BL , Imitación Molecular , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/química , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
10.
Curr Opin Hematol ; 17(3): 169-76, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20173635

RESUMEN

PURPOSE OF REVIEW: In 1985-1989, erythropoietin (EPO), its receptor (EPOR), and janus kinase 2 were cloned; established to be essential for definitive erythropoiesis; and initially intensely studied. Recently, new impetus, tools, and model systems have emerged to re-examine EPO/EPOR actions, and are addressed in this review. Impetus includes indications that EPO affects significantly more than standard erythroblast survival pathways, the development of novel erythropoiesis-stimulating agents, increasing evidence for EPO/EPOR cytoprotection of ischemically injured tissues, and potential EPO-mediated worsening of tumorigenesis. RECENT FINDINGS: New findings are reviewed in four functional contexts: (pro)erythroblast survival mechanisms, new candidate EPO/EPOR effects on erythroid cell development and new EPOR responses, EPOR downmodulation and trafficking, and novel erythropoiesis-stimulating agents. SUMMARY: As Current Opinion, this monograph seeks to summarize, and provoke, new EPO/EPOR action concepts. Specific problems addressed include: beyond (and before) BCL-XL, what key survival factors are deployed in early-stage proerythroblasts? Are distinct EPO/EPOR signals transduced in stage-selective fashions? Is erythroblast proliferation also modulated by EPO/EPOR signals? What functions are subserved by new noncanonical EPO/EPOR response factors (e.g. podocalyxin like-1, tribbles 3, reactive oxygen species, and nuclear factor kappa B)? What key regulators mediate EPOR inhibition and trafficking? And for emerging erythropoiesis-stimulating agents, to what extent do activities parallel EPOs (or differ in advantageous, potentially complicating ways, or both)?


Asunto(s)
Eritroblastos/fisiología , Eritropoyetina/metabolismo , Receptores de Eritropoyetina/metabolismo , Transducción de Señal , Animales , Humanos
11.
J Cell Biochem ; 109(2): 363-74, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19950207

RESUMEN

The signals that direct pluripotent stem cell differentiation into lineage-specific cells remain largely unknown. Here, we investigated the roles of BMP on vascular progenitor development from human embryonic stem cells (hESCs). In a serum-free condition, hESCs sequentially differentiated into CD34+CD31-, CD34+CD31+, and then CD34-CD31+ cells during vascular cell development. CD34+CD31+ cells contained vascular progenitor population that gives rise to endothelial cells and smooth muscle cells. BMP4 promoted hESC differentiation into CD34+CD31+ cells at an early stage. In contrast, TGFbeta suppressed BMP4-induced CD34+CD31+ cell development, and promoted CD34+CD31- cells that failed to give rise to either endothelial or smooth muscle cells. The BMP-Smad inhibitor, dorsomorphin, inhibited phosphorylation of Smad1/5/8, and blocked hESC differentiation to CD34+CD31+ progenitor cells, suggesting that BMP Smad-dependent signaling is critical for CD34+CD31+ vascular progenitor development. Our findings provide new insight into how pluripotent hESCs differentiate into vascular cells.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Células Madre Embrionarias/citología , Neovascularización Fisiológica , Proteínas Smad/metabolismo , Antígenos CD34 , Proteína Morfogenética Ósea 4/antagonistas & inhibidores , Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular/fisiología , Línea Celular , Medio de Cultivo Libre de Suero , Células Madre Embrionarias/metabolismo , Células Endoteliales/citología , Humanos , Músculo Liso Vascular/citología , Fosforilación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta , Pirazoles/farmacología , Pirimidinas/farmacología , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología
12.
Blood ; 112(3): 886-90, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18535204

RESUMEN

The adult erythron is maintained via dynamic modulation of erythroblast survival potentials. Toward identifying novel regulators of this process, murine splenic erythroblasts at 3 developmental stages were prepared, purified and profiled. Stage-to-stage modulated genes were then functionally categorized, with a focus on apoptotic factors. In parallel with BCL-X and NIX, death-associated protein kinase-2 (DAPK2) was substantially up-modulated during late erythropoiesis. Among hematopoietic lineages, DAPK2 was expressed predominantly in erythroid cells. In a Gata1-IE3.9int-DAPK2 transgenic mouse model, effects on steady-state reticulocyte and red blood cell (RBC) levels were limited. During hemolytic anemia, however, erythropoiesis was markedly deficient. Ex vivo ana-lyses revealed heightened apoptosis due to DAPK2 at a Kit(-)CD71(high)Ter119(-) stage, together with a subsequent multifold defect in late-stage Kit(-)CD71(high)Ter119(+) cell formation. In UT7epo cells, siRNA knock-down of DAPK2 enhanced survival due to cytokine withdrawal, and DAPK2's phosphorylation and kinase activity also were erythropoietin (EPO)-modulated. DAPK2 therefore comprises a new candidate attenuator of stress erythropoiesis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Eritroblastos/citología , Eritropoyesis/genética , Eritropoyetina/farmacología , Anemia Hemolítica , Animales , Proteínas Reguladoras de la Apoptosis/análisis , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Quinasas Dependientes de Calcio-Calmodulina/análisis , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Linaje de la Célula , Proteínas Quinasas Asociadas a Muerte Celular , Hemostasis , Ratones , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Bazo/citología , Regulación hacia Arriba/genética
13.
Blood ; 111(11): 5390-9, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18349318

RESUMEN

EPO functions primarily as an erythroblast survival factor, and its antiapoptotic actions have been proposed to involve predominantly PI3-kinase and BCL-X pathways. Presently, the nature of EPO-regulated survival genes has been investigated through transcriptome analyses of highly responsive, primary bone marrow erythroblasts. Two proapoptotic factors, Bim and FoxO3a, were rapidly repressed not only via the wild-type EPOR, but also by PY-deficient knocked-in EPOR alleles. In parallel, Pim1 and Pim3 kinases and Irs2 were induced. For this survival gene set, induction failed via a PY-null EPOR-HM allele, but was restored upon reconstitution of a PY343 STAT5-binding site within a related EPOR-H allele. Notably, EPOR-HM supports erythropoiesis at steady state but not during anemia, while EPOR-H exhibits near wild-type EPOR activities. EPOR-H and the wild-type EPOR (but not EPOR-HM) also markedly stimulated the expression of Trb3 pseudokinase, and intracellular serpin, Serpina-3G. For SERPINA-3G and TRB3, ectopic expression in EPO-dependent progenitors furthermore significantly inhibited apoptosis due to cytokine withdrawal. BCL-XL and BCL2 also were studied, but in highly responsive Kit(pos)CD71(high)Ter119(neg) erythroblasts, neither was EPO modulated. EPOR survival circuits therefore include the repression of Bim plus FoxO3a, and EPOR/PY343/STAT5-dependent stimulation of Pim1, Pim3, Irs2 plus Serpina-3G, and Trb3 as new antiapoptotic effectors.


Asunto(s)
Apoptosis/fisiología , Eritroblastos/fisiología , Regulación de la Expresión Génica/fisiología , Receptores de Eritropoyetina/fisiología , Animales , Supervivencia Celular , Citometría de Flujo , Expresión Génica , Perfilación de la Expresión Génica , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Exp Hematol ; 86: 28-42.e3, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32473295

RESUMEN

As important vectors for ectopic protein expression, gene silencing, and progenitor cell barcoding, lentiviruses continue to emerge as versatile research and clinical tools. For studies employing cell types that are relatively resistant to transduction, high-titer lentivirus preparations with low cytotoxicity are required. During lentivirus production, carryover plasmid DNA endotoxins, transfection reagents, damaged packaging cells, and virus concentration procedures are potential sources of cytotoxicity. As an often unevaluated property of lentivirus preparations, cytotoxicity can unwittingly skew estimates of functional titers and complicate interpretations of transduced cell phenotypes. By employing hematopoietic UT7epo cells cultured in erythropoietin (EPO) below maximal dosing, we first define a sensitive flow cytometric bioassay for critically assessing the cytotoxicity (and titers) of lentivirus preparations. Bioassay of custom preparations of research-grade lentiviruses from six commercial sources unexpectedly revealed substantial cytotoxicity (with certain preparations additionally registering titers several log below designated values). To overcome such limiting properties, we further report on unique, efficient workflows for reproducibly preparing and processing high-titer, low-cytotoxicity (HTLC) lentiviruses at research scale. These HTLC lentiviruses reliably transduce peripheral blood hematopoietic stem/progenitor cells (PB-HSPCs) at frequencies ≥40%, with low cytotoxicity. In addition, by employing cyclosporin H (to inhibit IFITM3), PB-HSPCs can be transduced at heightened efficiency with nominal cytotoxicity. Overall, this work provides straightforward approaches to (1) critical assessment of the cytotoxicity of lentivirus preparations; (2) reproducible generation (and concentration) of high-quality lentiviruses via a streamlined workflow; and (3) transduction of PB-HSPCs at benchmark levels with nominal cytotoxicity.


Asunto(s)
Eritropoyetina , Vectores Genéticos , Movilización de Célula Madre Hematopoyética , Lentivirus , Células Madre de Sangre Periférica/metabolismo , Transducción Genética , Línea Celular , Eritropoyetina/biosíntesis , Eritropoyetina/genética , Humanos , Células Madre de Sangre Periférica/citología
15.
Exp Hematol ; 84: 29-44, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32259549

RESUMEN

Erythroid cell formation critically depends on signals transduced via erythropoietin (EPO)/EPO receptor (EPOR)/JAK2 complexes. This includes not only core response modules (e.g., JAK2/STAT5, RAS/MEK/ERK), but also specialized effectors (e.g., erythroferrone, ASCT2 glutamine transport, Spi2A). By using phospho-proteomics and a human erythroblastic cell model, we identify 121 new EPO target proteins, together with their EPO-modulated domains and phosphosites. Gene ontology (GO) enrichment for "Molecular Function" identified adaptor proteins as one top EPO target category. This includes a novel EPOR/JAK2-coupled network of actin assemblage modifiers, with adaptors DLG-1, DLG-3, WAS, WASL, and CD2AP as prime components. "Cellular Component" GO analysis further identified 19 new EPO-modulated cytoskeletal targets including the erythroid cytoskeletal targets spectrin A, spectrin B, adducin 2, and glycophorin C. In each, EPO-induced phosphorylation occurred at pY sites and subdomains, which suggests coordinated regulation by EPO of the erythroid cytoskeleton. GO analysis of "Biological Processes" further revealed metabolic regulators as a likewise unexpected EPO target set. Targets included aldolase A, pyruvate dehydrogenase α1, and thioredoxin-interacting protein (TXNIP), with EPO-modulated p-Y sites in each occurring within functional subdomains. In TXNIP, EPO-induced phosphorylation occurred at novel p-T349 and p-S358 sites, and was paralleled by rapid increases in TXNIP levels. In UT7epo-E and primary human stem cell (HSC)-derived erythroid progenitor cells, lentivirus-mediated short hairpin RNA knockdown studies revealed novel pro-erythropoietic roles for TXNIP. Specifically, TXNIP's knockdown sharply inhibited c-KIT expression; compromised EPO dose-dependent erythroblast proliferation and survival; and delayed late-stage erythroblast formation. Overall, new insight is provided into EPO's diverse action mechanisms and TXNIP's contributions to EPO-dependent human erythropoiesis.


Asunto(s)
Eritropoyesis , Eritropoyetina/metabolismo , Fosfoproteínas/metabolismo , Proteómica , Eritropoyetina/genética , Humanos , Fosfoproteínas/genética
16.
Cell Signal ; 69: 109554, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32027948

RESUMEN

The formation of erythroid progenitor cells depends sharply upon erythropoietin (EPO), its cell surface receptor (erythropoietin receptor, EPOR), and Janus kinase 2 (JAK2). Clinically, recombinant human EPO (rhEPO) additionally is an important anti-anemia agent for chronic kidney disease (CKD), myelodysplastic syndrome (MDS) and chemotherapy, but induces hypertension, and can exert certain pro-tumorigenic effects. Cellular signals transduced by EPOR/JAK2 complexes, and the nature of EPO-modulated signal transduction factors, therefore are of significant interest. By employing phospho-tyrosine post-translational modification (p-Y PTM) proteomics and human EPO- dependent UT7epo cells, we have identified 22 novel kinases and phosphatases as novel EPO targets, together with their specific sites of p-Y modification. New kinases modified due to EPO include membrane palmitoylated protein 1 (MPP1) and guanylate kinase 1 (GUK1) guanylate kinases, together with the cytoskeleton remodeling kinases, pseudopodium enriched atypical kinase 1 (PEAK1) and AP2 associated kinase 1 (AAK1). Novel EPO- modified phosphatases include protein tyrosine phosphatase receptor type A (PTPRA), phosphohistidine phosphatase 1 (PHPT1), tensin 2 (TENC1), ubiquitin associated and SH3 domain containing B (UBASH3B) and protein tyrosine phosphatase non-receptor type 18 (PTPN18). Based on PTPN18's high expression in hematopoietic progenitors, its novel connection to JAK kinase signaling, and a unique EPO- regulated PTPN18-pY389 motif which is modulated by JAK2 inhibitors, PTPN18's actions in UT7epo cells were investigated. Upon ectopic expression, wt-PTPN18 promoted EPO dose-dependent cell proliferation, and survival. Mechanistically, PTPN18 sustained the EPO- induced activation of not only mitogen-activated protein kinases 1 and 3 (ERK1/2), AKT serine/threonine kinase 1-3 (AKT), and signal transducer and activator of transcription 5A and 5B (STAT5), but also JAK2. Each effect further proved to depend upon PTPN18's EPO- modulated (p)Y389 site. In analyses of the EPOR and the associated adaptor protein RHEX (regulator of hemoglobinization and erythroid cell expansion), wt-PTPN18 increased high molecular weight EPOR forms, while sharply inhibiting the EPO-induced phosphorylation of RHEX-pY141. Each effect likewise depended upon PTPN18-Y389. PTPN18 thus promotes signals for EPO-dependent hematopoietic cell growth, and may represent a new druggable target for myeloproliferative neoplasms.


Asunto(s)
Eritropoyesis , Eritropoyetina/metabolismo , Janus Quinasa 2/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/fisiología , Receptores de Eritropoyetina/metabolismo , Línea Celular , Humanos , Proteómica , Transducción de Señal
17.
J Clin Invest ; 116(3): 683-94, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16511603

RESUMEN

Anemia due to chronic disease or chemotherapy often is ameliorated by erythropoietin (Epo). Present studies reveal that, unlike steady-state erythropoiesis, erythropoiesis during anemia depends sharply on an Epo receptor-phosphotyrosine-343-Stat5 signaling axis. In mice expressing a phosphotyrosine-null (PY-null) Epo receptor allele (EpoR-HM), severe and persistent anemia was induced by hemolysis or 5-fluorouracil. In short-term transplantation experiments, donor EpoR-HM bone marrow cells also failed to efficiently repopulate the erythroid compartment. In each context, stress erythropoiesis was rescued to WT levels upon the selective restoration of an EpoR PY343 Stat5-binding site (EpoR-H allele). As studied using a unique primary culture system, EpoR-HM erythroblasts exhibited marked stage-specific losses in Epo-dependent growth and survival. EpoR-H PY343 signals restored efficient erythroblast expansion, and the selective Epo induction of the Stat5 target genes proviral integration site-1 (Pim-1) and oncostatin-M. Bcl2-like 1 (Bcl-x), in contrast, was not significantly induced via WT-EpoR, EpoR-HM, or EpoR-H alleles. In Kit+ CD71+ erythroblasts, EpoR-PY343 signals furthermore enhanced SCF growth effects, and SCF modulation of Pim-1 kinase and oncostatin-M expression. In maturing Kit- CD71+ erythroblasts, oncostatin-M exerted antiapoptotic effects that likewise depended on EpoR PY343-mediated events. Stress erythropoiesis, therefore, requires stage-specific EpoR-PY343-Stat5 signals, some of which selectively bolster SCF and oncostatin-M action.


Asunto(s)
Diferenciación Celular/fisiología , Eritropoyesis/fisiología , Fosfotirosina/metabolismo , Receptores de Eritropoyetina/fisiología , Factor de Transcripción STAT5/fisiología , Transducción de Señal/fisiología , Anemia/sangre , Anemia/inducido químicamente , Anemia/genética , Animales , Sitios de Unión/genética , Células de la Médula Ósea/citología , Diferenciación Celular/genética , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas , Eritroblastos/citología , Eritropoyesis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Fenilhidrazinas/administración & dosificación , Fosfotirosina/genética , Fosfotirosina/fisiología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores de Eritropoyetina/biosíntesis , Receptores de Eritropoyetina/genética , Transducción de Señal/genética
18.
Cell Signal ; 20(8): 1513-20, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18538998

RESUMEN

KIT and erythropoietin receptor (EpoR) mediated co-signaling is essential for normal erythroid cell expansion, however the intracellular signals that contribute to cooperative signaling are poorly understood. Here, we examined the role of intracellular tyrosine residues in KIT and EpoR cooperation by co-expressing tyrosine (Y) to phenylalanine (F) and deletion mutants of KIT and EpoR in 32D cells. Of the four EpoR mutants examined, only EpoR-Y343 induced proliferation to near wildtype EpoR levels. A modest increase in the growth was also observed in 32D cells expressing the EpoR-Y343F; however neither EpoR-W282R nor EpoR-F8 showed any increase in growth over baseline. Biochemical analysis revealed that EpoR-Y343 induced the activation of Stat5, PI-3Kinase/Akt and MAP kinase Erk1/2 to near wildtype EpoR levels, while the remaining mutants failed to activate any of these signals. Interestingly, none of the EpoR mutants cooperated with WT KIT, although EpoR-Y343 showed a modest increase in co-signaling. Loss of seven tyrosine residues in KIT (KIT-F7) completely abrogated EpoR induced co-signaling. Restoring the Src kinase binding sites in KIT-F7 alone or together with the PI3Kinase binding site restored KIT induced signals as well as co-signals with WT EpoR, although restoring the Src kinase binding sites along with the PLC-gamma binding site repressed both KIT induced signaling as well as co-signaling with WT EpoR. Taken together, these results suggest that KIT and EpoR mediated co-signaling requires intracellular tyrosine residues and tyrosine residues that bind Src kinases in the KIT receptor appear to be sufficient for restoring both KIT signaling as well as co-signaling with EpoR. In contrast, restoration of the PLC-gamma binding site in the context of Src binding sites appears to antagonize the positive signals induced via the Src kinase binding sites in the KIT receptor.


Asunto(s)
Proteínas Proto-Oncogénicas c-kit/química , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores de Eritropoyetina/química , Receptores de Eritropoyetina/metabolismo , Transducción de Señal , Tirosina/metabolismo , Sustitución de Aminoácidos , Animales , Línea Celular , Proliferación Celular , Ratones , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Receptores de Eritropoyetina/genética , Tirosina/genética
19.
BMC Dev Biol ; 8: 86, 2008 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-18801194

RESUMEN

BACKGROUND: Bone morphogenetic proteins (BMPs) regulate essential processes during organogenesis, and a functional understanding of these secreted proteins depends on identification of their target cells. In this study, we generate a transgenic reporter for organogenesis studies that we use to define BMP pathway activation in the developing kidney. RESULTS: Mouse strains reporting on BMP pathway activation were generated by transgenically expressing beta-galactosidase under the control of BMP responsive elements from Id1. Reporter expression corresponds well with immunoassays for pathway activation in all organs studied, validating the model. Using these reporters we have generated a detailed map of cellular targets of BMP signaling in the developing kidney. We find that SMAD dependent BMP signaling is active in collecting duct trunks, but not tips. Furthermore, glomerular endothelial cells, and proximal nephron tubules from the renal vesicle stage onward show pathway activation. Surprisingly, little activation is detected in the nephrogenic zone of the kidney, and in organ culture BMP treatment fails to activate SMAD dependent BMP signaling in nephron progenitor cells. In contrast, signaling is efficiently induced in collecting duct tips. CONCLUSION: Transgenic reporters driven by control elements from BMP responsive genes such as Id1 offer significant advantages in sensitivity and consistency over immunostaining for studies of BMP pathway activation. They also provide opportunities for analysis of BMP signaling in organ and primary cell cultures subjected to experimental manipulation. Using such a reporter, we made the surprising finding that SMAD dependent BMP signaling is inactive in nephron progenitors, and that these cells are refractory to activation by applied growth factors. Furthermore, we find that the BMP pathway is not normally active in collecting duct tips, but that it can be ectopically activated by BMP treatment, offering a possible explanation for the inhibitory effects of BMP treatment on collecting duct growth and branching.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Marcación de Gen , Genes Reporteros/fisiología , Riñón/embriología , Organogénesis/fisiología , Transducción de Señal/fisiología , Animales , Femenino , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Proteína Smad1/biosíntesis , Proteína Smad1/genética , Proteína Smad5/biosíntesis , Proteína Smad5/genética , Proteína Smad8/biosíntesis , Proteína Smad8/genética , beta-Galactosidasa/biosíntesis , beta-Galactosidasa/genética
20.
Blood ; 116(20): 4040-2, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21088142
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA