Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Biomedicines ; 12(2)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38398037

ABSTRACT

Proteolysis of the extracellular matrix (ECM) by matrix metalloproteinases (MMPs) plays a crucial role in the immune response to bacterial infections. Here we report the secretion of MMPs associated with proteolytic extracellular vesicles (EVs) released by macrophages in response to Salmonella enterica serovar Typhimurium infection. Specifically, we used global proteomics, in vitro, and in vivo approaches to investigate the composition and function of these proteolytic EVs. Using a model of S. Typhimurium infection in murine macrophages, we isolated and characterized a population of small EVs. Bulk proteomics analysis revealed significant changes in protein cargo of naïve and S. Typhimurium-infected macrophage-derived EVs, including the upregulation of MMP-9. The increased levels of MMP-9 observed in immune cells exposed to S. Typhimurium were found to be regulated by the toll-like receptor 4 (TLR-4)-mediated response to bacterial lipopolysaccharide. Macrophage-derived EV-associated MMP-9 enhanced the macrophage invasion through Matrigel as selective inhibition of MMP-9 reduced macrophage invasion. Systemic administration of fluorescently labeled EVs into immunocompromised mice demonstrated that EV-associated MMP activity facilitated increased accumulation of EVs in spleen and liver tissues. This study suggests that macrophages secrete proteolytic EVs to enhance invasion and ECM remodeling during bacterial infections, shedding light on an essential aspect of the immune response.

3.
Nat Methods ; 18(9): 1068-1074, 2021 09.
Article in English | MEDLINE | ID: mdl-34480152

ABSTRACT

In general, mRNAs are assumed to be loaded with ribosomes instantly upon entry into the cytoplasm. To measure ribosome density (RD) on nascent mRNA, we developed nascent Ribo-Seq by combining Ribo-Seq with progressive 4-thiouridine labeling. In mouse macrophages, we determined experimentally the lag between the appearance of nascent mRNA and its association with ribosomes, which was calculated to be 20-22 min for bulk mRNA. In mouse embryonic stem cells, nRibo-Seq revealed an even stronger lag of 35-38 min in ribosome loading. After stimulation of macrophages with lipopolysaccharide, the lag between cytoplasmic and translated mRNA leads to uncoupling between input and ribosome-protected fragments, which gives rise to distorted RD measurements under conditions where mRNA amounts are far from steady-state expression. As a result, we demonstrate that transcriptional changes affect RD in a passive way.


Subject(s)
Protein Biosynthesis , Ribosomes/genetics , Ribosomes/metabolism , Sequence Analysis, RNA/methods , Animals , Cytoplasm/genetics , Kinetics , Lipopolysaccharides/pharmacology , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/physiology , RAW 264.7 Cells , RNA, Messenger/genetics , Ribosomal Proteins/biosynthesis , Ribosomal Proteins/genetics , Ribosomes/drug effects , Time Factors
4.
Matrix Biol ; 96: 47-68, 2021 02.
Article in English | MEDLINE | ID: mdl-33246101

ABSTRACT

Identification of early processes leading to complex tissue pathologies, such as inflammatory bowel diseases, poses a major scientific and clinical challenge that is imperative for improved diagnosis and treatment. Most studies of inflammation onset focus on cellular processes and signaling molecules, while overlooking the environment in which they take place, the continuously remodeled extracellular matrix. In this study, we used colitis models for investigating extracellular-matrix dynamics during disease onset, while treating the matrix as a complete and defined entity. Through the analysis of matrix structure, stiffness and composition, we unexpectedly revealed that even prior to the first clinical symptoms, the colon displays its own unique extracellular-matrix signature and found specific markers of clinical potential, which were also validated in human subjects. We also show that the emergence of this pre-symptomatic matrix is mediated by subclinical infiltration of immune cells bearing remodeling enzymes. Remarkably, whether the inflammation is chronic or acute, its matrix signature converges at pre-symptomatic states. We suggest that the existence of a pre-symptomatic extracellular-matrix is general and relevant to a wide range of diseases.


Subject(s)
Biomarkers/metabolism , Colitis, Ulcerative/pathology , Extracellular Matrix/pathology , Interleukin-10/genetics , Animals , Case-Control Studies , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Female , Gene Knockdown Techniques , Humans , Machine Learning , Male , Mice , Piroxicam/adverse effects , Prognosis , Proteomics
5.
Mol Syst Biol ; 16(9): e9443, 2020 09.
Article in English | MEDLINE | ID: mdl-32960509

ABSTRACT

Tumor relapse as a consequence of chemotherapy resistance is a major clinical challenge in advanced stage breast tumors. To identify processes associated with poor clinical outcome, we took a mass spectrometry-based proteomic approach and analyzed a breast cancer cohort of 113 formalin-fixed paraffin-embedded samples. Proteomic profiling of matched tumors before and after chemotherapy, and tumor-adjacent normal tissue, all from the same patients, allowed us to define eight patterns of protein level changes, two of which correlate to better chemotherapy response. Supervised analysis identified two proteins of proline biosynthesis pathway, PYCR1 and ALDH18A1, that were significantly associated with resistance to treatment based on pattern dominance. Weighted gene correlation network analysis of post-treatment samples revealed that these proteins are associated with tumor relapse and affect patient survival. Functional analysis showed that knockdown of PYCR1 reduced invasion and migration capabilities of breast cancer cell lines. PYCR1 knockout significantly reduced tumor burden and increased drug sensitivity of orthotopically injected ER-positive tumor in vivo, thus emphasizing the role of PYCR1 in resistance to chemotherapy.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Neoadjuvant Therapy , Proteomics , Breast Neoplasms/pathology , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cell Proliferation , Citric Acid Cycle , Female , Gene Regulatory Networks , Humans , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Prognosis , Protein Interaction Maps , Pyrroline Carboxylate Reductases/metabolism , Recurrence , Survival Analysis , delta-1-Pyrroline-5-Carboxylate Reductase
6.
Cell ; 179(1): 236-250.e18, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31495571

ABSTRACT

Immunotherapy has revolutionized cancer treatment, yet most patients do not respond. Here, we investigated mechanisms of response by profiling the proteome of clinical samples from advanced stage melanoma patients undergoing either tumor infiltrating lymphocyte (TIL)-based or anti- programmed death 1 (PD1) immunotherapy. Using high-resolution mass spectrometry, we quantified over 10,300 proteins in total and ∼4,500 proteins across most samples in each dataset. Statistical analyses revealed higher oxidative phosphorylation and lipid metabolism in responders than in non-responders in both treatments. To elucidate the effects of the metabolic state on the immune response, we examined melanoma cells upon metabolic perturbations or CRISPR-Cas9 knockouts. These experiments indicated lipid metabolism as a regulatory mechanism that increases melanoma immunogenicity by elevating antigen presentation, thereby increasing sensitivity to T cell mediated killing both in vitro and in vivo. Altogether, our proteomic analyses revealed association between the melanoma metabolic state and the response to immunotherapy, which can be the basis for future improvement of therapeutic response.


Subject(s)
Immunotherapy/methods , Melanoma/metabolism , Melanoma/therapy , Mitochondria/metabolism , Proteomics/methods , Skin Neoplasms/metabolism , Skin Neoplasms/therapy , Adoptive Transfer/methods , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cohort Studies , Female , Humans , Lipid Metabolism/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/immunology , Treatment Outcome , Young Adult
7.
Cell ; 176(3): 505-519.e22, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30612738

ABSTRACT

Genomic instability can be a hallmark of both human genetic disease and cancer. We identify a deleterious UBQLN4 mutation in families with an autosomal recessive syndrome reminiscent of genome instability disorders. UBQLN4 deficiency leads to increased sensitivity to genotoxic stress and delayed DNA double-strand break (DSB) repair. The proteasomal shuttle factor UBQLN4 is phosphorylated by ATM and interacts with ubiquitylated MRE11 to mediate early steps of homologous recombination-mediated DSB repair (HRR). Loss of UBQLN4 leads to chromatin retention of MRE11, promoting non-physiological HRR activity in vitro and in vivo. Conversely, UBQLN4 overexpression represses HRR and favors non-homologous end joining. Moreover, we find UBQLN4 overexpressed in aggressive tumors. In line with an HRR defect in these tumors, UBQLN4 overexpression is associated with PARP1 inhibitor sensitivity. UBQLN4 therefore curtails HRR activity through removal of MRE11 from damaged chromatin and thus offers a therapeutic window for PARP1 inhibitor treatment in UBQLN4-overexpressing tumors.


Subject(s)
Carrier Proteins/genetics , Nuclear Proteins/genetics , Carrier Proteins/metabolism , Chromatin/metabolism , DNA , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA End-Joining Repair , DNA-Binding Proteins/metabolism , Female , Genomic Instability , Germ-Line Mutation , Homologous Recombination , Humans , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Male , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/metabolism , Primary Cell Culture , Recombinational DNA Repair
8.
Nucleic Acids Res ; 45(10): 5945-5957, 2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28460002

ABSTRACT

Precise regulation of mRNA translation is critical for proper cell division, but little is known about the factors that mediate it. To identify mRNA-binding proteins that regulate translation during mitosis, we analyzed the composition of polysomes from interphase and mitotic cells using unbiased quantitative mass-spectrometry (LC-MS/MS). We found that mitotic polysomes are enriched with a subset of proteins involved in RNA processing, including alternative splicing and RNA export. To demonstrate that these may indeed be regulators of translation, we focused on heterogeneous nuclear ribonucleoprotein C (hnRNP C) as a test case and confirmed that it is recruited to elongating ribosomes during mitosis. Then, using a combination of pulsed SILAC, metabolic labeling and ribosome profiling, we showed that knockdown of hnRNP C affects both global and transcript-specific translation rates and found that hnRNP C is specifically important for translation of mRNAs that encode ribosomal proteins and translation factors. Taken together, our results demonstrate how proteomic analysis of polysomes can provide insight into translation regulation under various cellular conditions of interest and suggest that hnRNP C facilitates production of translation machinery components during mitosis to provide daughter cells with the ability to efficiently synthesize proteins as they enter G1 phase.


Subject(s)
Mitosis/genetics , Polyribosomes/physiology , Protein Biosynthesis/physiology , Proteomics/methods , RNA Splicing Factors/physiology , Chromatography, Liquid , G1 Phase , Gene Knockdown Techniques , Gene Ontology , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein Group C/antagonists & inhibitors , Heterogeneous-Nuclear Ribonucleoprotein Group C/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group C/physiology , Humans , Interphase , RNA Interference , RNA Splicing Factors/isolation & purification , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA-Binding Proteins/isolation & purification , RNA-Binding Proteins/physiology , Ribosomal Proteins/genetics , Tandem Mass Spectrometry
9.
J Neurochem ; 141(5): 694-707, 2017 06.
Article in English | MEDLINE | ID: mdl-28306143

ABSTRACT

Eukaryotic translation initiation factor 2B (eIF2B) is a master regulator of protein synthesis under normal and stress conditions. Mutations in any of the five genes encoding its subunits lead to vanishing white matter (VWM) disease, a recessive genetic deadly illness caused by progressive loss of white matter in the brain. In this study we used fibroblasts, which are not involved in the disease, to demonstrate the involvement of eIF2B in mitochondrial function and abundance. Mass spectrometry of total proteome of mouse embryonic fibroblasts (MEFs) isolated from Eif2b5R132H/R132H mice revealed unbalanced stoichiometry of proteins involved in oxidative phosphorylation and of mitochondrial translation machinery components, among others. Mutant MEFs exhibit 55% decrease in oxygen consumption rate per mtDNA content and 47% increase in mitochondrial abundance (p < 0.005), reflecting adaptation to energy requirements. A more robust eIF2B-associated oxidative respiration deficiency was found in mutant primary astrocytes, which exhibit > 3-fold lower ATP-linked respiration per cell despite a 2-fold increase in mtDNA content (p < 0.03). The 2-fold increase in basal and stimulated glycolysis in mutant astrocytes (p ≤ 0.03), but not in MEFs, demonstrates their higher energetic needs and further explicates their involvement in the disease. The data demonstrate the critical role of eIF2B in tight coordination of expression from nuclear and mitochondrial genomes and illuminates the importance of mitochondrial function in VWM pathology. Further dissection of the signaling network associated with eIF2B function will help generating therapeutic strategies for VWM disease and possibly other neurodegenerative disorders.


Subject(s)
Astrocytes/ultrastructure , Eukaryotic Initiation Factor-2B/genetics , Mitochondria/genetics , Mutation/genetics , Oxidative Phosphorylation , Oxygen Consumption/genetics , Animals , Animals, Newborn , Antimycin A/pharmacology , Astrocytes/drug effects , Astrocytes/physiology , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Size , Cells, Cultured , Chloramphenicol/pharmacology , Eukaryotic Initiation Factor-2B/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/ultrastructure , Histocompatibility Antigens/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/physiology , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Protein Synthesis Inhibitors/pharmacology , Proton Ionophores/pharmacology , Reactive Oxygen Species/metabolism
10.
J Exp Med ; 213(11): 2315-2331, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27697834

ABSTRACT

Tumor-associated macrophages (TAMs) promote tumor development, invasion, and dissemination by various mechanisms. In this study, using an orthotopic colorectal cancer (CRC) model, we found that monocyte-derived TAMs advance tumor development by the remodeling of its extracellular matrix (ECM) composition and structure. Unbiased transcriptomic and proteomic analyses of (a) TAM-abundant and -deficient tumor tissues and (b) sorted tumor-associated and -resident colonic macrophage subpopulations defined a distinct TAM-induced ECM molecular signature composed of an ensemble of matricellular proteins and remodeling enzymes they provide to the tumor microenvironment. Remarkably, many of these ECM proteins are specifically increased in human CRC versus healthy colon. Specifically, we demonstrate that although differentiating into TAMs, monocytes up-regulate matrix-remodeling programs associated with the synthesis and assembly of collagenous ECM, specifically collagen types I, VI, and XIV. This finding was further established by advanced imaging showing that TAMs instruct the deposition, cross-linking, and linearization of collagen fibers during tumor development, especially at areas of tumor invasiveness. Finally, we show that cancer-associated fibroblasts are significantly outnumbered by TAMs in this model and that their expression of collagen XIV and I is reduced by TAM deficiency. Here, we outline a novel TAM protumoral function associated with building of the collagenous ECM niche.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Extracellular Matrix/metabolism , Fibrillar Collagens/metabolism , Macrophages/pathology , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/ultrastructure , Disease Models, Animal , Extracellular Matrix/ultrastructure , Extracellular Matrix Proteins/metabolism , Fibrillar Collagens/genetics , Gene Expression Regulation, Neoplastic , Humans , Macrophages/metabolism , Mice, Inbred C57BL , Proteomics , Receptors, CCR2/deficiency , Receptors, CCR2/metabolism , Transcriptome/genetics , Tumor Microenvironment
11.
PLoS Genet ; 11(10): e1005554, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26439921

ABSTRACT

Studying the complex relationship between transcription, translation and protein degradation is essential to our understanding of biological processes in health and disease. The limited correlations observed between mRNA and protein abundance suggest pervasive regulation of post-transcriptional steps and support the importance of profiling mRNA levels in parallel to protein synthesis and degradation rates. In this work, we applied an integrative multi-omic approach to study gene expression along the mammalian cell cycle through side-by-side analysis of mRNA, translation and protein levels. Our analysis sheds new light on the significant contribution of both protein synthesis and degradation to the variance in protein expression. Furthermore, we find that translation regulation plays an important role at S-phase, while progression through mitosis is predominantly controlled by changes in either mRNA levels or protein stability. Specific molecular functions are found to be co-regulated and share similar patterns of mRNA, translation and protein expression along the cell cycle. Notably, these include genes and entire pathways not previously implicated in cell cycle progression, demonstrating the potential of this approach to identify novel regulatory mechanisms beyond those revealed by traditional expression profiling. Through this three-level analysis, we characterize different mechanisms of gene expression, discover new cycling gene products and highlight the importance and utility of combining datasets generated using different techniques that monitor distinct steps of gene expression.


Subject(s)
Protein Biosynthesis , Proteolysis , RNA, Messenger/biosynthesis , Transcription, Genetic , Cell Division/genetics , Gene Expression Regulation , HeLa Cells , Humans , Protein Biosynthesis/genetics , Proteome/genetics , RNA, Messenger/genetics , Transcriptome/genetics
12.
Expert Rev Proteomics ; 12(1): 13-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25404501

ABSTRACT

Stable isotope labeling with amino acids in cell culture (SILAC) has risen as a powerful quantification technique in mass spectrometry (MS)-based proteomics in classical and modified forms. Previously, SILAC was limited to cultured cells because of the requirement of active protein synthesis; however, in recent years, it was expanded to model organisms and tissue samples. Specifically, the super-SILAC technique uses a mixture of SILAC-labeled cells as a spike-in standard for accurate quantification of unlabeled samples, thereby enabling quantification of human tissue samples. Here, we highlight the recent developments in super-SILAC and its application to the study of clinical samples, secretomes, post-translational modifications and organelle proteomes. Finally, we propose super-SILAC as a robust and accurate method that can be commercialized and applied to basic and clinical research.


Subject(s)
Amino Acids/metabolism , Biomarkers, Tumor/metabolism , Neoplasms/diagnosis , Proteomics/methods , Animals , Cells, Cultured , Humans , Isotope Labeling/methods , Isotope Labeling/trends , Proteomics/trends
SELECTION OF CITATIONS
SEARCH DETAIL
...