Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Genome Res ; 30(1): 127-137, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31831592

RESUMEN

Bone marrow-derived mesenchymal stem cells (MSCs) differentiate into osteoblasts upon stimulation by signals present in their niche. Because the global signaling cascades involved in the early phases of MSCs osteoblast (OB) differentiation are not well-defined, we used quantitative mass spectrometry to delineate changes in human MSCs proteome and phosphoproteome during the first 24 h of their OB lineage commitment. The temporal profiles of 6252 proteins and 15,059 phosphorylation sites suggested at least two distinct signaling waves: one peaking within 30 to 60 min after stimulation and a second upsurge after 24 h. In addition to providing a comprehensive view of the proteome and phosphoproteome dynamics during early MSCs differentiation, our analyses identified a key role of serine/threonine protein kinase D1 (PRKD1) in OB commitment. At the onset of OB differentiation, PRKD1 initiates activation of the pro-osteogenic transcription factor RUNX2 by triggering phosphorylation and nuclear exclusion of the histone deacetylase HDAC7.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Fosfoproteínas/metabolismo , Proteoma , Proteómica , Humanos , Filogenia , Proteómica/métodos
2.
Mol Cell Proteomics ; 18(Suppl 1): S118-S131, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30622161

RESUMEN

G-protein coupled receptors (GPCRs) belong to the seven transmembrane receptor superfamily that transduce signals via G proteins in response to external stimuli to initiate different intracellular signaling pathways which culminate in specific cellular responses. The expression of diverse GPCRs at the plasma membrane of human spermatozoa suggests their involvement in the regulation of sperm fertility. However, the signaling events downstream of many GPCRs in spermatozoa remain uncharacterized. Here, we selected the kappa-opioid receptor (KOR) as a study model and applied phosphoproteomic approach based on TMT labeling and LC-MS/MS analyses. Quantitative coverage of more than 5000 proteins with over 3500 phosphorylation sites revealed changes in the phosphorylation levels of sperm-specific proteins involved in the regulation of the sperm fertility in response to a specific agonist of KOR, U50488H. Further functional studies indicate that KOR could be involved in the regulation of sperm fertile capacity by modulation of calcium channels. Our findings suggest that human spermatozoa possess unique features in the molecular mechanisms downstream of GPCRs which could be key regulators of sperm fertility and improved knowledge of these specific processes may contribute to the development of useful biochemical tools for diagnosis and treatment of male infertility.


Asunto(s)
Fosfoproteínas/metabolismo , Proteómica , Receptores Opioides kappa/metabolismo , Espermatozoides/metabolismo , Reacción Acrosómica , Canales de Calcio/metabolismo , Humanos , Masculino , Fosforilación , Proteoma/metabolismo , Receptores Opioides kappa/agonistas
3.
Mol Cell Proteomics ; 16(8): 1433-1446, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28572092

RESUMEN

Cylindromatosis tumor suppressor protein (CYLD) is a deubiquitinase, best known as an essential negative regulator of the NFkB pathway. Previous studies have suggested an involvement of CYLD in epidermal growth factor (EGF)-dependent signal transduction as well, as it was found enriched within the tyrosine-phosphorylated complexes in cells stimulated with the growth factor. EGF receptor (EGFR) signaling participates in central cellular processes and its tight regulation, partly through ubiquitination cascades, is decisive for a balanced cellular homeostasis. Here, using a combination of mass spectrometry-based quantitative proteomic approaches with biochemical and immunofluorescence strategies, we demonstrate the involvement of CYLD in the regulation of the ubiquitination events triggered by EGF. Our data show that CYLD regulates the magnitude of ubiquitination of several major effectors of the EGFR pathway by assisting the recruitment of the ubiquitin ligase Cbl-b to the activated EGFR complex. Notably, CYLD facilitates the interaction of EGFR with Cbl-b through its Tyr15 phosphorylation in response to EGF, which leads to fine-tuning of the receptor's ubiquitination and subsequent degradation. This represents a previously uncharacterized strategy exerted by this deubiquitinase and tumors suppressor for the negative regulation of a tumorigenic signaling pathway.


Asunto(s)
Enzima Desubiquitinante CYLD/metabolismo , Receptores ErbB/metabolismo , Proteolisis , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Ubiquitinación , Cromatografía Liquida , Enzima Desubiquitinante CYLD/genética , Células HeLa , Humanos , Fosforilación , Proteómica , Espectrometría de Masas en Tándem , Tirosina/metabolismo
4.
J Proteome Res ; 17(1): 296-304, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29091453

RESUMEN

Modulation of protein activities by reversible post-translational modifications (PTMs) is a major molecular mechanism involved in the control of virtually all cellular processes. One of these PTMs is ubiquitination, which regulates key processes including protein degradation, cell cycle, DNA damage repair, and signal transduction. Because of its importance for numerous cellular functions, ubiquitination has become an intense topic of research in recent years, and proteomics tools have greatly facilitated the identification of many ubiquitination targets. Taking advantage of the StUbEx strategy for exchanging the endogenous ubiquitin with an epitope-tagged version, we created a modified system, StUbEx PLUS, which allows precise mapping of ubiquitination sites by mass spectrometry. Application of StUbEx PLUS to U2OS cells treated with proteasomal inhibitors resulted in the identification of 41 589 sites on 7762 proteins, which thereby revealed the ubiquitous nature of this PTM and demonstrated the utility of the approach for comprehensive ubiquitination studies at site-specific resolution.


Asunto(s)
Sitios de Unión , Péptidos/aislamiento & purificación , Ubiquitina/metabolismo , Ubiquitinación , Línea Celular , Humanos , Espectrometría de Masas , Péptidos/metabolismo , Procesamiento Proteico-Postraduccional
5.
Mol Cell Proteomics ; 15(6): 2076-92, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27067055

RESUMEN

Anti-cancer immunotherapies commonly rely on the use of interleukin-2 (IL-2) to promote the expansion of T lymphocytes. IL-2- dependent proliferation is the culmination of a complex network of phosphorylation-driven signaling events that impact on gene transcription through mechanisms that are not clearly understood. To study the role of IL-2 in the regulation of nuclear protein function we have performed an unbiased mass spectrometry-based study of the nuclear phosphoproteome of resting and IL-2-treated CD4(+) T lymphocytes. We detected 8521distinct phosphosites including many that are not yet reported in curated phosphorylation databases. Although most phosphorylation sites remained unaffected upon IL-2 treatment, 391 sites corresponding to 288 gene products showed robust IL-2-dependent regulation. Importantly, we show that ATP-citrate lyase (ACLY) is a key phosphoprotein effector of IL-2-mediated T-cell responses. ACLY becomes phosphorylated on serine 455 in T lymphocytes upon IL-2-driven activation of AKT, and depletion or inactivation of ACLY compromises IL-2-promoted T-cell growth. Mechanistically, we demonstrate that ACLY is required for enhancing histone acetylation levels and inducing the expression of cell cycle regulating genes in response to IL-2. Thus, the metabolic enzyme ACLY emerges as a bridge between cytokine signaling and proliferation of T lymphocytes, and may be an attractive candidate target for the development of more efficient anti-cancer immunotherapies.


Asunto(s)
ATP Citrato (pro-S)-Liasa/aislamiento & purificación , Linfocitos T CD4-Positivos/citología , Interleucina-2/farmacología , Proteómica/métodos , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Proteínas Nucleares/análisis , Proteínas Nucleares/efectos de los fármacos , Fosfoproteínas/análisis , Fosfoproteínas/efectos de los fármacos
6.
J Proteome Res ; 16(1): 106-121, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-27463037

RESUMEN

It remains a paradox that IL-2 and IL-15 can differentially modulate the immune response using the same signaling receptors. We have previously dissected the phosphotyrosine-driven signaling cascades triggered by both cytokines in Kit225 T-cells, unveiling subtle differences that may contribute to their functional dichotomy. In this study, we aimed to decipher the receptor complex assembly in IL-2- and IL-15-activated T-lymphocytes that is highly orchestrated by site-specific phosphorylation events. Comparing the cytokine-induced interactome of the interleukin receptor beta and gamma subunits shared by the two cytokines, we defined the components of the early IL-2 and IL-15 receptor-associated complex discovering novel constituents. Additionally, phosphopeptide-directed analysis allowed us to detect several cytokine-dependent and -independent phosphorylation events within the activated receptor complex including novel phosphorylated sites located in the cytoplasmic region of IL-2 receptor ß subunit (IL-2Rß). We proved that the distinct phosphorylations induced by the cytokines serve for recruiting different types of effectors to the initial receptor/ligand complex. Overall, our study sheds new light into the initial molecular events triggered by IL-2 and IL-15 and constitutes a further step toward a better understanding of the early signaling aspects of the two closely related cytokines in T-lymphocytes.


Asunto(s)
Subunidad gamma Común de Receptores de Interleucina/inmunología , Interleucina-15/farmacología , Subunidad beta del Receptor de Interleucina-2/inmunología , Interleucina-2/farmacología , Janus Quinasa 1/inmunología , Janus Quinasa 3/inmunología , Linfocitos T/efectos de los fármacos , Secuencia de Aminoácidos , Línea Celular Tumoral , Regulación de la Expresión Génica , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Interleucina-15/genética , Interleucina-15/inmunología , Interleucina-2/genética , Interleucina-2/inmunología , Subunidad beta del Receptor de Interleucina-2/genética , Janus Quinasa 1/genética , Janus Quinasa 3/genética , Activación de Linfocitos , Fosforilación , Fosfotirosina/genética , Fosfotirosina/inmunología , Unión Proteica , Mapeo de Interacción de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología , Transducción de Señal , Linfocitos T/citología , Linfocitos T/inmunología
7.
Proteomics ; 15(2-3): 520-31, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25142963

RESUMEN

Common γ-chain family of cytokines (IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, where IL stands for interleukin) are key regulators of the immune homeostasis that exhibit pleiotropic biological activities and even sometimes redundant roles as a result of the utilization of the same receptor subunit. However, they also exert distinct functions that make each of them to be indispensable. For instance, all family members can act as T-cell growth factors; however, we found that IL-15 but not IL-7 can replace IL-2 to promote and sustain the proliferation of Kit225T cells. In addition to the γ-chain, IL-2 and IL-15 share the ß-chain, which creates the paradox of how they can trigger diverse phenotypes despite signaling through the same receptors. To investigate this paradigm, we combined SILAC with enrichment of tyrosine-phosphorylated proteins and peptides followed by mass spectrometric analysis to quantitatively assess the signaling networks triggered downstream IL-2/IL-2R and IL-15/IL-15R. This study confirmed that the transduction pathways initiated by both cytokines are highly similar and revealed that the main signaling branches, JAK/STAT, RAS/MAPK and PI3K/AKT, were nearly equivalently activated in response to both ILs. Despite that, our study revealed that receptor internalization rates differ in IL-2- and IL-15-treated cells indicating a discrete modulation of cytokine signaling. All MS data have been deposited in the ProteomeXchange with identifier PXD001129 (http://proteomecentral.proteomexchange.org/dataset/PXD001129).


Asunto(s)
Interleucina-15/inmunología , Interleucina-2/inmunología , Transducción de Señal , Linfocitos T/inmunología , Línea Celular Tumoral , Proliferación Celular , Endocitosis , Humanos , Interleucina-7/inmunología , Fosforilación , Proteómica , Linfocitos T/citología
8.
J Proteome Res ; 14(8): 3348-61, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26074025

RESUMEN

Muscle stem cells, or satellite cells, play an important role in the maintenance and repair of muscle tissue and have the capacity to proliferate and differentiate in response to physiological or environmental changes. Although they have been extensively studied, the key regulatory steps and the complex temporal protein dynamics accompanying the differentiation of primary human muscle cells remain poorly understood. Here, we demonstrate the advantages of applying a MS-based quantitative approach, stable isotope labeling by amino acids in cell culture (SILAC), for studying human myogenesis in vitro and characterize the fine-tuned changes in protein expression underlying the dramatic phenotypic conversion of primary mononucleated human muscle cells during in vitro differentiation to form multinucleated myotubes. Using an exclusively optimized triple encoding SILAC procedure, we generated dynamic expression profiles during the course of myogenic differentiation and quantified 2240 proteins, 243 of which were regulated. These changes in protein expression occurred in sequential waves and underlined vast reprogramming in key processes governing cell fate decisions, i.e., cell cycle withdrawal, RNA metabolism, cell adhesion, proteolysis, and cytoskeletal organization. In silico transcription factor target analysis demonstrated that the observed dynamic changes in the proteome could be attributed to a cascade of transcriptional events involving key myogenic regulatory factors as well as additional regulators not yet known to act on muscle differentiation. In addition, we created of a dynamic map of the developing myofibril, providing valuable insights into the formation and maturation of the contractile apparatus in vitro. Finally, our SILAC-based quantitative approach offered the possibility to follow the expression profiles of several muscle disease-associated proteins simultaneously and therefore could be a valuable resource for future studies investigating pathogenesis of degenerative muscle disorders as well as assessing new therapeutic strategies.


Asunto(s)
Diferenciación Celular , Fibras Musculares Esqueléticas/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Células Satélite del Músculo Esquelético/metabolismo , Aminoácidos/metabolismo , Western Blotting , Células Cultivadas , Cromatografía Liquida , Análisis por Conglomerados , Humanos , Inmunohistoquímica , Recién Nacido , Marcaje Isotópico/métodos , Cinética , Fibras Musculares Esqueléticas/citología , Proteoma/clasificación , Células Satélite del Músculo Esquelético/citología , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem , Factores de Tiempo
9.
J Proteome Res ; 13(9): 4192-204, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25093938

RESUMEN

Post-translational modification of proteins with the small polypeptide ubiquitin plays a pivotal role in many cellular processes, altering protein lifespan, location, and function and regulating protein-protein interactions. Ubiquitination exerts its diverse functions through complex mechanisms by formation of different polymeric chains and subsequent recognition of the ubiquitin signal by specific protein interaction domains. Despite some recent advances in the analytical tools for the analysis of ubiquitination by mass spectrometry, there is still a need for additional strategies suitable for investigation of cellular ubiquitination at the proteome level. Here, we present a stable tagged ubiquitin exchange (StUbEx) cellular system in which endogenous ubiquitin is replaced with an epitope-tagged version, thereby allowing specific and efficient affinity purification of ubiquitinated proteins for global analyses of protein ubiquitination. Importantly, the overall level of ubiquitin in the cell remains virtually unchanged, thus avoiding ubiquitination artifacts associated with overexpression. The efficiency and reproducibility of the method were assessed through unbiased analysis of epidermal growth factor (EGF) signaling by quantitative mass spectrometry, covering over 3400 potential ubiquitinated proteins. The StUbEx system is applicable to virtually any cell line and can be readily adapted to any of the ubiquitin-like post-translational modifications.


Asunto(s)
Marcaje Isotópico/métodos , Proteómica/métodos , Ubiquitina/química , Ubiquitina/metabolismo , Cromatografía de Afinidad/métodos , Bases de Datos de Proteínas , Células HeLa , Histidina , Humanos , Oligopéptidos , Proteínas Recombinantes de Fusión , Reproducibilidad de los Resultados , Ubiquitinación
10.
Mol Cell Proteomics ; 11(10): 989-1007, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22801418

RESUMEN

It is well established that bone forming cells (osteoblasts) secrete proteins with autocrine, paracrine, and endocrine function. However, the identity and functional role for the majority of these secreted and differentially expressed proteins during the osteoblast (OB) differentiation process, is not fully established. To address these questions, we quantified the temporal dynamics of the human stromal (mesenchymal, skeletal) stem cell (hMSC) secretome during ex vivo OB differentiation using stable isotope labeling by amino acids in cell culture (SILAC). In addition, we employed pulsed SILAC labeling to distinguish genuine secreted proteins from intracellular contaminants. We identified 466 potentially secreted proteins that were quantified at 5 time-points during 14-days ex vivo OB differentiation including 41 proteins known to be involved in OB functions. Among these, 315 proteins exhibited more than 2-fold up or down-regulation. The pulsed SILAC method revealed a strong correlation between the fraction of isotope labeling and the subset of proteins known to be secreted and involved in OB differentiation. We verified SILAC data using qRT-PCR analysis of 9 identified potential novel regulators of OB differentiation. Furthermore, we studied the biological effects of one of these proteins, the hormone stanniocalcin 2 (STC2) and demonstrated its autocrine effects in enhancing osteoblastic differentiation of hMSC. In conclusion, combining complete and pulsed SILAC labeling facilitated the identification of novel factors produced by hMSC with potential role in OB differentiation. Our study demonstrates that the secretome of osteoblastic cells is more complex than previously reported and supports the emerging evidence that osteoblastic cells secrete proteins with endocrine functions and regulate cellular processes beyond bone formation.


Asunto(s)
Glicoproteínas/farmacología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/metabolismo , Proteoma/metabolismo , Aminoácidos/metabolismo , Diferenciación Celular , Línea Celular Transformada , Células Cultivadas , Cromatografía Liquida , Glicoproteínas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Marcaje Isotópico , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Proteoma/genética , Proteómica , Espectrometría de Masas en Tándem
11.
Nature ; 439(7073): 204-7, 2006 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-16306937

RESUMEN

Toll-like receptors (TLRs) are activated by pathogen-associated molecular patterns to induce innate immune responses and production of pro-inflammatory cytokines, interferons and anti-inflammatory cytokines. TLRs activate downstream effectors through adaptors that contain Toll/interleukin-1 receptor (TIR) domains, but the mechanisms accounting for diversification of TLR effector functions are unclear. To dissect biochemically TLR signalling, we established a system for isolating signalling complexes assembled by dimerized adaptors. Using MyD88 as a prototypical adaptor, we identified TNF receptor-associated factor 3 (TRAF3) as a new component of TIR signalling complexes that is recruited along with TRAF6. Using myeloid cells from TRAF3- and TRAF6-deficient mice, we show that TRAF3 is essential for the induction of type I interferons (IFN) and the anti-inflammatory cytokine interleukin-10 (IL-10), but is dispensable for expression of pro-inflammatory cytokines. In fact, TRAF3-deficient cells overproduce pro-inflammatory cytokines owing to defective IL-10 production. Despite their structural similarity, the functions of TRAF3 and TRAF6 are largely distinct. TRAF3 is also recruited to the adaptor TRIF (Toll/IL-1 receptor domain-containing adaptor-inducing IFN-beta) and is required for marshalling the protein kinase TBK1 (also called NAK) into TIR signalling complexes, thereby explaining its unique role in activation of the IFN response.


Asunto(s)
Transducción de Señal , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Receptores Toll-Like/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Antígenos de Diferenciación/química , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Línea Celular , Dimerización , Regulación de la Expresión Génica , Inmunidad Innata , Interferones/biosíntesis , Interleucina-10/biosíntesis , Ratones , Células Mieloides/metabolismo , Factor 88 de Diferenciación Mieloide , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Especificidad por Sustrato , Factor 6 Asociado a Receptor de TNF/deficiencia , Factor 6 Asociado a Receptor de TNF/genética
12.
Mol Cell Proteomics ; 9(11): 2482-96, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20631206

RESUMEN

During recent years, increased efforts have focused on elucidating the secretory function of skeletal muscle. Through secreted molecules, skeletal muscle affects local muscle biology in an auto/paracrine manner as well as having systemic effects on other tissues. Here we used a quantitative proteomics platform to investigate the factors secreted during the differentiation of murine C2C12 skeletal muscle cells. Using triple encoding stable isotope labeling by amino acids in cell culture, we compared the secretomes at three different time points of muscle differentiation and followed the dynamics of protein secretion. We identified and quantitatively analyzed 635 secreted proteins, including 35 growth factors, 40 cytokines, and 36 metallopeptidases. The extensive presence of these proteins that can act as potent signaling mediators to other cells and tissues strongly highlights the important role of the skeletal muscle as a prominent secretory organ. In addition to previously reported molecules, we identified many secreted proteins that have not previously been shown to be released from skeletal muscle cells nor shown to be differentially released during the process of myogenesis. We found 188 of these secreted proteins to be significantly regulated during the process of myogenesis. Comparative analyses of selected secreted proteins revealed little correlation between their mRNA and protein levels, indicating pronounced regulation by posttranscriptional mechanisms. Furthermore, analyses of the intracellular levels of members of the semaphorin family and their corresponding secretion dynamics demonstrated that the release of secreted proteins is tightly regulated by the secretory pathway, the stability of the protein, and/or the processing of secreted proteins. Finally, we provide 299 unique hydroxyproline sites mapping to 48 distinct secreted proteins and have discovered a novel hydroxyproline motif.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas Musculares , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular , Marcaje Isotópico , Espectrometría de Masas/métodos , Ratones , Datos de Secuencia Molecular , Proteínas Musculares/química , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mioblastos/citología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteómica/métodos , Semaforinas/genética , Semaforinas/metabolismo
13.
Mol Cell Proteomics ; 8(5): 959-70, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19151416

RESUMEN

Stable isotope labeling by amino acids in cell culture (SILAC) is a powerful quantitative proteomics platform for comprehensive characterization of complex biological systems. However, the potential of SILAC-based approaches has not been fully utilized in human embryonic stem cell (hESC) research mainly because of the complex nature of hESC culture conditions. Here we describe complete SILAC labeling of hESCs with fully preserved pluripotency, self-renewal capabilities, and overall proteome status that was quantitatively analyzed to a depth of 1556 proteins and 527 phosphorylation events. SILAC-labeled hESCs appear to be perfectly suitable for functional studies, and we exploited a SILAC-based proteomics strategy for discovery of hESC-specific surface markers. We determined and quantitatively compared the membrane proteomes of the self-renewing versus differentiating cells of two distinct human embryonic stem cell lines. Of the 811 identified membrane proteins, six displayed significantly higher expression levels in the undifferentiated state compared with differentiating cells. This group includes the established marker CD133/Prominin-1 as well as novel candidates for hESC surface markers: Glypican-4, Neuroligin-4, ErbB2, receptor-type tyrosine-protein phosphatase zeta (PTPRZ), and Glycoprotein M6B. Our study also revealed 17 potential markers of hESC differentiation as their corresponding protein expression levels displayed a dramatic increase in differentiated embryonic stem cell populations.


Asunto(s)
Aminoácidos/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Marcaje Isotópico , Proteínas de la Membrana/análisis , Proteoma/análisis , Animales , Biomarcadores/metabolismo , Proliferación Celular , Células Cultivadas , Medios de Cultivo Condicionados , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Ratones , Fosfoproteínas/análisis , Células Madre Pluripotentes/citología , Proteómica , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
Proc Natl Acad Sci U S A ; 105(7): 2451-6, 2008 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-18268350

RESUMEN

The insulin signaling pathway is of pivotal importance in metabolic diseases, such as diabetes, and in cellular processes, such as aging. Insulin activates a tyrosine phosphorylation cascade that branches to create a complex network affecting multiple biological processes. To understand the full spectrum of the tyrosine phosphorylation cascade, we have defined the tyrosine-phosphoproteome of the insulin signaling pathway, using high resolution mass spectrometry in combination with phosphotyrosine immunoprecipitation and stable isotope labeling by amino acids in cell culture (SILAC) in differentiated brown adipocytes. Of 40 identified insulin-induced effectors, 7 have not previously been described in insulin signaling, including SDR, PKCdelta binding protein, LRP-6, and PISP/PDZK11, a potential calcium ATPase binding protein. A proteomic interaction screen with PISP/PDZK11 identified the calcium transporting ATPase SERCA2, supporting a connection to calcium signaling. The combination of quantitative phosphoproteomics with cell culture models provides a powerful strategy to dissect the insulin signaling pathways in intact cells.


Asunto(s)
Insulina/farmacología , Transducción de Señal/efectos de los fármacos , Adipocitos Marrones/citología , Adipocitos Marrones/efectos de los fármacos , Aminoácidos/química , Aminoácidos/metabolismo , Animales , Transporte Biológico , Calcio/metabolismo , Proteínas Portadoras/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Ratones , Miosinas/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica , Proteómica , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo
15.
Sci Rep ; 11(1): 3583, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-33574425

RESUMEN

Human sperm protein associated with the nucleus on the X chromosome (SPANX) genes encode a protein family (SPANX-A, -B, -C and -D), whose expression is limited to the testis and spermatozoa in normal tissues and various tumour cells. SPANX-A/D proteins have been detected in metastatic melanoma cells, but their contribution to cancer development and the underlying molecular mechanisms of skin tumourigenesis remain unknown. Combining functional and proteomic approaches, the present work describes the presence of SPANX-A/D in primary and metastatic human melanoma cells and how it promotes pro-tumoural processes such as cell proliferation, motility and migration. We provide insights into the molecular features of skin tumourigenesis, describing for the first time a multifunctional role of the SPANX-A/D protein family in nuclear function, energy metabolism and cell survival, considered key hallmarks of cancer. A better comprehension of the SPANX-A/D protein subfamily and its molecular mechanisms will help to describe new aspects of tumour cell biology and develop new therapeutic targets and tumour-directed pharmacological drugs for skin tumours.


Asunto(s)
Carcinogénesis/genética , Melanoma/genética , Proteínas Nucleares/genética , Proteómica , Secuencia de Aminoácidos/genética , Núcleo Celular/genética , Núcleo Celular/patología , Cromosomas Humanos X/genética , Humanos , Masculino , Melanoma/patología , Proteínas Nucleares/clasificación , Homología de Secuencia de Aminoácido , Espermatozoides/metabolismo , Espermatozoides/patología , Testículo/crecimiento & desarrollo , Testículo/patología
16.
Sci Rep ; 11(1): 21839, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34750429

RESUMEN

The intimate association between obesity and type II diabetes urges for a deeper understanding of adipocyte function. We and others have previously delineated a role for the tumor suppressor p53 in adipocyte biology. Here, we show that mice haploinsufficient for MDM2, a key regulator of p53, in their adipose stores suffer from overt obesity, glucose intolerance, and hepatic steatosis. These mice had decreased levels of circulating palmitoleic acid [non-esterified fatty acid (NEFA) 16:1] concomitant with impaired visceral adipose tissue expression of Scd1 and Ffar4. A similar decrease in Scd and Ffar4 expression was found in in vitro differentiated adipocytes with perturbed MDM2 expression. Lowered MDM2 levels led to nuclear exclusion of the transcriptional cofactors, MORC2 and LIPIN1, and thereby possibly hampered adipocyte function by antagonizing LIPIN1-mediated PPARγ coactivation. Collectively, these data argue for a hitherto unknown interplay between MDM2 and MORC2/LIPIN1 involved in balancing adipocyte function.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Resistencia a la Insulina/fisiología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos Monoinsaturados/sangre , Hígado Graso/genética , Hígado Graso/metabolismo , Femenino , Redes Reguladoras de Genes , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Haploinsuficiencia/genética , Haploinsuficiencia/fisiología , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , PPAR gamma/metabolismo , Fosfatidato Fosfatasa , Proteínas Proto-Oncogénicas c-mdm2/deficiencia , Proteínas Proto-Oncogénicas c-mdm2/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
17.
J Proteome Res ; 9(5): 2734-42, 2010 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-20222723

RESUMEN

We have used stable isotope labeling by amino acids in cell culture (SILAC), in combination with high-resolution mass spectrometry, to identify common and discrete components of the respective receptor tyrosine kinase-dependent phosphotyrosine-associated networks induced by acute stimulation of A549 lung adenocarcinoma cells with EGF or HGF. In total, we obtained quantitative information for 274 proteins, which respond to either or both stimuli by >1.5 fold changes in enrichment, following immuno-precipitation with antiphosphotyrosine antibodies. The data reveal a high degree of overlap between the respective signaling networks but also clear points of departure. A small number of HGF specific effectors were identified including myosin-X, galectin-1, ELMO2 and EphrinB1, while a larger set of EGF specific effectors (39 proteins) includes both novel (e.g., MAP4K3) and established components of receptor tyrosine kinase receptor signaling pathways. Using available protein-interaction data the identified proteins have been assembled into a highly connected network that can be visualized using the Cytoscape tool.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Fosfotirosina/metabolismo , Proteoma/análisis , Proteómica/métodos , Western Blotting , Línea Celular Tumoral , Humanos , Inmunoprecipitación , Marcaje Isotópico , Mapeo de Interacción de Proteínas , Proteínas Tirosina Quinasas/metabolismo , Proteoma/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal
18.
Sci Rep ; 10(1): 5625, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32221341

RESUMEN

Human sperm protein associated with the nucleus on the X chromosome (SPANX) genes encode a protein family (SPANX-A, -B, -C and -D), whose expression is limited to the testis and spermatozoa in normal tissues and to a wide variety of tumour cells. Present only in hominids, SPANX-A/D is exclusively expressed in post-meiotic spermatids and mature spermatozoa. However, the biological role of the protein family in human spermatozoa is largely unknown. Combining proteomics and molecular approaches, the present work describes the presence of all isoforms of SPANX-A/D in human spermatozoa and novel phosphorylation sites of this protein family. In addition, we identify 307 potential SPANX-A/D interactors related to nuclear envelop, chromatin organisation, metabolism and cilia movement. Specifically, SPANX-A/D interacts with fumarate hydratase and colocalises with both fumarate hydratase and Tektin 1 proteins, involved in meeting energy demands for sperm motility, and with nuclear pore complex nucleoporins. We provide insights into the molecular features of sperm physiology describing for the first time a multifunctional role of SPANX-A/D protein family in nuclear envelope, sperm movement and metabolism, considered key functions for human spermatozoa. SPANX-A/D family members, therefore, might be promising targets for sperm fertility management.


Asunto(s)
Proteínas Nucleares/metabolismo , Motilidad Espermática/fisiología , Espermatozoides/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/genética , Células HEK293 , Células HeLa , Hominidae/metabolismo , Humanos , Masculino , Membrana Nuclear/metabolismo , Fosforilación/genética , Isoformas de Proteínas/metabolismo , Proteómica/métodos , Homología de Secuencia de Aminoácido , Espermátides/metabolismo , Testículo/metabolismo , Factores de Transcripción/metabolismo , Cromosoma X/genética
19.
Reprod Biol ; 20(3): 300-306, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32684427

RESUMEN

The kappa-opioid receptor (KOR) is involved in the regulation of the fertilizing capacity of human sperm. Recently, a testicular-specific protein family, SPANX-A/D, has also been found to be involved in regulating this process. In order to determine if KOR has a role in the regulation of sperm fertility through the SPANX-A/D protein family, we activated the kappa opioid receptor adding its selective agonist, U50488H to normozoospermic human spermatozoa. Then, we performed immunofluorescence assays and immunoprecipitation experiments followed by LC-MS/MS. According to our results, KOR activation may cause the translocation of SPANX-A/D into the nucleus of human spermatozoa. Phosphoproteomic studies show that KOR does not cause phosphorylation changes in SPANX-A/D residues. However, interactome assays demonstrate that KOR activation provokes changes in SPANX-A/D potential interactors involved in sperm motility, energy metabolism and nuclear processes. Taking these results into account, KOR may regulate human sperm fertility through SPANX-A/D protein family, modifying its subcellular location and interactions. Although further studies are needed, this finding could help us describing the molecular mechanisms underlying sperm fertility as well as developing new strategies for treating infertility.


Asunto(s)
Proteínas Nucleares/metabolismo , Receptores Opioides kappa/metabolismo , Espermatozoides/metabolismo , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Analgésicos no Narcóticos/farmacología , Humanos , Masculino , Fosforilación/efectos de los fármacos , Motilidad Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Espectrometría de Masas en Tándem
20.
Oncol Rep ; 41(2): 742-752, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30483808

RESUMEN

Colon cancer is one of the most frequently occurring types of cancers in the world. Primary tumours are treated very efficiently, but the metastatic cases are known to have severe outcomes. Therefore, the aim of the present study was to obtain a greater understanding of the transformation of primary colon cancer cells into metastatic phenotypes. Small changes in protein expression provoke the metastatic phenotype transformation. More sensitive methods to detect small variations are required. A murine colon cancer cell line with metastatic characteristics in a very early phase was created in order to investigate the first steps of transformation using a murine liver metastasis model. The protein expression patterns of metastatic and non­metastatic cells were compared using the stable isotope labelling by amino acids in cell culture method in combination with mass spectrometry. Quantitative proteomics data indicated that nicotinamide adenine dinucleotide hydride (NADH) dehydrogenase complex I was overexpressed in metastatic cells with respect to non­metastatic cells. Since the NADH dehydrogenase complex catalyses the oxidation of NADH to NAD+, the functionality of the complex was studied by measuring the amount of NADH. The results revealed that metastatic cells accumulate more NADH and reactive oxygen species. In addition, the mitochondrial membrane potential of metastatic cells was lower than that of non­metastatic cells, indicating that the activity of NADH dehydrogenase and the mitochondrial oxidative chain were decreased in metastatic cells. During the incipient transformation of primary cancer cells, NADH dehydrogenase complex I was overexpressed but then became inactive due to the Warburg effect, which inhibits mitochondrial activity. In the first step of transformation, the high energy demand required in an adverse environment is fulfilled by overexpressing components of the respiratory chain, a fact that should be considered for future anti­metastatic therapies.


Asunto(s)
Neoplasias del Colon/patología , Complejo I de Transporte de Electrón/metabolismo , Neoplasias Hepáticas/patología , Mitocondrias/patología , NADH Deshidrogenasa/metabolismo , Animales , Línea Celular Tumoral , Neoplasias Hepáticas/secundario , Masculino , Ratones , Ratones Endogámicos BALB C , Mitocondrias/metabolismo , NAD/análisis , NAD/metabolismo , Especies Reactivas de Oxígeno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA