Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Immunity ; 53(1): 217-232.e5, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32668225

RESUMEN

B cells are capable of a wide range of effector functions including antibody secretion, antigen presentation, cytokine production, and generation of immunological memory. A consistent strategy for classifying human B cells by using surface molecules is essential to harness this functional diversity for clinical translation. We developed a highly multiplexed screen to quantify the co-expression of 351 surface molecules on millions of human B cells. We identified differentially expressed molecules and aligned their variance with isotype usage, VDJ sequence, metabolic profile, biosynthesis activity, and signaling response. Based on these analyses, we propose a classification scheme to segregate B cells from four lymphoid tissues into twelve unique subsets, including a CD45RB+CD27- early memory population, a class-switched CD39+ tonsil-resident population, and a CD19hiCD11c+ memory population that potently responds to immune activation. This classification framework and underlying datasets provide a resource for further investigations of human B cell identity and function.


Asunto(s)
Subgrupos de Linfocitos B/clasificación , Subgrupos de Linfocitos B/inmunología , Isotipos de Inmunoglobulinas/metabolismo , Proteínas de la Membrana/metabolismo , 5'-Nucleotidasa/metabolismo , Apirasa/metabolismo , Antígeno CD11c/metabolismo , Femenino , Proteínas Ligadas a GPI/metabolismo , Humanos , Memoria Inmunológica/inmunología , Antígenos Comunes de Leucocito/metabolismo , Persona de Mediana Edad , Transducción de Señal/inmunología , Receptor fas/metabolismo
2.
Blood ; 133(7): 688-696, 2019 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-30593445

RESUMEN

Transforming growth factor ß (TGF-ß) is well known for its important function in hematopoietic stem cell (HSC) quiescence. However, the molecular mechanism underlining this function remains obscure. Endoglin (Eng), a type III receptor for the TGF-ß superfamily, has been shown to selectively mark long-term HSCs; however, its necessity in adult HSCs is unknown due to embryonic lethality. Using conditional deletion of Eng combined with serial transplantation, we show that this TGF-ß receptor is critical to maintain the HSC pool. Transplantation of Eng-deleted whole bone marrow or purified HSCs into lethally irradiated mice results in a profound engraftment defect in tertiary and quaternary recipients. Cell cycle analysis of primary grafts revealed decreased frequency of HSCs in G0, suggesting that lack of Eng impairs reentry of HSCs to quiescence. Using cytometry by time of flight (CyTOF) to evaluate the activity of signaling pathways in individual HSCs, we find that Eng is required within the Lin-Sca+Kit+-CD48- CD150+ fraction for canonical and noncanonical TGF-ß signaling, as indicated by decreased phosphorylation of SMAD2/3 and the p38 MAPK-activated protein kinase 2, respectively. These findings support an essential role for Eng in positively modulating TGF-ß signaling to ensure maintenance of HSC quiescence.


Asunto(s)
Endoglina/metabolismo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Ciclo Celular , Endoglina/genética , Ratones , Ratones Endogámicos C57BL , Fosforilación , Transducción de Señal
3.
Stem Cells ; 32(8): 2072-83, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24677751

RESUMEN

Embryonic stem cells (ESCs) represent an ideal model to study how lineage decisions are established during embryonic development. Using a doxycycline-inducible mouse ESC line, we have previously shown that expression of the transcriptional activator Pax3 in early mesodermal cells leads to the robust generation of paraxial mesoderm progenitors that ultimately differentiate into skeletal muscle precursors. Here, we show that the ability of this transcription factor to induce the skeletal myogenic cell fate occurs at the expenses of the cardiac lineage. Our results show that the PDGFRα+FLK1--subfraction represents the main population affected by Pax3, through downregulation of several transcripts encoding for proteins involved in cardiac development. We demonstrate that although Nkx2-5, Tbx5, and Gata4 negatively affect Pax3 skeletal myogenic activity, the cardiac potential of embryoid body-derived cultures is restored solely by forced expression of Tbx5. Taking advantage of this model, we used an unbiased genome-wide approach to identify genes whose expression is rescued by Tbx5, and which could represent important regulators of cardiac development. These findings elucidate mechanisms regulating the commitment of mesodermal cells in the early embryo and identify the Tbx5 cardiac transcriptome.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/metabolismo , Músculo Esquelético/citología , Miocardio/citología , Factores de Transcripción Paired Box/metabolismo , Proteínas de Dominio T Box/metabolismo , Animales , Western Blotting , Linaje de la Célula , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Músculo Esquelético/embriología , Factor de Transcripción PAX3 , Técnicas de Placa-Clamp , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis
4.
Blood ; 119(23): 5417-28, 2012 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-22535663

RESUMEN

Much remains unknown about the signals that induce early mesoderm to initiate hematopoietic differentiation. Here, we show that endoglin (Eng), a receptor for the TGFß superfamily, identifies all cells with hematopoietic fate in the early embryo. These arise in an Eng(+)Flk1(+) mesodermal precursor population at embryonic day 7.5 (E7.5), a cell fraction also endowed with endothelial potential. In Eng-knockout embryos, hematopoietic colony activity and numbers of CD71(+)Ter119(+) erythroid progenitors were severely reduced. This coincided with severely reduced expression of embryonic globin and key bone morphogenic protein (BMP) target genes, including the hematopoietic regulators Scl, Gata1, Gata2, and Msx-1. To interrogate molecular pathways active in the earliest hematopoietic progenitors, we applied transcriptional profiling to sorted cells from E7.5 embryos. Eng(+)Flk-1(+) progenitors coexpressed TGFß and BMP receptors and target genes. Furthermore, Eng(+)Flk-1(+) cells presented high levels of phospho-SMAD1/5, indicating active TGFß and/or BMP signaling. Remarkably, under hematopoietic serum-free culture conditions, hematopoietic outgrowth of Eng-expressing cells was dependent on the TGFß superfamily ligands BMP4, BMP2, or TGF-ß1. These data demonstrate that the E(+)F(+) fraction at E7.5 represents mesodermal cells competent to respond to TGFß1, BMP4, or BMP2, shaping their hematopoietic development, and that Eng acts as a critical regulator in this process by modulating TGF/BMP signaling.


Asunto(s)
Embrión de Mamíferos/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/ultraestructura , Desarrollo Embrionario , Endoglina , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Células Madre Hematopoyéticas/citología , Péptidos y Proteínas de Señalización Intracelular/análisis , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Stem Cells ; 31(9): 1893-901, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23712751

RESUMEN

Endoglin (Eng), an ancillary receptor of the transforming growth factor beta (TGFß) signaling pathway superfamily, has been well recognized for its important function in vascular development and angiogenesis since its discovery more than a decade ago. Recent studies show that this receptor is also critical for the emergence of blood during embryonic development, and that at E7.5, endoglin together with Flk-1 identifies early mesoderm progenitors that are endowed with hematopoietic and endothelial potential. These two lineages emerge in very close association during embryogenesis, and because they share the expression of the same surface markers, it has been difficult to distinguish the earliest hematopoietic from endothelial cells. Here, we evaluated the function of endoglin in hematopoiesis as development progresses past E7.5, and found that the hematopoietic and endothelial progenitors can be distinguished by the levels of endoglin in E9.5 yolk sacs. Whereas endothelial cells are Eng(bright), hematopoietic activity is primarily restricted to a subset of cells that display dim expression of endoglin (Eng(dim)). Molecular characterization of these subfractions showed that endoglin-mediated induction of hematopoiesis occurs in concert with BMP2/BMP4 signaling. This pathway is highly active in Eng(dim) cells but significantly downregulated in the Eng knockout. Taken together, our findings show an important function for endoglin in mediating BMP2/BMP4 signaling during yolk sac hematopoietic development and suggest that the levels of this receptor modulate TGFß versus bone morphogenetic protein (BMP) signaling.


Asunto(s)
Células Endoteliales/citología , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Péptidos y Proteínas de Señalización Intracelular/genética , Saco Vitelino/citología , Saco Vitelino/embriología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Endoglina , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Modelos Biológicos , Transducción de Señal , Fracciones Subcelulares/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Saco Vitelino/metabolismo
6.
Stem Cells ; 30(8): 1611-23, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22628281

RESUMEN

During embryogenesis, the endothelial and the hematopoietic lineages first appear during gastrulation in the blood island of the yolk sac. We have previously reported that an Ets variant gene 2 (Etv2/ER71) mutant embryo lacks hematopoietic and endothelial lineages; however, the precise roles of Etv2 in yolk sac development remains unclear. In this study, we define the role of Etv2 in yolk sac blood island development using the Etv2 mutant and a novel Etv2-EYFP reporter transgenic line. Both the hematopoietic and the endothelial lineages are absent in the Etv2 mutant yolk sac. In the Etv2-EYFP transgenic mouse, the EYFP reporter is activated in the nascent mesoderm, expressed in the endothelial and blood progenitors, and in the Tie2(+), c-kit(+), and CD41(+) hematopoietic population. The hematopoietic activity in the E7.75 yolk sac was exclusively localized to the Etv2-EYFP(+) population. In the Etv2 mutant yolk sac, Tie2(+) cells are present but do not express hematopoietic or endothelial markers. In addition, these cells do not form hematopoietic colonies, indicating an essential role of Etv2 in the specification of the hematopoietic lineage. Forced overexpression of Etv2 during embryoid body differentiation induces the hematopoietic and the endothelial lineages, and transcriptional profiling in this context identifies Lmo2 as a downstream target. Using electrophoretic mobility shift assay, chromatin immunoprecipitation, transcriptional assays, and mutagenesis, we demonstrate that Etv2 binds to the Lmo2 enhancer and transactivates its expression. Collectively, our studies demonstrate that Etv2 is expressed during and required for yolk sac hematoendothelial development, and that Lmo2 is one of the downstream targets of Etv2.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Endoteliales/citología , Células Madre Hematopoyéticas/metabolismo , Proteínas con Dominio LIM/metabolismo , Factores de Transcripción/metabolismo , Saco Vitelino/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Humanos , Inmunohistoquímica , Proteínas con Dominio LIM/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Factores de Transcripción/genética , Transfección , Saco Vitelino/citología
7.
bioRxiv ; 2023 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-37693547

RESUMEN

Hematopoietic stem and progenitor cell (HSPC) transplantation is an essential therapy for hematological conditions, but finer definitions of human HSPC subsets with associated function could enable better tuning of grafts and more routine, lower-risk application. To deeply phenotype HSPCs, following a screen of 328 antigens, we quantified 41 surface proteins and functional regulators on millions of CD34+ and CD34- cells, spanning four primary human hematopoietic tissues: bone marrow, mobilized peripheral blood, cord blood, and fetal liver. We propose more granular definitions of HSPC subsets and provide new, detailed differentiation trajectories of erythroid and myeloid lineages. These aspects of our revised human hematopoietic model were validated with corresponding epigenetic analysis and in vitro clonal differentiation assays. Overall, we demonstrate the utility of using molecular regulators as surrogates for cellular identity and functional potential, providing a framework for description, prospective isolation, and cross-tissue comparison of HSPCs in humans.

8.
Circ Res ; 107(12): 1460-9, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-20947832

RESUMEN

RATIONALE: several studies demonstrate that hematopoietic tissues are a source of endothelial progenitor cells, which contribute to newly formed blood vessels during tissue repair in adults. However, it is not clear which cell type in these hematopoietic tissues gives rise to endothelial progenitor cells. OBJECTIVE: to identity the origin of endothelial progenitors within the hematopoietic hierarchy and to assess their in vivo revascularization potential. METHODS AND RESULTS: using a single-cell sorting approach and in vitro multilineage differentiation assays, here we show that individual CD34(+)CD45(+)CD133(+)CD38(+) cells from cord blood uniquely have the ability to differentiate into T- and B-lymphoid, myeloid, and endothelial cells. The latter were characterized by the expression of VE-cadherin, KDR, von Willebrand factor, endothelial nitric oxide synthase, the lack of CD45, CD133, and c-fms (colony stimulating factor-1 receptor). Unexpectedly when transplanted into hindlimb ischemic NOD-scid IL2Rγ(null) mice, freshly isolated CD34(+)CD45(+)CD133(+)CD38(+) cells maintained their hematopoietic identity and were rarely found to integrate into host blood vessels. Nevertheless, they significantly improved perfusion, most likely through a paracrine mechanism. On the other hand, CD34(+)CD45(+)CD133(+)CD38(+) cells differentiated in vitro into endothelial cells were able to form vessels in vivo in both Matrigel plug and hindlimb ischemia transplantation assays. CONCLUSIONS: these findings indicate that the CD34(+)CD45(+)CD133(+)CD38(+) cell fraction contains a common progenitor for the hematopoietic and vascular lineages and may represent a valuable cell source for therapeutic applications.


Asunto(s)
Células Endoteliales , Sangre Fetal/citología , Células Madre/citología , Antígeno AC133 , ADP-Ribosil Ciclasa 1 , Animales , Antígenos CD , Antígenos CD34 , Diferenciación Celular , Linaje de la Célula , Células Clonales/citología , Células Endoteliales/citología , Glicoproteínas , Humanos , Antígenos Comunes de Leucocito , Células Progenitoras Linfoides/citología , Glicoproteínas de Membrana , Ratones , Ratones SCID , Células Progenitoras Mieloides/citología , Péptidos , Trasplante de Células Madre
9.
STAR Protoc ; 3(2): 101280, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35434655

RESUMEN

Granulocytes encompass diverse roles, from fighting off pathogens to regulating inflammatory processes in allergies. These roles are represented by distinct cellular phenotypes that we captured with mass cytometry (CyTOF). Our protocol enables simultaneous evaluation of human basophils, eosinophils, and neutrophils under homeostasis and upon immune activation by anti-Immunoglobulin E (anti-IgE) or interleukin-3 (IL-3). Granulocyte integrity and detection of protein markers were optimized so that rare granulocyte populations could be deeply characterized by single cell mass cytometry. For complete details on the use and execution of this protocol, please refer to Vivanco Gonzalez et al. (2020).


Asunto(s)
Eosinófilos , Neutrófilos , Basófilos , Eosinófilos/metabolismo , Citometría de Flujo/métodos , Humanos , Recuento de Leucocitos
10.
Cell Rep Methods ; 2(3)2022 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-35463156

RESUMEN

Master transcription factors (TFs) directly regulate present and future cell states by binding DNA regulatory elements and driving gene-expression programs. Their abundance influences epigenetic priming to different cell fates at the chromatin level, especially in the context of differentiation. In order to link TF protein abundance to changes in TF motif accessibility and open chromatin, we developed InTAC-seq, a method for simultaneous quantification of genome-wide chromatin accessibility and intracellular protein abundance in fixed cells. Our method produces high-quality data and is a cost-effective alternative to single-cell techniques. We showcase our method by purifying bone marrow (BM) progenitor cells based on GATA-1 protein levels and establish high GATA-1-expressing BM cells as both epigenetically and functionally similar to erythroid-committed progenitors.


Asunto(s)
Cromatina , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Cromatina/genética , Linaje de la Célula/genética , Regulación de la Expresión Génica , ADN/metabolismo
11.
Biochim Biophys Acta ; 1793(5): 806-16, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19268501

RESUMEN

Glioblastoma multiforme is highly aggressive and is the most common glial tumor type. Although there have been advances in treatment, the average survival expectancy is 12-15 months. Several genes have been shown to influence glioblastoma progression. In the present work, we demonstrate that the RhoGTPase Activating Protein 21 (ARHGAP21) is expressed in the nuclear and perinuclear regions of several cell lines. In T98G and U138MG, glioblastoma derived cell lines, ARHGAP21 interacts with the C-terminal region of Focal Adhesion Kinase (FAK). ARHGAP21 depletion by shRNAi in T98G cells alters cellular morphology and increases: FAK phosphorylation states and activation of downstream signaling; the activity state of Cdc42; the production of metalloproteinase 2 (MMP-2) and cell migration rates. These modifications were found to be mainly due to the loss of ARHGAP21 action on FAK and, consequently, the activation of downstream effectors. These results suggest not only that ARHGAP21 might act as a tumor suppressor gene, but also indicate that ARHGAP21 might be a master regulator of migration having a crucial role in controlling the progression of different tumor types.


Asunto(s)
Movimiento Celular/fisiología , Quinasa 1 de Adhesión Focal/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Glioblastoma/metabolismo , Transducción de Señal/fisiología , Actinas/metabolismo , Animales , Línea Celular Tumoral , Forma de la Célula , Proteína Sustrato Asociada a CrK/metabolismo , Citoesqueleto/metabolismo , Quinasa 1 de Adhesión Focal/genética , Proteínas Activadoras de GTPasa/genética , Glioblastoma/patología , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Interferencia de ARN , Proteína de Unión al GTP cdc42/metabolismo , Familia-src Quinasas/metabolismo
12.
iScience ; 23(11): 101724, 2020 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-33205028

RESUMEN

Basophils, the rarest granulocyte, play critical roles in parasite- and allergen-induced inflammation. We applied mass cytometry (CyTOF) to simultaneously asses 44 proteins to phenotype and functionally characterize neutrophils, eosinophils, and basophils from 19 healthy donors. There was minimal heterogeneity seen in eosinophils and neutrophils, but data-driven analyses revealed four unique subpopulations within phenotypically basophilic granulocytes (PBG; CD45+HLA-DR-CD123+). Through CyTOF and fluorescence-activated cell sorting (FACS), we classified these four PBG subpopulations as (I) CD16lowFcεRIhighCD244high (88.5 ± 1.2%), (II) CD16highFcεRIhighCD244high (9.1 ± 0.4%), (III) CD16lowFcεRIlowCD244low (2.3 ± 1.3), and (IV) CD16highFcεRIlowCD244low (0.4 ± 0.1%). Prospective isolation confirmed basophilic-morphology of PBG I-III, but neutrophilic-morphology of PBG IV. Functional interrogation via IgE-crosslinking or IL-3 stimulation demonstrated that PBG I-II had significant increases in CD203c expression, whereas PBG III-IV remained unchanged compared with media-alone conditions. Thus, PBG III-IV could serve roles in non-IgE-mediated immunity. Our findings offer new perspectives in human basophil heterogeneity and the varying functional potential of these new subsets in health and disease.

13.
Nat Commun ; 10(1): 1185, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862852

RESUMEN

Cellular products derived from the activity of DNA, RNA, and protein synthesis collectively control cell identity and function. Yet there is little information on how these three biosynthesis activities are coordinated during transient and sparse cellular processes, such as activation and differentiation. Here, we describe Simultaneous Overview of tri-Molecule Biosynthesis (SOM3B), a molecular labeling and simultaneous detection strategy to quantify DNA, RNA, and protein synthesis in individual cells. Comprehensive interrogation of biosynthesis activities during transient cell states, such as progression through cell cycle or cellular differentiation, is achieved by partnering SOM3B with parallel quantification of select biomolecules with conjugated antibody reagents. Here, we investigate differential de novo DNA, RNA, and protein synthesis dynamics in transformed human cell lines, primary activated human immune cells, and across the healthy human hematopoietic continuum, all at a single-cell resolution.


Asunto(s)
ADN/biosíntesis , Biosíntesis de Proteínas , ARN/biosíntesis , Análisis de la Célula Individual/métodos , Médula Ósea/metabolismo , Ciclo Celular , Células HEK293 , Células HeLa , Voluntarios Sanos , Humanos , Células Jurkat , Leucocitos Mononucleares , Cultivo Primario de Células , Coloración y Etiquetado/métodos
14.
Nat Biotechnol ; 37(3): 259-266, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30742126

RESUMEN

Selective differentiation of naive T cells into multipotent T cells is of great interest clinically for the generation of cell-based cancer immunotherapies. Cellular differentiation depends crucially on division state and time. Here we adapt a dye dilution assay for tracking cell proliferative history through mass cytometry and uncouple division, time and regulatory protein expression in single naive human T cells during their activation and expansion in a complex ex vivo milieu. Using 23 markers, we defined groups of proteins controlled predominantly by division state or time and found that undivided cells account for the majority of phenotypic diversity. We next built a map of cell state changes during naive T-cell expansion. By examining cell signaling on this map, we rationally selected ibrutinib, a BTK and ITK inhibitor, and administered it before T cell activation to direct differentiation toward a T stem cell memory (TSCM)-like phenotype. This method for tracing cell fate across division states and time can be broadly applied for directing cellular differentiation.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Activación de Linfocitos/genética , Células Madre Multipotentes/citología , Rastreo Celular/métodos , Humanos , Análisis de la Célula Individual/métodos , Linfocitos T/citología
15.
Methods Mol Biol ; 1989: 55-81, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31077099

RESUMEN

The advent of mass cytometry (CyTOF®) has permitted simultaneous detection of more than 40 antibody parameters at the single-cell level, although a limited number of metal-labeled antibodies are commercially available. Here we present optimized and scalable protocols for conjugation of lanthanide as well as bismuth ions to immunoglobulin (Ig) using a maleimide-functionalized chelating polymer and for characterization of the conjugate. The maleimide functional group is reactive with cysteine sulfhydryl groups generated through partial reduction of the Ig Fc region. Incubation of Ig with polymer pre-loaded with lanthanide ions produces metal-labeled Ig without disrupting antigen specificity. Antibody recovery rates can be determined by UV spectrophotometry and frequently exceeds 60%. Each custom-conjugated antibody is validated using positive and negative cellular control populations and is titrated for optimal staining at concentrations ranging from 0.1 to 10 µg/mL. The preparation of metal-labeled antibodies can be completed in 4.5 h, and titration requires an additional 3-5 h.


Asunto(s)
Anticuerpos/química , Células/citología , Citometría de Flujo/métodos , Inmunofenotipificación/métodos , Isótopos/análisis , Espectrometría de Masas/métodos , Análisis de la Célula Individual/métodos , Quelantes/química , Humanos , Inmunoglobulinas/inmunología , Metales/química , Polímeros/química , Compuestos de Sulfhidrilo/química
16.
Biochem Biophys Res Commun ; 374(4): 641-6, 2008 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-18662671

RESUMEN

ARHGAP21 is highly expressed in the heart, which demonstrates activity over Cdc42 and interacts with proteins of the cytoskeleton and adherent junctions. The main cause of cardiac hypertrophy is mechanical stimulus; therefore we analyzed ARHGAP21 expression after acute mechanical stress in the myocardium and its association with FAK and PKCzeta. We demonstrated that ARHGAP21 is relocated to Z-lines and costameres after pressure overload, and interacts with PKCzeta and FAK in control rats (sham), rats submitted to aortic clamping and spontaneously hypertensive rats (SHR). Co-transfection using ARHGAP21 and PKCzeta constructions demonstrated that ARHGAP21 associates with PKCzeta-GST and endogenous FAK. Pulldown assay showed that ARHGAP21 binds to the C-terminal region of FAK. Moreover, ARHGAP21 binds to PKCzeta phosphorylated on Thr410 in sham and SHR. However, ARHGAP21 only binds to FAK phosphorylated on Tyr925 of SHR. Additionally, PKCzeta is phosphorylated by mechanical stimuli. These results suggest that ARHGAP21 may act as a signaling or scaffold protein of FAK and PKCzeta signaling pathways, developing an important function during cardiac stress.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/metabolismo , Chaperonas Moleculares/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Núcleo Celular , Modelos Animales de Enfermedad , Hipertrofia Ventricular Izquierda/patología , Masculino , Ratones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Fosforilación , Presión , Ratas , Ratas Endogámicas SHR , Ratas Wistar
17.
Nat Commun ; 7: 13101, 2016 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-27713415

RESUMEN

Mechanisms of haematopoietic and cardiac patterning remain poorly understood. Here we show that the BMP and Wnt signalling pathways are integrated in an endoglin (Eng)-dependent manner in cardiac and haematopoietic lineage specification. Eng is expressed in early mesoderm and marks both haematopoietic and cardiac progenitors. In the absence of Eng, yolk sacs inappropriately express the cardiac marker, Nkx2.5. Conversely, high levels of Eng in vitro and in vivo increase haematopoiesis and inhibit cardiogenesis. Levels of Eng determine the activation of both BMP and Wnt pathways, which are integrated downstream of Eng by phosphorylation of Smad1 by Gsk3. By interrogating Eng-dependent Wnt-mediated transcriptional changes, we identify Jdp2 as a key Eng-dependent Wnt target, sufficient to establish haematopoietic fate in early mesoderm when BMP and Wnt crosstalk is disturbed. These studies provide mechanistic insight into the integration of BMP and Wnt signalling in the establishment of haematopoietic and cardiac progenitors during embryogenesis.


Asunto(s)
Tipificación del Cuerpo/genética , Proteína Morfogenética Ósea 4/genética , Endoglina/genética , Hematopoyesis/genética , Proteína Wnt3/genética , Proteínas de Pez Cebra/genética , Animales , Tipificación del Cuerpo/fisiología , Línea Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Hematopoyesis/fisiología , Proteína Homeótica Nkx-2.5/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Transducción de Señal/genética , Proteína Smad1/metabolismo , Vía de Señalización Wnt/genética , Pez Cebra , beta Catenina/genética
18.
Nat Commun ; 4: 1549, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23462992

RESUMEN

Duchenne muscular dystrophy is a progressive and incurable neuromuscular disease caused by genetic and biochemical defects of the dystrophin-glycoprotein complex. Here we show the regenerative potential of myogenic progenitors derived from corrected dystrophic induced pluripotent stem cells generated from fibroblasts of mice lacking both dystrophin and utrophin. We correct the phenotype of dystrophic induced pluripotent stem cells using a Sleeping Beauty transposon system carrying the micro-utrophin gene, differentiate these cells into skeletal muscle progenitors and transplant them back into dystrophic mice. Engrafted muscles displayed large numbers of micro-utrophin-positive myofibers, with biochemically restored dystrophin-glycoprotein complex and improved contractile strength. The transplanted cells seed the satellite cell compartment, responded properly to injury and exhibit neuromuscular synapses. We also detect muscle engraftment after systemic delivery of these corrected progenitors. These results represent an important advance towards the future treatment of muscular dystrophies using genetically corrected autologous induced pluripotent stem cells.


Asunto(s)
Terapia Genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/terapia , Animales , Compartimento Celular , Distrofina/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Contracción Isométrica , Ratones , Ratones Noqueados , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Distrofia Muscular Animal/patología , Distrofia Muscular Animal/fisiopatología , Recuperación de la Función , Regeneración , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/patología , Sinapsis/metabolismo , Utrofina/genética
19.
Exp Hematol ; 40(10): 837-46, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22728030

RESUMEN

Increasing evidence points to endoglin (Eng), an accessory receptor for the transforming growth factor-ß superfamily commonly associated with the endothelial lineage, as an important regulator of the hematopoietic lineage. We have shown that lack of Eng results in reduced numbers of primitive erythroid colonies as well as downregulation of key hematopoietic genes. To determine the effect of Eng overexpression in hematopoietic development, we generated a doxycycline-inducible embryonic stem cell line. Our results demonstrate that induction of Eng during embryoid body differentiation leads to a significant increase in the frequency of hematopoietic progenitors, in particular, the erythroid lineage, which correlated with upregulation of Scl, Gata1, Runx1, and embryonic globin. Interestingly, activation of the hematopoietic program happened at the expense of endothelial and cardiac cells, as differentiation into these mesoderm lineages was compromised. Eng-induced enhanced erythroid activity was accompanied by high levels of Smad1 phosphorylation. This effect was attenuated by addition of a bone morphogenetic protein (BMP) signaling inhibitor to these cultures. Among the BMPs, BMP4 is well known for its role in hematopoietic specification from mesoderm by promoting expression of several hematopoietic genes, including Scl. Because Scl is considered the master regulator of the hematopoietic program, we investigated whether Scl would be capable of rescuing the defective hematopoietic phenotype observed in Eng(-/-) embryonic stem cells. Scl expression in Eng-deficient embryonic stem cells resulted in increased erythroid colony-forming activity and upregulation of Gata1 and Gata2, positioning Eng upstream of Scl. Taken together, these findings support the premise that Eng modulates the hematopoietic transcriptional network, most likely through regulation of BMP4 signaling.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Cuerpos Embrioides/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Regulación de la Expresión Génica/fisiología , Hematopoyesis/fisiología , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Cuerpos Embrioides/citología , Endoglina , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Globinas/genética , Globinas/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Fosforilación/fisiología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/fisiología , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA