Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(19): e2212118120, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37126683

RESUMEN

The prognosis and treatment outcomes of heart failure (HF) patients rely heavily on disease etiology, yet the majority of underlying signaling mechanisms are complex and not fully elucidated. Phosphorylation is a major point of protein regulation with rapid and profound effects on the function and activity of protein networks. Currently, there is a lack of comprehensive proteomic and phosphoproteomic studies examining cardiac tissue from HF patients with either dilated dilated cardiomyopathy (DCM) or ischemic cardiomyopathy (ICM). Here, we used a combined proteomic and phosphoproteomic approach to identify and quantify more than 5,000 total proteins with greater than 13,000 corresponding phosphorylation sites across explanted left ventricle (LV) tissue samples, including HF patients with DCM vs. nonfailing controls (NFC), and left ventricular infarct vs. noninfarct, and periinfarct vs. noninfarct regions of HF patients with ICM. Each pair-wise comparison revealed unique global proteomic and phosphoproteomic profiles with both shared and etiology-specific perturbations. With this approach, we identified a DCM-associated hyperphosphorylation cluster in the cardiomyocyte intercalated disc (ICD) protein, αT-catenin (CTNNA3). We demonstrate using both ex vivo isolated cardiomyocytes and in vivo using an AAV9-mediated overexpression mouse model, that CTNNA3 phosphorylation at these residues plays a key role in maintaining protein localization at the cardiomyocyte ICD to regulate conductance and cell-cell adhesion. Collectively, this integrative proteomic/phosphoproteomic approach identifies region- and etiology-associated signaling pathways in human HF and describes a role for CTNNA3 phosphorylation in the pathophysiology of DCM.


Asunto(s)
Cardiomiopatía Dilatada , Insuficiencia Cardíaca , Animales , Ratones , Humanos , Cardiomiopatía Dilatada/metabolismo , Ventrículos Cardíacos/metabolismo , Fosforilación , Proteómica , Miocardio/metabolismo , Insuficiencia Cardíaca/metabolismo , alfa Catenina/metabolismo
2.
Proteomics ; 23(21-22): e2200289, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37691071

RESUMEN

Heart disease remains a leading cause of death in North America and worldwide. Despite advances in therapies, the chronic nature of cardiovascular diseases ultimately results in frequent hospitalizations and steady rates of mortality. Systems biology approaches have provided a new frontier toward unraveling the underlying mechanisms of cell, tissue, and organ dysfunction in disease. Mapping the complex networks of molecular functions across the genome, transcriptome, proteome, and metabolome has enormous potential to advance our understanding of cardiovascular disease, discover new disease biomarkers, and develop novel therapies. Computational workflows to interpret these data-intensive analyses as well as integration between different levels of interrogation remain important challenges in the advancement and application of systems biology-based analyses in cardiovascular research. This review will focus on summarizing the recent developments in network biology-level profiling in the heart, with particular emphasis on modeling of human heart failure. We will provide new perspectives on integration between different levels of large "omics" datasets, including integration of gene regulatory networks, protein-protein interactions, signaling networks, and metabolic networks in the heart.


Asunto(s)
Enfermedades Cardiovasculares , Humanos , Enfermedades Cardiovasculares/genética , Multiómica , Biología de Sistemas , Genoma , Metaboloma , Biología Computacional/métodos
3.
Nat Methods ; 16(8): 737-742, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31308550

RESUMEN

Protein complexes are key macromolecular machines of the cell, but their description remains incomplete. We and others previously reported an experimental strategy for global characterization of native protein assemblies based on chromatographic fractionation of biological extracts coupled to precision mass spectrometry analysis (chromatographic fractionation-mass spectrometry, CF-MS), but the resulting data are challenging to process and interpret. Here, we describe EPIC (elution profile-based inference of complexes), a software toolkit for automated scoring of large-scale CF-MS data to define high-confidence multi-component macromolecules from diverse biological specimens. As a case study, we used EPIC to map the global interactome of Caenorhabditis elegans, defining 612 putative worm protein complexes linked to diverse biological processes. These included novel subunits and assemblies unique to nematodes that we validated using orthogonal methods. The open source EPIC software is freely available as a Jupyter notebook packaged in a Docker container (https://hub.docker.com/r/baderlab/bio-epic/).


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Complejos Multiproteicos/aislamiento & purificación , Complejos Multiproteicos/metabolismo , Mapeo de Interacción de Proteínas , Proteoma/análisis , Programas Informáticos , Animales , Proteínas de Caenorhabditis elegans/aislamiento & purificación
4.
J Mol Cell Cardiol ; 160: 97-110, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34216608

RESUMEN

Angiotensin II (Ang II) presents a critical mediator in various pathological conditions such as non-genetic cardiomyopathy. Osmotic pump infusion in rodents is a commonly used approach to model cardiomyopathy associated with Ang II. However, profound differences in electrophysiology and pharmacokinetics between rodent and human cardiomyocytes may limit predictability of animal-based experiments. This study investigates the application of an Organ-on-a-chip (OOC) system in modeling Ang II-induced progressive cardiomyopathy. The disease model is constructed to recapitulate myocardial response to Ang II in a temporal manner. The long-term tissue cultivation and non-invasive functional readouts enable monitoring of both acute and chronic cardiac responses to Ang II stimulation. Along with mapping of cytokine secretion and proteomic profiles, this model presents an opportunity to quantitatively measure the dynamic pathological changes that could not be otherwise identified in animals. Further, we present this model as a testbed to evaluate compounds that target Ang II-induced cardiac remodeling. Through assessing the effects of losartan, relaxin, and saracatinib, the drug screening data implicated multifaceted cardioprotective effects of relaxin in restoring contractile function and reducing fibrotic remodeling. Overall, this study provides a controllable platform where cardiac activities can be explicitly observed and tested over the pathological process. The facile and high-content screening can facilitate the evaluation of potential drug candidates in the pre-clinical stage.


Asunto(s)
Angiotensina II/efectos adversos , Cardiomiopatías/inducido químicamente , Cardiomiopatías/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Animales , Cardiomiopatías/patología , Cardiotónicos/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Evaluación Preclínica de Medicamentos/métodos , Fibroblastos/metabolismo , Fibrosis , Humanos , Células Madre Pluripotentes Inducidas/citología , Dispositivos Laboratorio en un Chip , Losartán/farmacología , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proyectos Piloto , Proteoma , Proteómica/métodos , Proteínas Recombinantes/farmacología , Relaxina/farmacología , Remodelación Ventricular/efectos de los fármacos
5.
J Proteome Res ; 20(5): 2867-2881, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33789425

RESUMEN

Heart failure (HF) is associated with pathological remodeling of the myocardium, including the initiation of fibrosis and scar formation by activated cardiac fibroblasts (CFs). Although early CF-dependent scar formation helps prevent cardiac rupture by maintaining the heart's structural integrity, ongoing deposition of the extracellular matrix in the remote and infarct regions can reduce tissue compliance, impair cardiac function, and accelerate progression to HF. In our study, we conducted mass spectrometry (MS) analysis to identify differentially altered proteins and signaling pathways between CFs isolated from 7 day sham and infarcted murine hearts. Surprisingly, CFs from both the remote and infarct regions of injured hearts had a wide number of similarly altered proteins and signaling pathways that were consistent with fibrosis and activation into pathological myofibroblasts. Specifically, proteins enriched in CFs isolated from MI hearts were involved in pathways pertaining to cell-cell and cell-matrix adhesion, chaperone-mediated protein folding, and collagen fibril organization. These results, together with principal component analyses, provided evidence of global CF activation postinjury. Interestingly, however, direct comparisons between CFs from the remote and infarct regions of injured hearts identified 15 differentially expressed proteins between MI remote and MI infarct CFs. Eleven of these proteins (Gpc1, Cthrc1, Vmac, Nexn, Znf185, Sprr1a, Specc1, Emb, Limd2, Pawr, and Mcam) were higher in MI infarct CFs, whereas four proteins (Gstt1, Gstm1, Tceal3, and Inmt) were higher in MI remote CFs. Collectively, our study shows that MI injury induced global changes to the CF proteome, with the magnitude of change reflecting their relative proximity to the site of injury.


Asunto(s)
Infarto del Miocardio , Remodelación Ventricular , Animales , Modelos Animales de Enfermedad , Fibroblastos/patología , Fibrosis , Proteínas con Dominio LIM , Ratones , Proteínas de Microfilamentos , Infarto del Miocardio/genética , Miocardio/patología , Miofibroblastos/patología
6.
Am J Physiol Heart Circ Physiol ; 320(1): H417-H423, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33185114

RESUMEN

Cardiovascular diseases remain the most rapidly rising contributing factor of all-cause mortality and the leading cause of inpatient hospitalization worldwide, with costs exceeding $30 billion annually in North America. Cell surface and membrane-associated proteins play an important role in cardiomyocyte biology and are involved in the pathogenesis of many human heart diseases. In cardiomyocytes, membrane proteins serve as critical signaling receptors, Ca2+ cycling regulators, and electrical propagation regulators, all functioning in concert to maintain spontaneous and synchronous contractions of cardiomyocytes. Membrane proteins are excellent pharmaceutical targets due to their uniquely exposed position within the cell. Perturbations in cardiac membrane protein localization and function have been implicated in the progression and pathogenesis of many heart diseases. However, previous attempts at profiling the cardiac membrane proteome have yielded limited results due to poor technological developments for isolating hydrophobic, low-abundance membrane proteins. Comprehensive mapping and characterization of the cardiac membrane proteome thereby remains incomplete. This review will focus on recent advances in mapping the cardiac membrane proteome and the role of novel cardiac membrane proteins in the healthy and the diseased heart.


Asunto(s)
Membrana Celular/metabolismo , Cardiopatías/metabolismo , Proteínas de la Membrana/metabolismo , Miocitos Cardíacos/metabolismo , Proteómica , Animales , Difusión de Innovaciones , Predicción , Cardiopatías/patología , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Miocitos Cardíacos/patología , Proteómica/historia , Proteómica/tendencias
7.
Proc Natl Acad Sci U S A ; 113(44): 12592-12597, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27742792

RESUMEN

Phospholamban (PLN) plays a central role in Ca2+ homeostasis in cardiac myocytes through regulation of the sarco(endo)plasmic reticulum Ca2+-ATPase 2A (SERCA2A) Ca2+ pump. An inherited mutation converting arginine residue 9 in PLN to cysteine (R9C) results in dilated cardiomyopathy (DCM) in humans and transgenic mice, but the downstream signaling defects leading to decompensation and heart failure are poorly understood. Here we used precision mass spectrometry to study the global phosphorylation dynamics of 1,887 cardiac phosphoproteins in early affected heart tissue in a transgenic R9C mouse model of DCM compared with wild-type littermates. Dysregulated phosphorylation sites were quantified after affinity capture and identification of 3,908 phosphopeptides from fractionated whole-heart homogenates. Global statistical enrichment analysis of the differential phosphoprotein patterns revealed selective perturbation of signaling pathways regulating cardiovascular activity in early stages of DCM. Strikingly, dysregulated signaling through the Notch-1 receptor, recently linked to cardiomyogenesis and embryonic cardiac stem cell development and differentiation but never directly implicated in DCM before, was a prominently perturbed pathway. We verified alterations in Notch-1 downstream components in early symptomatic R9C transgenic mouse cardiomyocytes compared with wild type by immunoblot analysis and confocal immunofluorescence microscopy. These data reveal unexpected connections between stress-regulated cell signaling networks, specific protein kinases, and downstream effectors essential for proper cardiac function.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Fosfoproteínas/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Transducción de Señal , Secuencia de Aminoácidos , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Cardiomiopatía Dilatada/genética , Modelos Animales de Enfermedad , Humanos , Ratones Transgénicos , Mutación , Miocardio/metabolismo , Miocardio/patología , Fosfoproteínas/genética , Fosforilación , Proteoma/genética
8.
BMC Biochem ; 17(1): 17, 2016 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-27474398

RESUMEN

BACKGROUND: Human tyrosine-protein phosphatase non-receptor type substrate 1α (SIRPA) is a surface marker identified in cardiomyocytes differentiated from human embryonic stem cells. Our objective was to determine if circulating SIRPA levels can serve as a biomarker of cardiac injury in children undergoing open heart surgery. RESULTS: Paired pre- and post-operative serum samples from 48 pediatric patients undergoing open heart surgery and from 6 pediatric patients undergoing non-cardiac surgery (controls) were tested for SIRPA protein levels using commercially available SIRPA ELISA kits from two manufacturers. Post-operative SIRPA concentrations were significantly higher in patients after cardiac surgery compared to non-cardiac surgery when tested using SIRPA ELISA kits from both manufacturers. To verify the identity of the protein detected, recombinant human SIRPA protein (rhSIRPA) was tested on both ELISA kits. The calibrator from both ELISA kits was analyzed by Western blot as well as by Mass Spectrometry (MS). Western blot analysis of calibrators from both kits did not identity SIRPA. MS analysis of calibrators from both ELISA kits identified several inflammatory markers and albumin but no SIRPA was detected. CONCLUSIONS: We conclude that commercially available ELISA kits for SIRPA give false-positive results. Verifying protein identity using robust protein characterization is critical to avoid false biomarker discovery when using commercial ELISA kits.


Asunto(s)
Antígenos de Diferenciación/sangre , Biomarcadores/sangre , Receptores Inmunológicos/sangre , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Western Blotting , Calibración , Estudios de Casos y Controles , Niño , Ensayo de Inmunoadsorción Enzimática/normas , Lesiones Cardíacas/sangre , Lesiones Cardíacas/cirugía , Humanos , Espectrometría de Masas , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes/análisis , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación
9.
Proteomics ; 15(23-24): 4080-95, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26227301

RESUMEN

Fertilization triggers a dynamic symphony of molecular transformations induced by a rapid rise in intracellular calcium. Most prominent are surface alterations, metabolic activation, cytoskeletal reorganization, and cell-cycle reentry. While the activation process appears to be broadly evolutionarily conserved, and protein phosphorylation is known to play a key role, the signaling networks mediating the response to fertilization are not well described. To address this gap, we performed a time course phosphoproteomic analysis of egg activation in the sea urchin Strongylocentrotus purpuratus, a system that offers biochemical tractability coupled with exquisite synchronicity. By coupling large-scale phosphopeptide enrichment with unbiased quantitative MS, we identified striking changes in global phosphoprotein patterns at 2- and 5-min postfertilization as compared to unfertilized eggs. Overall, we mapped 8796 distinct phosphosite modifications on 2833 phosphoproteins, of which 15% were differentially regulated in early egg activation. Activated kinases were identified by phosphosite mapping, while enrichment analyses revealed conserved signaling cascades not previously associated with egg activation. This work represents the most comprehensive study of signaling associated with egg activation to date, suggesting novel mechanisms that can be experimentally tested and providing a valuable resource for the broader research community. All MS data have been deposited in the ProteomeXchange with identifier PXD002239 (http://proteomecentral.proteomexchange.org/dataset/PXD002239).


Asunto(s)
Proteómica , Erizos de Mar/metabolismo , Strongylocentrotus purpuratus/metabolismo , Animales , Calcio/metabolismo
10.
BMC Med ; 11: 31, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23390961

RESUMEN

Aberrant and dysregulated protein glycosylation is a well-established event in the process of oncogenesis and cancer progression. Years of study on the glycobiology of cancer have been focused on the development of clinically viable diagnostic applications of this knowledge. However, for a number of reasons, there has been only sparse and varied success. The causes of this range from technical to biological issues that arise when studying protein glycosylation and attempting to apply it to practical applications. This review focuses on the pitfalls, advances, and future directions to be taken in the development of clinically applicable quantitative assays using glycan moieties from serum-based proteins as analytes. Topics covered include the development and progress of applications of lectins, mass spectrometry, and other technologies towards this purpose. Slowly but surely, novel applications of established and development of new technologies will eventually provide us with the tools to reach the ultimate goal of quantification of the full scope of heterogeneity associated with the glycosylation of biomarker candidate glycoproteins in a clinically applicable fashion.


Asunto(s)
Biomarcadores de Tumor/sangre , Glicoproteínas/sangre , Neoplasias/sangre , Neoplasias/diagnóstico , Animales , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Glicoproteínas/genética , Glicosilación , Humanos , Espectrometría de Masas/métodos , Espectrometría de Masas/normas , Neoplasias/genética
11.
Clin Chem ; 59(1): 296-305, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23136250

RESUMEN

BACKGROUND: Kallikrein-related peptidase 6 (KLK6), a member of the serine protease family of kallikrein (KLK) genes, is dysregulated in ovarian carcinomas (OCa) and its overexpression is associated with poor prognosis. Regulation of its expression is poorly understood and is likely to be influenced by multiple mechanisms. The KLK locus is subject to copy number changes and heterogeneity in serous OCas. These copy number imbalances generally correlate with KLK6 protein expression; however, this is not always the case. In this study we explored the role of miRNAs in the posttranscriptional control of KLK6 expression and the contributions of copy numbers, not only of the KLK locus, but also of the miRNAs predicted to regulate it. METHODS AND RESULTS: By miRNA profiling of the KLK6-overexpressing OCa cell line, OVCAR-3, we identified overexpressed and underexpressed miRNAs. Publically available miRNA databases identified the human miRNA lethal 7 (hsa-let-7) family members as putative regulating miRNAs, from which hsa-let-7a was chosen for functional analysis. The transient transfection of hsa-let-7a to OVCAR-3 resulted in a decrease of KLK6 secreted protein. Moreover, such transfection was also able to weakly affect the expression of another member of the KLK gene family, KLK10 (kallikrein-related peptidase 10). Cytogenomic analysis, including array comparative genomic hybridization, fluorescence in situ hybridization, and spectral karyotyping revealed the overall net copy number losses of hsa-let-7a and other miRNAs predicted to target KLK6. CONCLUSIONS: The hsa-let-7 family member hsa-let-7a is a modulator of KLK6 protein expression that is independent of the KLK6 copy number status.


Asunto(s)
Dosificación de Gen , Calicreínas/metabolismo , MicroARNs/genética , Neoplasias Ováricas/genética , Western Blotting , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Calicreínas/genética , Neoplasias Ováricas/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Clin Proteomics ; 10(1): 5, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23635041

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is the most common type of dementia affecting people over 65 years of age. The hallmarks of AD are the extracellular deposits known as amyloid ß plaques and the intracellular neurofibrillary tangles, both of which are the principal players involved in synaptic loss and neuronal cell death. Tau protein and Aß fragment 1-42 have been investigated so far in cerebrospinal fluid as a potential AD biomarkers. However, an urgent need to identify novel biomarkers which will capture disease in the early stages and with better specificity remains. High-throughput proteomic and pathway analysis of hippocampal tissue provides a valuable source of disease-related proteins and biomarker candidates, since it represents one of the earliest affected brain regions in AD. RESULTS: In this study 2954 proteins were identified (with at least 2 peptides for 1203 proteins) from both control and AD brain tissues. Overall, 204 proteins were exclusively detected in AD and 600 proteins in control samples. Comparing AD and control exclusive proteins with cerebrospinal fluid (CSF) literature-based proteome, 40 out of 204 AD related proteins and 106 out of 600 control related proteins were also present in CSF. As most of these proteins were extracellular/secretory origin, we consider them as a potential source of candidate biomarkers that need to be further studied and verified in CSF samples. CONCLUSIONS: Our semiquantitative proteomic analysis provides one of the largest human hippocampal proteome databases. The lists of AD and control related proteins represent a panel of proteins potentially involved in AD pathogenesis and could also serve as prospective AD diagnostic biomarkers.

13.
Clin Chem Lab Med ; 51(7): 1467-76, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23241603

RESUMEN

BACKGROUND: Ovarian cancer is the leading cause of death among all gynecological disorders. Aberrant glycosylation, or more specifically, increased sialylation of proteins has been observed in ovarian cancer. Several sialyltransferase genes have been shown to be up-regulated at both the mRNA and protein levels in a number of cancers, including that of the ovary. ST6GAL1 (ß-galactosamide α2,6-sialyltranferase 1) gene expression has previously been shown to be upregulated in ovarian cancers of all major subtypes. METHODS: We have identified the sialome (i.e., sialic acid containing glycoproteins) of biological fluids from ovarian cancer patients and ovarian cancer cell lines utilizing tandem mass spectrometry as a potential pool of novel biomarker candidates. The sialoglycopeptides from four ovarian cancer cell lines, pooled ascites (n=13) and ovarian cyst (n=14) fluids from ovarian cancer patients were enriched utilizing affinity to agarose-immobilized Elderberry lectin (Sambucus nigra agglutinin) and magnetic hydrazide beads folowing periodate-mediated oxidation of sialic acids. Benign ovarian cyst (n=10) and peritoneal effusion (n=20) fluids were analyzed in the same fashion to serve as controls. PNGase F deglycosylated peptides were identified using electrospray ionization-LTQ Orbitrap tandem mass spectrometry. RESULTS: In all of the samples analyzed in the glycoproteomic portion of the study, we have identified 579 glycosylation sites on 333 proteins. Of these, 13 were exclusively identified in biological fluids from ovarian cancer patients, and another eight were common to these fluids and the ovarian cancer cell line supernatants. CONCLUSIONS: The proteins identified in the present study could form the basis for future studies examining and quantifying their sialylation status as biomarkers of ovarian cancer.


Asunto(s)
Adenocarcinoma de Células Claras/diagnóstico , Adenocarcinoma Mucinoso/diagnóstico , Biomarcadores de Tumor/aislamiento & purificación , Cistadenocarcinoma Seroso/diagnóstico , Glicoproteínas/aislamiento & purificación , Neoplasias Ováricas/diagnóstico , Sialiltransferasas/aislamiento & purificación , Adenocarcinoma de Células Claras/genética , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma Mucinoso/genética , Adenocarcinoma Mucinoso/metabolismo , Adulto , Secuencia de Aminoácidos , Biomarcadores de Tumor/genética , Estudios de Casos y Controles , Línea Celular Tumoral , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Femenino , Glicoproteínas/genética , Glicosilación , Humanos , Lectinas/química , Persona de Mediana Edad , Anotación de Secuencia Molecular , Datos de Secuencia Molecular , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Proteómica , Sialiltransferasas/genética , Espectrometría de Masas en Tándem
14.
J Clin Invest ; 133(9)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37115698

RESUMEN

Inflammation promotes adverse ventricular remodeling, a common antecedent of heart failure. Here, we set out to determine how inflammatory cells affect cardiomyocytes in the remodeling heart. Pathogenic cardiac macrophages induced an IFN response in cardiomyocytes, characterized by upregulation of the ubiquitin-like protein IFN-stimulated gene 15 (ISG15), which posttranslationally modifies its targets through a process termed ISGylation. Cardiac ISG15 is controlled by type I IFN signaling, and ISG15 or ISGylation is upregulated in mice with transverse aortic constriction or infused with angiotensin II; rats with uninephrectomy and DOCA-salt, or pulmonary artery banding; cardiomyocytes exposed to IFNs or CD4+ T cell-conditioned medium; and ventricular tissue of humans with nonischemic cardiomyopathy. By nanoscale liquid chromatography-tandem mass spectrometry, we identified the myofibrillar protein filamin-C as an ISGylation target. ISG15 deficiency preserved cardiac function in mice with transverse aortic constriction and led to improved recovery of mouse hearts ex vivo. Metabolomics revealed that ISG15 regulates cardiac amino acid metabolism, whereas ISG15 deficiency prevented misfolded filamin-C accumulation and induced cardiomyocyte autophagy. In sum, ISG15 upregulation is a feature of pathological ventricular remodeling, and protein ISGylation is an inflammation-induced posttranslational modification that may contribute to heart failure development by altering cardiomyocyte protein turnover.


Asunto(s)
Citocinas , Insuficiencia Cardíaca , Humanos , Ratas , Ratones , Animales , Citocinas/genética , Citocinas/metabolismo , Filaminas , Remodelación Ventricular/genética , Insuficiencia Cardíaca/metabolismo , Inflamación , Ubiquitinas/genética
15.
Proteomics ; 12(6): 799-809, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22539431

RESUMEN

Kallikrein 6 (KLK6) has been shown to be aberrantly glycosylated in ovarian cancer. Here, we report a novel HPLC anion exchange method, coupled to a KLK6-specific ELISA, capable of differentiating KLK6 glycoform subgroups in biological fluids. Biological fluids were fractionated using anion exchange and resulting fractions were analyzed for KLK6 content by ELISA producing a four-peak elution profile. Using this assay, the KLK6 elution profile and distribution across peaks of a set (n = 7) of ovarian cancer patient matched serum and ascites fluid samples was found to be different than the profile of serum and cerebrospinal fluid (CSF) of normal individuals (n = 7). Glycosylation patterns of recombinant KLK6 (rKLK6) were characterized using tandem mass spectrometry (MS/MS), and found to consist of a highly heterogeneous KLK6 population. This protein was found to contain all of the four diagnostic KLK6 peaks present in the previously assayed biological fluids. The rKLK6 glycoform composition of each peak was assessed by lectin affinity and MS/MS based glycopeptide quantification by product ion monitoring. The combined results showed an increase in terminal alpha 2-6 linked sialic acid in the N-glycans found on KLK6 from ovarian cancer serum and ascites, as opposed to CSF and serum of normal individuals.


Asunto(s)
Cromatografía por Intercambio Iónico , Calicreínas/sangre , Calicreínas/líquido cefalorraquídeo , Neoplasias Ováricas/sangre , Neoplasias Ováricas/líquido cefalorraquídeo , Ensayo de Inmunoadsorción Enzimática , Femenino , Glicopéptidos/análisis , Glicosilación , Células HEK293 , Humanos , Calicreínas/análisis , Proteínas Recombinantes/análisis , Espectrometría de Masas en Tándem
16.
Commun Biol ; 5(1): 955, 2022 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-36097051

RESUMEN

Functional oncogenic links between ErbB2 and ERRα in HER2+ breast cancer patients support a therapeutic benefit of co-targeted therapies. However, ErbB2 and ERRα also play key roles in heart physiology, and this approach could pose a potential liability to cardiovascular health. Herein, using integrated phosphoproteomic, transcriptomic and metabolic profiling, we uncovered molecular mechanisms associated with the adverse remodeling of cardiac functions in mice with combined attenuation of ErbB2 and ERRα activity. Genetic disruption of both effectors results in profound effects on cardiomyocyte architecture, inflammatory response and metabolism, the latter leading to a decrease in fatty acyl-carnitine species further increasing the reliance on glucose as a metabolic fuel, a hallmark of failing hearts. Furthermore, integrated omics signatures of ERRα loss-of-function and doxorubicin treatment exhibit common features of chemotherapeutic cardiotoxicity. These findings thus reveal potential cardiovascular risks in discrete combination therapies in the treatment of breast and other cancers.


Asunto(s)
Receptores de Estrógenos , Remodelación Ventricular , Animales , Doxorrubicina/farmacología , Ratones , Miocitos Cardíacos/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptor Relacionado con Estrógeno ERRalfa
17.
Mol Cell Proteomics ; 8(4): 791-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19088065

RESUMEN

Ovarian cancer causes more deaths than any other gynecological disorder. Perturbed glycosylation is one of the hallmarks of this malignancy. Kallikrein 6 (KLK6) elevation in serum is a diagnostic and prognostic indicator in ovarian cancer. The majority of ovarian carcinomas express high levels of KLK6, which diffuses into the circulation. Under physiological conditions, KLK6 is expressed highly in the central nervous system and found at high levels in cerebrospinal fluid from where it enters the circulation. Our aim was to characterize and compare the N-glycosylation status of this protein in ovarian cancer ascites fluid and cerebrospinal fluid. Anion-exchange chromatography was used to reveal different post-translational modifications on the two isoforms. Mobility gel shift Western blot analysis coupled with glycosidase digestion showed that the molecular weight difference between the two isoforms was because of differential glycosylation patterns. The presence of a single N-glycosylation site on KLK6 was confirmed by site-directed mutagenesis. Using a Sambucus nigra agglutinin-monoclonal antibody sandwich enzyme-linked immunosorbent assay approach, it was shown that ovarian cancer-derived KLK6 was modified with alpha2-6-linked sialic acid. The structure and composition of glycans of both KLK6 isoforms was elucidated by glycopeptide monitoring with electrospray ionization-Orbitrap tandem mass spectrometry. Therefore, the extensive and almost exclusive sialylation of KLK6 from ovarian cancer cells could lead to the development of an improved biomarker for the early diagnosis of ovarian carcinoma.


Asunto(s)
Sistema Nervioso Central/enzimología , Calicreínas/metabolismo , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Secuencia de Aminoácidos , Aniones , Anticuerpos , Western Blotting , Líquidos Corporales/enzimología , Cromatografía por Intercambio Iónico , Ensayo de Inmunoadsorción Enzimática , Femenino , Glicopéptidos/química , Glicósido Hidrolasas/metabolismo , Glicosilación , Humanos , Calicreínas/química , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Lectinas de Plantas/metabolismo , Proteínas Inactivadoras de Ribosomas/metabolismo , Espectrometría de Masas en Tándem
18.
Nat Biomed Eng ; 4(9): 889-900, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32661320

RESUMEN

Study of the molecular basis of myocardial fibrosis is hampered by limited access to tissues from human patients and by confounding variables associated with sample accessibility, collection, processing and storage. Here, we report an integrative strategy based on mass spectrometry for the phosphoproteomic profiling of normal and fibrotic cardiac tissue obtained from surgical explants from patients with hypertrophic cardiomyopathy, from a transaortic-constriction mouse model of cardiac hypertrophy and fibrosis, and from a heart-on-a-chip model of cardiac fibrosis. We used the integrative approach to map the relative abundance of thousands of proteins, phosphoproteins and phosphorylation sites specific to each tissue source, to identify key signalling pathways driving fibrosis and to screen for anti-fibrotic compounds targeting glycogen synthase kinase 3, which has a consistent role as a key mediator of fibrosis in all three types of tissue specimen. The integrative disease-modelling strategy may reveal new insights into mechanisms of cardiac disease and serve as a test bed for drug screening.


Asunto(s)
Miocardio/patología , Proteómica/métodos , Transducción de Señal , Animales , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Fibrosis , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Ratones , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteoma/metabolismo , Ingeniería de Tejidos
19.
Sci Data ; 7(1): 425, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33262348

RESUMEN

In the current study we examined several proteomic- and RNA-Seq-based datasets of cardiac-enriched, cell-surface and membrane-associated proteins in human fetal and mouse neonatal ventricular cardiomyocytes. By integrating available microarray and tissue expression profiles with MGI phenotypic analysis, we identified 173 membrane-associated proteins that are cardiac-enriched, conserved amongst eukaryotic species, and have not yet been linked to a 'cardiac' Phenotype-Ontology. To highlight the utility of this dataset, we selected several proteins to investigate more carefully, including FAM162A, MCT1, and COX20, to show cardiac enrichment, subcellular distribution and expression patterns in disease. We performed three-dimensional confocal imaging analysis to validate subcellular localization and expression in adult mouse ventricular cardiomyocytes. FAM162A, MCT1, and COX20 were expressed differentially at the transcriptomic and proteomic levels in multiple models of mouse and human heart diseases and may represent potential diagnostic and therapeutic targets for human dilated and ischemic cardiomyopathies. Altogether, we believe this comprehensive cardiomyocyte membrane proteome dataset will prove instrumental to future investigations aimed at characterizing heart disease markers and/or therapeutic targets for heart failure.


Asunto(s)
Proteínas de la Membrana/análisis , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Proteoma , Animales , Biología Computacional , Conjuntos de Datos como Asunto , Ratones , RNA-Seq , Transcriptoma
20.
Physiol Rep ; 7(14): e14144, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31325238

RESUMEN

Phospholamban (PLN) is an important Ca2+ modulator at the sarcoplasmic reticulum (SR) of striated muscles. It physically interacts and inhibits sarcoplasmic reticulum Ca2+ ATPase (SERCA2) function, whereas a protein kinase A (PKA)-dependent phosphorylation at its serine 16 reverses the inhibition. The underlying mechanism of this post-translational modification, however, remains not fully understood. Using publicly available databases, we identified A-kinase anchoring protein 6 (AKAP6) as a candidate that might play some roles in PLN phosphorylation. Immunofluorescence showed colocalization between GFP-AKAP6 and PLN in transfected HEK-293T cells and cultured mouse neonatal cardiomyocytes (CMNCs). Co-immunoprecipitation confirmed the functional interaction between AKAP6 and PLN in HEK-293T and isolated adult rat cardiomyocytes in response to isoproterenol stimulation. Functionally, AKAP6 promoted Ca2+ uptake activity of SERCA1 in cotransfected HEK-293T cells despite the presence of PLN. These results were further confirmed in adult rat cardiomyocytes. Immunofluorescence showed colocalization of both proteins around the perinuclear region, while protein-protein interaction was corroborated by immunoprecipitation of the nucleus-enriched fraction of rat hearts. Our findings suggest AKAP6 as a novel interacting partner to PLN in HEK-293T and murine cardiomyocytes.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas de Unión al Calcio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Señalización del Calcio , Células Cultivadas , Células HEK293 , Humanos , Ratones , Unión Proteica , Ratas , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA