Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Proteome Res ; 20(6): 3134-3149, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34014671

RESUMEN

Multiple myeloma is an incurable hematological malignancy that impacts tens of thousands of people every year in the United States. Treatment for eligible patients involves induction, consolidation with stem cell rescue, and maintenance. High-dose therapy with a DNA alkylating agent, melphalan, remains the primary drug for consolidation therapy in conjunction with autologous stem-cell transplantation; as such, melphalan resistance remains a relevant clinical challenge. Here, we describe a proteometabolomic approach to examine mechanisms of acquired melphalan resistance in two cell line models. Drug metabolism, steady-state metabolomics, activity-based protein profiling (ABPP, data available at PRIDE: PXD019725), acute-treatment metabolomics, and western blot analyses have allowed us to further elucidate metabolic processes associated with melphalan resistance. Proteometabolomic data indicate that drug-resistant cells have higher levels of pentose phosphate pathway metabolites. Purine, pyrimidine, and glutathione metabolisms were commonly altered, and cell-line-specific changes in metabolite levels were observed, which could be linked to the differences in steady-state metabolism of naïve cells. Inhibition of selected enzymes in purine synthesis and pentose phosphate pathways was evaluated to determine their potential to improve melphalan's efficacy. The clinical relevance of these proteometabolomic leads was confirmed by comparison of tumor cell transcriptomes from newly diagnosed MM patients and patients with relapsed disease after treatment with high-dose melphalan and autologous stem-cell transplantation. The observation of common and cell-line-specific changes in metabolite levels suggests that omic approaches will be needed to fully examine melphalan resistance in patient specimens and define personalized strategies to optimize the use of high-dose melphalan.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Mieloma Múltiple , Humanos , Melfalán/farmacología , Metabolómica , Mieloma Múltiple/tratamiento farmacológico , Trasplante Autólogo
2.
Electrophoresis ; 33(24): 3786-97, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23161537

RESUMEN

MS-based proteomics has been the method of choice for biomarker discovery in the field of traumatic brain injury (TBI). Due to its high sensitivity and specificity, MS is now being explored for biomarker quantitative validation in tissue and biofluids. In this study, we demonstrate the use of MS in both qualitative protein identification and targeted detection of acute TBI biomarkers released from degenerating cultured rat cortical mixed neuronal cells, mimicking intracellular fluid in the central nervous system after TBI. Calpain activation was induced by cell treatment with maitotoxin (MTX), a known calcium channel opener. Separate plates of mixed neuronal-glial culture were subjected to excitotoxin N-methyl-D-aspartate (NMDA) and apoptotic inducer staurosporine. Acute TBI biomarkers, GFAP and UCH-L1, were first detected and assessed in the culture media by Western blot. The cell-conditioned media were then trypsinized and subjected to bottom up proteomic analysis. GFAP was readily detected by data-dependent scanning but not UCH-L1. As a proof-of-principle study, rat glia-enriched cell cultures treated with MTX were used to investigate the time-dependent release of GFAP breakdown product by Western blot and for isotope dilution MS absolute quantitation method development. Absolute quantitation of the GFAP release was conducted using the three cortical mixed neuronal cell cultures treated with different agents. Other differentially expressed proteins identified in the glial-enriched and cortical mixed neuronal cell culture models were further analyzed by bioinformatic tools. In summary, this study demonstrates the use of MS in both protein identification and targeted quantitation of acute TBI biomarkers and is the preliminary step toward development of TBI biomarker validation by targeted MS.


Asunto(s)
Lesiones Encefálicas/metabolismo , Corteza Cerebral/metabolismo , Espectrometría de Masas/métodos , Neuroglía/metabolismo , Neuronas/metabolismo , Proteómica/métodos , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/análisis , Biomarcadores/metabolismo , Lesiones Encefálicas/patología , Células Cultivadas , Corteza Cerebral/química , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/análisis , Proteína Ácida Fibrilar de la Glía/química , Proteína Ácida Fibrilar de la Glía/metabolismo , Toxinas Marinas/farmacología , N-Metilaspartato/farmacología , Necrosis/metabolismo , Neuroglía/química , Neuroglía/citología , Neuronas/química , Neuronas/citología , Oxocinas/farmacología , Mapas de Interacción de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Estaurosporina/farmacología , Ubiquitina Tiolesterasa/análisis , Ubiquitina Tiolesterasa/metabolismo
3.
Hepatol Commun ; 6(3): 513-525, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34811964

RESUMEN

Alcoholic fatty liver disease (AFLD) is characterized by lipid accumulation and inflammation and can progress to cirrhosis and cancer in the liver. AFLD diagnosis currently relies on histological analysis of liver biopsies. Early detection permits interventions that would prevent progression to cirrhosis or later stages of the disease. Herein, we have conducted the first comprehensive time-course study of lipids using novel state-of-the art lipidomics methods in plasma and liver in the early stages of a mouse model of AFLD, i.e., Lieber-DeCarli diet model. In ethanol-treated mice, changes in liver tissue included up-regulation of triglycerides (TGs) and oxidized TGs and down-regulation of phosphatidylcholine, lysophosphatidylcholine, and 20-22-carbon-containing lipid-mediator precursors. An increase in oxidized TGs preceded histological signs of early AFLD, i.e., steatosis, with these changes observed in both the liver and plasma. The major lipid classes dysregulated by ethanol play important roles in hepatic inflammation, steatosis, and oxidative damage. Conclusion: Alcohol consumption alters the liver lipidome before overt histological markers of early AFLD. This introduces the exciting possibility that specific lipids may serve as earlier biomarkers of AFLD than those currently being used.


Asunto(s)
Hígado Graso Alcohólico , Hígado Graso , Hepatopatías Alcohólicas , Animales , Biomarcadores/metabolismo , Etanol/efectos adversos , Hígado Graso Alcohólico/diagnóstico , Inflamación , Lipidómica , Cirrosis Hepática , Hepatopatías Alcohólicas/diagnóstico , Ratones , Oxidación-Reducción , Triglicéridos
4.
Electrophoresis ; 32(13): 1692-705, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21706495

RESUMEN

Protein tyrosine nitration is a post-translational modification commonly used as a marker of cellular oxidative stress associated with numerous pathophysiological conditions. We focused on ubiquitin carboxyl terminal hydrolase-L1 (UCH-L1) and glyceraldehyde-3-phosphate (GAPDH) which are high-abundant brain proteins that have been identified to be highly susceptible to oxidative modification. Both UCH-L1 and GAPDH have been linked to the pathogenesis of Alzheimer's and Parkinson's disease, however specific nitration sites have not been elucidated. Identification of specific nitration sites and quantitation of endogenous nitrated proteins are important in correlating this modification to disease pathology. In this study, purified UCH-L1 and GAPDH were nitrated in vitro with peroxynitrite and the presence of nitrated proteins was confirmed by anti-3-nitrotyrosine Western blots. Data-dependent LC-MS/MS analysis identified several distinct tyrosine nitration sites in UCH-L1 (Tyr-80) and GAPDH (Tyr-47, Tyr-92, and Tyr-312). Subsequent validation with synthetic peptides was conducted for selected nitropeptides. An LC-MS/MS method was developed for semi-quantitative determination of the synthetic nitropeptides: KGQEVSPKVY(*) (UCH-L1) and mFQY(*) DSTHGKF (GAPDH). The nitropeptides were detectable in the mid-attomole range and the peak area response was linear over three orders of magnitude. Targeted analysis of endogenous UCH-L1 and GAPDH nitration was then conducted in an in vivo second-hand smoke rat model to evaluate the utility of this approach.


Asunto(s)
Gliceraldehído-3-Fosfato Deshidrogenasas/química , Estrés Oxidativo/fisiología , Contaminación por Humo de Tabaco , Tirosina/análogos & derivados , Ubiquitina Tiolesterasa/química , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Química Encefálica , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Humanos , Immunoblotting , Masculino , Datos de Secuencia Molecular , Nitrosación , Oxidación-Reducción , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Conejos , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Tirosina/análisis , Tirosina/química , Tirosina/metabolismo , Ubiquitina Tiolesterasa/metabolismo
5.
Methods Mol Biol ; 1996: 273-296, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31127562

RESUMEN

Drug resistance remains a critical problem for the treatment of multiple myeloma (MM), which can serve as a specific example for a highly prevalent unmet medical need across almost all cancer types. In MM, the therapeutic arsenal has expanded and diversified, yet we still lack in-depth molecular understanding of drug mechanisms of action and cellular pathways to therapeutic escape. For those reasons, preclinical models of drug resistance are developed and characterized using different approaches to gain insights into tumor biology and elucidate mechanisms of drug resistance. For MM, numerous drugs are used for treatment, including conventional chemotherapies (e.g., melphalan or L-phenylalanine nitrogen mustard), proteasome inhibitors (e.g., Bortezomib), and immunomodulators (e.g., Lenalidomide). These agents have diverse effects on the myeloma cells, and several mechanisms of drug resistance have been previously described. The disparity of these mechanisms and the complexity of these biological processes lead to the formation of complicated hypotheses that require omics approaches for efficient and effective analysis of model systems that can then be interpreted for patient benefit. Here, we describe the combination of metabolomics and proteomics to assess melphalan resistance in MM by examining three specific areas: drug metabolism, modulation of endogenous metabolites to assist in therapeutic escape, and changes in protein activity gauged by ATP probe uptake.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Melfalán/farmacología , Metabolómica/métodos , Mieloma Múltiple/tratamiento farmacológico , Proteómica/métodos , Antineoplásicos Alquilantes/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión/métodos , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Melfalán/uso terapéutico , Metaboloma/efectos de los fármacos , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Espectrometría de Masas en Tándem/métodos
6.
Front Neurol ; 7: 198, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27920753

RESUMEN

Traumatic brain injury (TBI) represents a critical health problem of which diagnosis, management, and treatment remain challenging. TBI is a contributing factor in approximately one-third of all injury-related deaths in the United States. The Centers for Disease Control and Prevention estimate that 1.7 million people suffer a TBI in the United States annually. Efforts continue to focus on elucidating the complex molecular mechanisms underlying TBI pathophysiology and defining sensitive and specific biomarkers that can aid in improving patient management and care. Recently, the area of neuroproteomics-systems biology is proving to be a prominent tool in biomarker discovery for central nervous system injury and other neurological diseases. In this work, we employed the controlled cortical impact (CCI) model of experimental TBI in rat model to assess the temporal-global proteome changes after acute (1 day) and for the first time, subacute (7 days), post-injury time frame using the established cation-anion exchange chromatography-1D SDS gel electrophoresis LC-MS/MS platform for protein separation combined with discrete systems biology analyses to identify temporal biomarker changes related to this rat TBI model. Rather than focusing on any one individual molecular entity, we used in silico systems biology approach to understand the global dynamics that govern proteins that are differentially altered post-injury. In addition, gene ontology analysis of the proteomic data was conducted in order to categorize the proteins by molecular function, biological process, and cellular localization. Results show alterations in several proteins related to inflammatory responses and oxidative stress in both acute (1 day) and subacute (7 days) periods post-TBI. Moreover, results suggest a differential upregulation of neuroprotective proteins at 7 days post-CCI involved in cellular functions such as neurite growth, regeneration, and axonal guidance. Our study is among the first to assess temporal neuroproteome changes in the CCI model. Data presented here unveil potential neural biomarkers and therapeutic targets that could be used for diagnosis, for treatment and, most importantly, for temporal prognostic assessment following brain injury. Of interest, this work relies on in silico bioinformatics approach to draw its conclusion; further work is conducted for functional studies to validate and confirm the omics data obtained.

7.
Int J Bioinform Res Appl ; 10(1): 27-42, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24449691

RESUMEN

Mass spectrometry (MS) has become the method of choice to study the proteome of brain injury. The high throughput nature of MS-based proteomic experiments generates massive amount of mass spectral data presenting great challenges in downstream interpretation. Currently, different bioinformatics platforms are available for functional analysis and data mining of MS-generated proteomic data. These tools provide a way to convert data sets to biologically interpretable results and functional outcomes. In this review, a brief overview of the currently available bioinformatics strategies applied to neuroproteomic studies is presented. Application of commercially available bioinformatics software to different brain injury studies demonstrates integration of the data mining and analysis applications into neuroproteomic workflows that can identify major protein markers as well as highlight the biological processes and molecular functions involved.


Asunto(s)
Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Biología Computacional/métodos , Mapeo Peptídico/métodos , Proteoma/metabolismo , Proteómica/métodos , Programas Informáticos , Algoritmos , Animales , Humanos
8.
OMICS ; 18(2): 111-31, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24410486

RESUMEN

The post-genomics era has brought about new Omics biotechnologies, such as proteomics and metabolomics, as well as their novel applications to personal genomics and the quantified self. These advances are now also catalyzing other and newer post-genomics innovations, leading to convergences between Omics and nanotechnology. In this work, we systematically contextualize and exemplify an emerging strand of post-genomics life sciences, namely, nanoproteomics and its applications in health and integrative biological systems. Nanotechnology has been utilized as a complementary component to revolutionize proteomics through different kinds of nanotechnology applications, including nanoporous structures, functionalized nanoparticles, quantum dots, and polymeric nanostructures. Those applications, though still in their infancy, have led to several highly sensitive diagnostics and new methods of drug delivery and targeted therapy for clinical use. The present article differs from previous analyses of nanoproteomics in that it offers an in-depth and comparative evaluation of the attendant biotechnology portfolio and their applications as seen through the lens of post-genomics life sciences and biomedicine. These include: (1) immunosensors for inflammatory, pathogenic, and autoimmune markers for infectious and autoimmune diseases, (2) amplified immunoassays for detection of cancer biomarkers, and (3) methods for targeted therapy and automatically adjusted drug delivery such as in experimental stroke and brain injury studies. As nanoproteomics becomes available both to the clinician at the bedside and the citizens who are increasingly interested in access to novel post-genomics diagnostics through initiatives such as the quantified self, we anticipate further breakthroughs in personalized and targeted medicine.


Asunto(s)
Nanotecnología/métodos , Medicina de Precisión/métodos , Proteómica/métodos , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/terapia , Disciplinas de las Ciencias Biológicas , Técnicas Biosensibles , Enfermedades Transmisibles/diagnóstico , Enfermedades Transmisibles/terapia , Humanos , Inmunoensayo , Terapia Molecular Dirigida , Nanoestructuras/uso terapéutico , Nanotecnología/instrumentación , Nanotecnología/tendencias , Neoplasias/diagnóstico , Neoplasias/terapia , Medicina de Precisión/instrumentación , Proteómica/instrumentación
9.
PM R ; 3(6 Suppl 1): S139-47, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21703572

RESUMEN

Although traumatic brain injury (TBI) remains a major health problem, with approximately 2 million incidents occurring annually in the United States, no therapeutic agents to treat TBI have been approved by the Food and Drug Administration despite several clinical trials. It is estimated that 3.5 million Americans now have a lifelong condition that might be termed "chronic traumatic brain injury disease.'' Some health care providers categorize TBI as an "event" for which patients require brief periods of rehabilitation with no further treatment. On the contrary, TBI should be seen as a chronic disease process that fits the World Health Organization definition as being a non-reversible pathologic condition requiring special rehabilitation training. Among the major obstacles that contribute to this type of misconception is the absence of brain injury-specific diagnostic biomarker(s) that can indicate and monitor the long-term health status of patients with TBI after use of conventional therapeutics and a rehabilitation process. It is of interest that recent advances in genomics, proteomics, and systems biology have enabled us to use these high throughput-based approaches in developing biomarkers and therapeutic targets in the area of TBI. One aim of this article is to provide an overview that evaluates the current status of TBI biomarker discovery using neuroproteomics/systems biology techniques, along with their clinical utilization. In addition, we discuss the need for strengthening the role of biomarker-based neuroproteomics/systems biology and its potential utility in the field of rehabilitation, which would lead to the establishment of rehabilomics studies, where biomarkers would indicate and predict the long-term efficacy and health status of patients with chronic TBI conditions.


Asunto(s)
Productos Biológicos/uso terapéutico , Biomarcadores/metabolismo , Investigación Biomédica , Lesiones Encefálicas/rehabilitación , Genómica/métodos , Proteómica/métodos , Biología de Sistemas/métodos , Lesiones Encefálicas/metabolismo , Humanos , Estados Unidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA