24 results on '"Maliga Z"'
Search Results
2. Motor-Independent Targeting of CLASPs to Kinetochores by CENP-E Promotes Microtubule Turnover and Poleward Flux
- Author
-
Maffini, S, Maia, ARR, Manning, AL, Maliga, Z, Pereira, AL, Junqueira, M, Shevchenko, A, Hyman, A, Yates, JR, Galjart, N, Compton, DA, Maiato, H, and Instituto de Biologia Molecular e Celular
- Subjects
Flux ,Mitotic Spindle ,Mitosis ,Kinetochores ,Microtubule Turnover - Abstract
Efficient chromosome segregation during mitosis relies on the coordinated activity of molecular motors with proteins that regulate kinetochore attachments to dynamic spindle microtubules [1]. CLASPs are conserved kinetochore- and microtubule-associated proteins encoded by two paralogue genes, clasp1 and clasp2, and have been previously implicated in the regulation of kinetochore-microtubule dynamics [2-4]. However, it remains unknown how CLASPs work in concert with other proteins to form a functional kinetochore-microtubule interface. Here we have identified mitotic interactors of human CLASP1 using a proteomic approach. Among these, the microtubule plus-end directed motor CENP-E [5] was found to form a complex with CLASP1 that co-localizes to multiple structures of the mitotic apparatus in human cells. We found that CENP-E recruits both CLASP1 and CLASP2 to kinetochores independent of its motor activity or the presence of microtubules. Depletion of CLASPs or CENP-E by RNAi in human cells causes a significant and comparable reduction of kinetochore-microtubule poleward flux and turnover rates, as well as rescues spindle bipolarity in Kif2a-depleted cells. We conclude that CENP-E integrates two critical functions that are important for accurate chromosome movement and spindle architecture: one relying directly on its motor activity and the other involving the targeting of key microtubule regulators to kinetochores.
- Published
- 2009
- Full Text
- View/download PDF
3. Multiplexed 3D Analysis of Immune States and Niches in Human Tissue.
- Author
-
Yapp C, Nirmal AJ, Zhou F, Maliga Z, Tefft JB, Llopis PM, Murphy GF, Lian CG, Danuser G, Santagata S, and Sorger PK
- Abstract
Tissue homeostasis and the emergence of disease are controlled by changes in the proportions of resident and recruited cells, their organization into cellular neighbourhoods, and their interactions with acellular tissue components. Highly multiplexed tissue profiling (spatial omics)
1 makes it possible to study this microenvironment in situ , usually in 4-5 micron thick sections (the standard histopathology format)2 . Microscopy-based tissue profiling is commonly performed at a resolution sufficient to determine cell types but not to detect subtle morphological features associated with cytoskeletal reorganisation, juxtracrine signalling, or membrane trafficking3 . Here we describe a high-resolution 3D imaging approach able to characterize a wide variety of organelles and structures at sub-micron scale while simultaneously quantifying millimetre-scale spatial features. This approach combines cyclic immunofluorescence (CyCIF) imaging4 of over 50 markers with confocal microscopy of archival human tissue thick enough (30-40 microns) to fully encompass two or more layers of intact cells. 3D imaging of entire cell volumes substantially improves the accuracy of cell phenotyping and allows cell proximity to be scored using plasma membrane apposition, not just nuclear position. In pre-invasive melanoma in situ5 , precise phenotyping shows that adjacent melanocytic cells are plastic in state and participate in tightly localised niches of interferon signalling near sites of initial invasion into the underlying dermis. In this and metastatic melanoma, mature and precursor T cells engage in an unexpectedly diverse array of juxtracrine and membrane-membrane interactions as well as looser "neighbourhood" associations6 whose morphologies reveal functional states. These data provide new insight into the transitions occurring during early tumour formation and immunoediting and demonstrate the potential for phenotyping of tissues at a level of detail previously restricted to cultured cells and organoids.- Published
- 2024
- Full Text
- View/download PDF
4. SpatialCells: automated profiling of tumor microenvironments with spatially resolved multiplexed single-cell data.
- Author
-
Wan G, Maliga Z, Yan B, Vallius T, Shi Y, Khattab S, Chang C, Nirmal AJ, Yu KH, Liu D, Lian CG, DeSimone MS, Sorger PK, and Semenov YR
- Subjects
- Humans, Neoplasms pathology, Machine Learning, Computational Biology methods, Tumor Microenvironment, Single-Cell Analysis methods, Software
- Abstract
Cancer is a complex cellular ecosystem where malignant cells coexist and interact with immune, stromal and other cells within the tumor microenvironment (TME). Recent technological advancements in spatially resolved multiplexed imaging at single-cell resolution have led to the generation of large-scale and high-dimensional datasets from biological specimens. This underscores the necessity for automated methodologies that can effectively characterize molecular, cellular and spatial properties of TMEs for various malignancies. This study introduces SpatialCells, an open-source software package designed for region-based exploratory analysis and comprehensive characterization of TMEs using multiplexed single-cell data. The source code and tutorials are available at https://semenovlab.github.io/SpatialCells. SpatialCells efficiently streamlines the automated extraction of features from multiplexed single-cell data and can process samples containing millions of cells. Thus, SpatialCells facilitates subsequent association analyses and machine learning predictions, making it an essential tool in advancing our understanding of tumor growth, invasion and metastasis., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
5. SpatialCells: Automated Profiling of Tumor Microenvironments with Spatially Resolved Multiplexed Single-Cell Data.
- Author
-
Wan G, Maliga Z, Yan B, Vallius T, Shi Y, Khattab S, Chang C, Nirmal AJ, Yu KH, Liu D, Lian CG, DeSimone MS, Sorger PK, and Semenov YR
- Abstract
Background: Cancer is a complex cellular ecosystem where malignant cells coexist and interact with immune, stromal, and other cells within the tumor microenvironment. Recent technological advancements in spatially resolved multiplexed imaging at single-cell resolution have led to the generation of large-scale and high-dimensional datasets from biological specimens. This underscores the necessity for automated methodologies that can effectively characterize the molecular, cellular, and spatial properties of tumor microenvironments for various malignancies., Results: This study introduces SpatialCells, an open-source software package designed for region-based exploratory analysis and comprehensive characterization of tumor microenvironments using multiplexed single-cell data., Conclusions: SpatialCells efficiently streamlines the automated extraction of features from multiplexed single-cell data and can process samples containing millions of cells. Thus, SpatialCells facilitates subsequent association analyses and machine learning predictions, making it an essential tool in advancing our understanding of tumor growth, invasion, and metastasis.
- Published
- 2023
- Full Text
- View/download PDF
6. Immune Profiling of Dermatologic Adverse Events from Checkpoint Blockade using Tissue Cyclic Immunofluorescence.
- Author
-
Maliga Z, Kim DY, Bui AN, Lin JR, Dewan AK, Murphy GF, Nirmal AJ, Lian CG, Sorger PK, and LeBoeuf NR
- Abstract
In this study, we demonstrate the utility of whole-slide CyCIF (tissue-based cyclic immunofluorescence) imaging for characterizing immune cell infiltrates in immune checkpoint inhibitor (ICI)-induced dermatologic adverse events (dAEs). We analyzed six cases of ICI-induced dAEs, including lichenoid, bullous pemphigoid, psoriasis, and eczematous eruptions, comparing immune profiling results obtained using both standard immunohistochemistry (IHC) and CyCIF. Our findings indicate that CyCIF provides more detailed and precise single-cell characterization of immune cell infiltrates than IHC, which relies on semi-quantitative scoring by pathologists. This pilot study highlights the potential of CyCIF to advance our understanding of the immune environment in dAEs by revealing tissue-level spatial patterns of immune cell infiltrates, allowing for more precise phenotypic distinctions and deeper exploration of disease mechanisms. By demonstrating that CyCIF can be performed on friable tissues, such as bullous pemphigoid, we provide a foundation for future studies to examine the drivers of specific dAEs using larger cohorts of phenotyped toxicity and suggest a broader role for highly multiplexed tissue imaging in phenotyping the immune mediated disease that they resemble.
- Published
- 2023
- Full Text
- View/download PDF
7. UnMICST: Deep learning with real augmentation for robust segmentation of highly multiplexed images of human tissues.
- Author
-
Yapp C, Novikov E, Jang WD, Vallius T, Chen YA, Cicconet M, Maliga Z, Jacobson CA, Wei D, Santagata S, Pfister H, and Sorger PK
- Subjects
- Humans, Animals, Image Processing, Computer-Assisted methods, Machine Learning, Cell Nucleus, Mammals, Deep Learning
- Abstract
Upcoming technologies enable routine collection of highly multiplexed (20-60 channel), subcellular resolution images of mammalian tissues for research and diagnosis. Extracting single cell data from such images requires accurate image segmentation, a challenging problem commonly tackled with deep learning. In this paper, we report two findings that substantially improve image segmentation of tissues using a range of machine learning architectures. First, we unexpectedly find that the inclusion of intentionally defocused and saturated images in training data substantially improves subsequent image segmentation. Such real augmentation outperforms computational augmentation (Gaussian blurring). In addition, we find that it is practical to image the nuclear envelope in multiple tissues using an antibody cocktail thereby better identifying nuclear outlines and improving segmentation. The two approaches cumulatively and substantially improve segmentation on a wide range of tissue types. We speculate that the use of real augmentations will have applications in image processing outside of microscopy., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
8. Spatial intra-tumor heterogeneity is associated with survival of lung adenocarcinoma patients.
- Author
-
Wu HJ, Temko D, Maliga Z, Moreira AL, Sei E, Minussi DC, Dean J, Lee C, Xu Q, Hochart G, Jacobson CA, Yapp C, Schapiro D, Sorger PK, Seeley EH, Navin N, Downey RJ, and Michor F
- Abstract
Intra-tumor heterogeneity (ITH) of human tumors is important for tumor progression, treatment response, and drug resistance. However, the spatial distribution of ITH remains incompletely understood. Here, we present spatial analysis of ITH in lung adenocarcinomas from 147 patients using multi-region mass spectrometry of >5,000 regions, single-cell copy number sequencing of ~2,000 single cells, and cyclic immunofluorescence of >10 million cells. We identified two distinct spatial patterns among tumors, termed clustered and random geographic diversification (GD). These patterns were observed in the same samples using both proteomic and genomic data. The random proteomic GD pattern, which is characterized by decreased cell adhesion and lower levels of tumor-interacting endothelial cells, was significantly associated with increased risk of recurrence or death in two independent patient cohorts. Our study presents comprehensive spatial mapping of ITH in lung adenocarcinoma and provides insights into the mechanisms and clinical consequences of GD., Competing Interests: DECLARATION OF INTERESTS The other authors declare no competing financial interests.
- Published
- 2022
- Full Text
- View/download PDF
9. Obesity Shapes Metabolism in the Tumor Microenvironment to Suppress Anti-Tumor Immunity.
- Author
-
Ringel AE, Drijvers JM, Baker GJ, Catozzi A, García-Cañaveras JC, Gassaway BM, Miller BC, Juneja VR, Nguyen TH, Joshi S, Yao CH, Yoon H, Sage PT, LaFleur MW, Trombley JD, Jacobson CA, Maliga Z, Gygi SP, Sorger PK, Rabinowitz JD, Sharpe AH, and Haigis MC
- Subjects
- Adiposity, Animals, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Cell Proliferation, Diet, High-Fat, Fatty Acids metabolism, HEK293 Cells, Humans, Hypoxia-Inducible Factor-Proline Dioxygenases metabolism, Kinetics, Lymphocytes, Tumor-Infiltrating, Mice, Inbred C57BL, Mice, Knockout, Oxidation-Reduction, Principal Component Analysis, Procollagen-Proline Dioxygenase metabolism, Proteomics, Immunity, Neoplasms immunology, Neoplasms metabolism, Obesity metabolism, Tumor Microenvironment
- Abstract
Obesity is a major cancer risk factor, but how differences in systemic metabolism change the tumor microenvironment (TME) and impact anti-tumor immunity is not understood. Here, we demonstrate that high-fat diet (HFD)-induced obesity impairs CD8
+ T cell function in the murine TME, accelerating tumor growth. We generate a single-cell resolution atlas of cellular metabolism in the TME, detailing how it changes with diet-induced obesity. We find that tumor and CD8+ T cells display distinct metabolic adaptations to obesity. Tumor cells increase fat uptake with HFD, whereas tumor-infiltrating CD8+ T cells do not. These differential adaptations lead to altered fatty acid partitioning in HFD tumors, impairing CD8+ T cell infiltration and function. Blocking metabolic reprogramming by tumor cells in obese mice improves anti-tumor immunity. Analysis of human cancers reveals similar transcriptional changes in CD8+ T cell markers, suggesting interventions that exploit metabolism to improve cancer immunotherapy., Competing Interests: Declaration of Interests A.H.S. has patents/pending royalties on intellectual property on the PD-1 pathway from Roche and Novartis. A.H.S. is on advisory boards for Surface Oncology, Elstar, SQZ Biotechnologies, Selecta, Elpiscience, and Monopteros and has research funding from Novartis, Roche, Ipsen, Quark, and Merck. M.C.H. has patents pending on the PHD3 pathway, is on the scientific advisory board for Pori Therapeutics, and has research funding from Roche. J.M.D. has consulted for ElevateBio and Third Rock Ventures. The remaining authors declare no competing interests., (Copyright © 2020. Published by Elsevier Inc.)- Published
- 2020
- Full Text
- View/download PDF
10. Vascular Disease and Thrombosis in SARS-CoV-2-Infected Rhesus Macaques.
- Author
-
Aid M, Busman-Sahay K, Vidal SJ, Maliga Z, Bondoc S, Starke C, Terry M, Jacobson CA, Wrijil L, Ducat S, Brook OR, Miller AD, Porto M, Pellegrini KL, Pino M, Hoang TN, Chandrashekar A, Patel S, Stephenson K, Bosinger SE, Andersen H, Lewis MG, Hecht JL, Sorger PK, Martinot AJ, Estes JD, and Barouch DH
- Subjects
- Aged, 80 and over, Animals, Bronchoalveolar Lavage, C-Reactive Protein analysis, COVID-19 blood, COVID-19 pathology, Complement Activation, Cytokines blood, Female, Humans, Inflammation blood, Inflammation immunology, Inflammation virology, Lung pathology, Macaca mulatta, Macrophages immunology, Male, Platelet Activation, Thrombosis blood, Thrombosis pathology, Transcriptome, Vascular Diseases blood, Vascular Diseases pathology, COVID-19 complications, COVID-19 immunology, SARS-CoV-2 genetics, Thrombosis complications, Vascular Diseases complications
- Abstract
The COVID-19 pandemic has led to extensive morbidity and mortality throughout the world. Clinical features that drive SARS-CoV-2 pathogenesis in humans include inflammation and thrombosis, but the mechanistic details underlying these processes remain to be determined. In this study, we demonstrate endothelial disruption and vascular thrombosis in histopathologic sections of lungs from both humans and rhesus macaques infected with SARS-CoV-2. To define key molecular pathways associated with SARS-CoV-2 pathogenesis in macaques, we performed transcriptomic analyses of bronchoalveolar lavage and peripheral blood and proteomic analyses of serum. We observed macrophage infiltrates in lung and upregulation of macrophage, complement, platelet activation, thrombosis, and proinflammatory markers, including C-reactive protein, MX1, IL-6, IL-1, IL-8, TNFα, and NF-κB. These results suggest a model in which critical interactions between inflammatory and thrombosis pathways lead to SARS-CoV-2-induced vascular disease. Our findings suggest potential therapeutic targets for COVID-19., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
11. Author Correction: Immunogenomic profiling determines responses to combined PARP and PD-1 inhibition in ovarian cancer.
- Author
-
Färkkilä A, Gulhan DC, Casado J, Jacobson CA, Nguyen H, Kochupurakkal B, Maliga Z, Yapp C, Chen YA, Schapiro D, Zhou Y, Graham JR, Dezube BJ, Munster P, Santagata S, Garcia E, Rodig S, Lako A, Chowdhury D, Shapiro GI, Matulonis UA, Park PJ, Hautaniemi S, Sorger PK, Swisher EM, D'Andrea AD, and Konstantinopoulos PA
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
12. Immunogenomic profiling determines responses to combined PARP and PD-1 inhibition in ovarian cancer.
- Author
-
Färkkilä A, Gulhan DC, Casado J, Jacobson CA, Nguyen H, Kochupurakkal B, Maliga Z, Yapp C, Chen YA, Schapiro D, Zhou Y, Graham JR, Dezube BJ, Munster P, Santagata S, Garcia E, Rodig S, Lako A, Chowdhury D, Shapiro GI, Matulonis UA, Park PJ, Hautaniemi S, Sorger PK, Swisher EM, D'Andrea AD, and Konstantinopoulos PA
- Subjects
- Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized pharmacology, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, B7-H1 Antigen immunology, B7-H1 Antigen metabolism, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, DNA Mutational Analysis, Drug Monitoring methods, Female, Gene Amplification, Humans, Indazoles pharmacology, Indazoles therapeutic use, Interferons immunology, Interferons metabolism, Macrophages drug effects, Macrophages immunology, Macrophages metabolism, Middle Aged, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local immunology, Neoplasm Recurrence, Local pathology, Ovarian Neoplasms genetics, Ovarian Neoplasms immunology, Ovarian Neoplasms pathology, Ovary pathology, Piperidines pharmacology, Piperidines therapeutic use, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Programmed Cell Death 1 Ligand 2 Protein genetics, Programmed Cell Death 1 Ligand 2 Protein metabolism, Recombinational DNA Repair genetics, Single-Cell Analysis, Treatment Outcome, Tumor Microenvironment drug effects, Tumor Microenvironment genetics, Tumor Microenvironment immunology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, B7-H1 Antigen genetics, Neoplasm Recurrence, Local drug therapy, Ovarian Neoplasms drug therapy, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Programmed Cell Death 1 Receptor antagonists & inhibitors
- Abstract
Combined PARP and immune checkpoint inhibition has yielded encouraging results in ovarian cancer, but predictive biomarkers are lacking. We performed immunogenomic profiling and highly multiplexed single-cell imaging on tumor samples from patients enrolled in a Phase I/II trial of niraparib and pembrolizumab in ovarian cancer (NCT02657889). We identify two determinants of response; mutational signature 3 reflecting defective homologous recombination DNA repair, and positive immune score as a surrogate of interferon-primed exhausted CD8 + T-cells in the tumor microenvironment. Presence of one or both features associates with an improved outcome while concurrent absence yields no responses. Single-cell spatial analysis reveals prominent interactions of exhausted CD8 + T-cells and PD-L1 + macrophages and PD-L1 + tumor cells as mechanistic determinants of response. Furthermore, spatial analysis of two extreme responders shows differential clustering of exhausted CD8 + T-cells with PD-L1 + macrophages in the first, and exhausted CD8 + T-cells with cancer cells harboring genomic PD-L1 and PD-L2 amplification in the second.
- Published
- 2020
- Full Text
- View/download PDF
13. Highly multiplexed immunofluorescence images and single-cell data of immune markers in tonsil and lung cancer.
- Author
-
Rashid R, Gaglia G, Chen YA, Lin JR, Du Z, Maliga Z, Schapiro D, Yapp C, Muhlich J, Sokolov A, Sorger P, and Santagata S
- Subjects
- Algorithms, Formaldehyde, Humans, Paraffin Embedding, Software, Tissue Fixation, Biomarkers, Tumor immunology, Fluorescent Antibody Technique, Lung Neoplasms immunology, Palatine Tonsil immunology, Single-Cell Analysis
- Abstract
In this data descriptor, we document a dataset of multiplexed immunofluorescence images and derived single-cell measurements of immune lineage and other markers in formaldehyde-fixed and paraffin-embedded (FFPE) human tonsil and lung cancer tissue. We used tissue cyclic immunofluorescence (t-CyCIF) to generate fluorescence images which we artifact corrected using the BaSiC tool, stitched and registered using the ASHLAR algorithm, and segmented using ilastik software and MATLAB. We extracted single-cell features from these images using HistoCAT software. The resulting dataset can be visualized using image browsers and analyzed using high-dimensional, single-cell methods. This dataset is a valuable resource for biological discovery of the immune system in normal and diseased states as well as for the development of multiplexed image analysis and viewing tools.
- Published
- 2019
- Full Text
- View/download PDF
14. Phase Transitions Drive the Formation of Vesicular Stomatitis Virus Replication Compartments.
- Author
-
Heinrich BS, Maliga Z, Stein DA, Hyman AA, and Whelan SPJ
- Subjects
- Animals, Cell Compartmentation, Cell Line, Chlorocebus aethiops, RNA, Viral, Vero Cells, Viral Proteins genetics, Cytoplasmic Granules virology, Host Microbial Interactions, Inclusion Bodies, Viral physiology, Vesiculovirus physiology, Viral Proteins metabolism, Virus Replication
- Abstract
RNA viruses that replicate in the cell cytoplasm typically concentrate their replication machinery within specialized compartments. This concentration favors enzymatic reactions and shields viral RNA from detection by cytosolic pattern recognition receptors. Nonsegmented negative-strand (NNS) RNA viruses, which include some of the most significant human, animal, and plant pathogens extant, form inclusions that are sites of RNA synthesis and are not circumscribed by a membrane. These inclusions share similarities with cellular protein/RNA structures such as P granules and nucleoli, which are phase-separated liquid compartments. Here we show that replication compartments of vesicular stomatitis virus (VSV) have the properties of liquid-like compartments that form by phase separation. Expression of the individual viral components of the replication machinery in cells demonstrates that the 3 viral proteins required for replication are sufficient to drive cytoplasmic phase separation. Therefore, liquid-liquid phase separation, previously linked to organization of P granules, nucleolus homeostasis, and cell signaling, plays a key role in host-pathogen interactions. This work suggests novel therapeutic approaches to the problem of combating NNS RNA viral infections. IMPORTANCE RNA viruses compartmentalize their replication machinery to evade detection by host pattern recognition receptors and concentrate the machinery of RNA synthesis. For positive-strand RNA viruses, RNA replication occurs in a virus-induced membrane-associated replication organelle. For NNS RNA viruses, the replication compartment is a cytoplasmic inclusion that is not circumscribed by a cellular membrane. Such structures were first observed in the cell bodies of neurons from humans infected with rabies virus and were termed Negri bodies. How the replication machinery that forms this inclusion remains associated in the absence of a membrane has been an enduring mystery. In this article, we present evidence that the VSV replication compartments form through phase separation. Phase separation is increasingly recognized as responsible for cellular structures as diverse as processing bodies (P-bodies) and nucleoli and was recently demonstrated for rabies virus. This article further links the fields of host-pathogen interaction with that of phase separation., (Copyright © 2018 Heinrich et al.)
- Published
- 2018
- Full Text
- View/download PDF
15. Extracellular poly(ADP-ribose) is a pro-inflammatory signal for macrophages.
- Author
-
Krukenberg KA, Kim S, Tan ES, Maliga Z, and Mitchison TJ
- Subjects
- Animals, Cell Line, Cytokines metabolism, Dimerization, Humans, Macrophages cytology, Macrophages metabolism, Mice, Microscopy, Confocal, Poly Adenosine Diphosphate Ribose chemistry, Structure-Activity Relationship, Toll-Like Receptor 2 antagonists & inhibitors, Toll-Like Receptor 2 metabolism, Toll-Like Receptor 4 antagonists & inhibitors, Toll-Like Receptor 4 metabolism, Tumor Necrosis Factor-alpha metabolism, Poly Adenosine Diphosphate Ribose pharmacology, Poly(ADP-ribose) Polymerases metabolism, Signal Transduction drug effects
- Abstract
Poly(ADP-ribose) polymerase 1 (PARP1) synthesizes poly(ADP-ribose) (PAR), an essential post-translational modification whose function is important in many cellular processes including DNA damage signaling, cell death, and inflammation. All known PAR biology is intracellular, but we suspected it might also play a role in cell-to-cell communication during inflammation. We found that PAR activated cytokine release in human and mouse macrophages, a hallmark of innate immune activation, and determined structure-activity relationships. PAR was rapidly internalized by murine macrophages, while the monomer, ADP-ribose, was not. Inhibitors of Toll-like receptor 2 (TLR2) and TLR4 signaling blocked macrophage responses to PAR, and PAR induced TLR2 and TLR4 signaling in reporter cell lines suggesting it was recognized by these TLRs, much like bacterial pathogens. We propose that PAR acts as an extracellular damage associated molecular pattern that drives inflammatory signaling., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
16. A tethering mechanism controls the processivity and kinetochore-microtubule plus-end enrichment of the kinesin-8 Kif18A.
- Author
-
Stumpff J, Du Y, English CA, Maliga Z, Wagenbach M, Asbury CL, Wordeman L, and Ohi R
- Subjects
- Chromosome Positioning, HeLa Cells, Humans, Kinesins genetics, Kinesins metabolism, Kinetochores metabolism, Microtubules metabolism
- Abstract
Metaphase chromosome positioning depends on Kif18A, a kinesin-8 that accumulates at and suppresses the dynamics of K-MT plus ends. By engineering Kif18A mutants that suppress MT dynamics but fail to concentrate at K-MT plus ends, we identify a mechanism that allows Kif18A to accumulate at K-MT plus ends to a level required to suppress chromosome movements. Enrichment of Kif18A at K-MT plus ends depends on its C-terminal tail domain, while the ability of Kif18A to suppress MT growth is conferred by the N-terminal motor domain. The Kif18A tail contains a second MT-binding domain that diffuses along the MT lattice, suggesting that it tethers the motor to the MT track. Consistently, the tail enhances Kif18A processivity and is crucial for it to accumulate at K-MT plus ends. The heightened processivity of Kif18A, conferred by its tail domain, thus promotes concentration of Kif18A at K-MT plus ends, where it suppresses their dynamics to control chromosome movements., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
17. Loop L5 acts as a conformational latch in the mitotic kinesin Eg5.
- Author
-
Behnke-Parks WM, Vendome J, Honig B, Maliga Z, Moores C, and Rosenfeld SS
- Subjects
- Animals, Catalytic Domain, Humans, Kinesins genetics, Mutagenesis, Site-Directed, Protein Structure, Secondary, Computer Simulation, Kinesins chemistry, Models, Molecular
- Abstract
All members of the kinesin superfamily of molecular motors contain an unusual structural motif consisting of an α-helix that is interrupted by a flexible loop, referred to as L5. We have examined the function of L5 in the mitotic kinesin Eg5 by combining site-directed mutagenesis of L5 with transient state kinetics, molecular dynamics simulations, and docking using cryo electron microscopy density. We find that mutation of a proline residue located at a turn within this loop profoundly slows nucleotide-induced structural changes both at the catalytic site as well as at the microtubule binding domain and the neck linker. Molecular dynamics simulations reveal that this mutation affects the dynamics not only of L5 itself but also of the switch I structural elements that sense ATP binding to the catalytic site. Our results lead us to propose that L5 regulates the rate of conformational change in key elements of the nucleotide binding site through its interactions with α3 and in so doing controls the speed of movement and force generation in kinesin motors.
- Published
- 2011
- Full Text
- View/download PDF
18. A protein domain-based interactome network for C. elegans early embryogenesis.
- Author
-
Boxem M, Maliga Z, Klitgord N, Li N, Lemmens I, Mana M, de Lichtervelde L, Mul JD, van de Peut D, Devos M, Simonis N, Yildirim MA, Cokol M, Kao HL, de Smet AS, Wang H, Schlaitz AL, Hao T, Milstein S, Fan C, Tipsword M, Drew K, Galli M, Rhrissorrakrai K, Drechsel D, Koller D, Roth FP, Iakoucheva LM, Dunker AK, Bonneau R, Gunsalus KC, Hill DE, Piano F, Tavernier J, van den Heuvel S, Hyman AA, and Vidal M
- Subjects
- Animals, Cell Division, Protein Interaction Domains and Motifs, Proteome, Two-Hybrid System Techniques, Caenorhabditis elegans embryology, Embryo, Nonmammalian metabolism, Embryonic Development, Protein Interaction Mapping
- Abstract
Many protein-protein interactions are mediated through independently folding modular domains. Proteome-wide efforts to model protein-protein interaction or "interactome" networks have largely ignored this modular organization of proteins. We developed an experimental strategy to efficiently identify interaction domains and generated a domain-based interactome network for proteins involved in C. elegans early-embryonic cell divisions. Minimal interacting regions were identified for over 200 proteins, providing important information on their domain organization. Furthermore, our approach increased the sensitivity of the two-hybrid system, resulting in a more complete interactome network. This interactome modeling strategy revealed insights into C. elegans centrosome function and is applicable to other biological processes in this and other organisms.
- Published
- 2008
- Full Text
- View/download PDF
19. Influence of HDL-cholesterol-elevating drugs on the in vitro activity of the HDL receptor SR-BI.
- Author
-
Nieland TJ, Shaw JT, Jaipuri FA, Maliga Z, Duffner JL, Koehler AN, and Krieger M
- Subjects
- Anticholesteremic Agents chemical synthesis, Cells, Cultured, Clofibric Acid pharmacology, Dose-Response Relationship, Drug, Fenofibrate pharmacology, Humans, Scavenger Receptors, Class B antagonists & inhibitors, Thiourea analogs & derivatives, Thiourea chemical synthesis, Thiourea pharmacology, Anticholesteremic Agents pharmacology, Cholesterol, HDL metabolism, Lipoproteins, HDL metabolism, Receptors, Lipoprotein metabolism, Scavenger Receptors, Class B metabolism
- Abstract
Treatment of atherosclerotic disease often focuses on reducing plasma LDL-cholesterol or increasing plasma HDL-cholesterol. We examined in vitro the effects on HDL receptor [scavenger receptor class B type I (SR-BI)] activity of three classes of clinical and experimental plasma HDL-cholesterol-elevating compounds: niacin, fibrates, and HDL376. Fenofibrate (FF) and HDL376 were potent (IC(50) approximately 1 microM), direct inhibitors of SR-BI-mediated lipid transport in cells and in liposomes reconstituted with purified SR-BI. FF, a prodrug, was a more potent inhibitor of SR-BI than an activator of peroxisome proliferator-activated receptor alpha, a target of its active fenofibric acid (FFA) derivative. Nevertheless, FFA, four other fibrates (clofibrate, gemfibrozil, ciprofibrate, and bezafibrate), and niacin had little, if any, effect on SR-BI, suggesting that they do not directly target SR-BI in vivo. However, similarities of HDL376 treatment and SR-BI gene knockout on HDL metabolism in vivo (increased HDL-cholesterol and HDL particle sizes) and structure-activity relationship analysis suggest that SR-BI may be a target of HDL376 in vivo. HDL376 and other inhibitors may help elucidate SR-BI function in diverse mammalian models and determine the therapeutic potential of SR-BI-directed pharmaceuticals.
- Published
- 2007
- Full Text
- View/download PDF
20. A pathway of structural changes produced by monastrol binding to Eg5.
- Author
-
Maliga Z, Xing J, Cheung H, Juszczak LJ, Friedman JM, and Rosenfeld SS
- Subjects
- Anisotropy, Binding Sites, Crystallography, X-Ray, Dose-Response Relationship, Drug, Fluorescence Resonance Energy Transfer, Kinetics, Mitosis, Models, Chemical, Models, Molecular, Nucleotides chemistry, Protein Binding, Protein Structure, Secondary, Protein Structure, Tertiary, Spectrometry, Fluorescence, Spectrophotometry, Spectrum Analysis, Raman, Substrate Specificity, Time Factors, Tryptophan chemistry, Ultraviolet Rays, Kinesins chemistry, Pyrimidines chemistry, Thiones chemistry
- Abstract
Monastrol is a small molecule inhibitor that is specific for Eg5, a member of the kinesin 5 family of mitotic motors. Crystallographic models of Eg5 in the presence and absence of monastrol revealed that drug binding produces a variety of structural changes in the motor, including in loop L5 and the neck linker. What is not clear from static crystallographic models, however, is the sequence of structural changes produced by drug binding. Furthermore, because crystallographic structures can be influenced by the packing forces in the crystal, it also remains unclear whether these drug-induced changes occur in solution, at physiologically active concentrations of monastrol or of other drugs that target this site. We have addressed these issues by using a series of spectroscopic probes to monitor the structural consequences of drug binding. Our results demonstrated that the crystallographic model of an Eg5-ADP-monastrol ternary complex is consistent with several solution-based spectroscopic probes. Furthermore, the kinetics of these spectroscopic signal changes allowed us to determine the temporal sequence of drug-induced structural transitions. These results suggested that L5 may be an element in the pathway that links the state of the nucleotide-binding site to the neck linker in kinesin motors.
- Published
- 2006
- Full Text
- View/download PDF
21. Small-molecule and mutational analysis of allosteric Eg5 inhibition by monastrol.
- Author
-
Maliga Z and Mitchison TJ
- Abstract
Background: A recent crystal structure of monastrol in a ternary complex with the kinesin Eg5 motor domain highlights a novel, induced-fit drug binding site at atomic resolution. Mutational obliteration of the monastrol binding site results in a monastrol-resistant, but otherwise catalytically active Eg5 motor domain. However, considering the conformational changes at this site, it is unclear what specific interactions stabilize the interaction between monastrol and the Eg5 motor domain., Results: To study the molecular complementarity of the monastrol-Eg5 interaction, we used a combination of synthetic chemistry and targeted mutations in Eg5 to measure the contribution of specific contacts to inhibition of Eg5 in vitro and in cultured cells. Structure-activity data on chemical derivatives, sequence analysis of Eg5 homologs from different species, and the effect of mutations near the drug binding site were consistent with the crystal structure., Conclusion: The mechanism of monastrol revealed by our data rationalizes its specificity for Eg5 over other kinesins and highlights a potential mechanism of drug resistance for anti-cancer therapy targeting this site in Eg5.
- Published
- 2006
- Full Text
- View/download PDF
22. Mechanistic analysis of the mitotic kinesin Eg5.
- Author
-
Cochran JC, Sontag CA, Maliga Z, Kapoor TM, Correia JJ, and Gilbert SP
- Subjects
- Adenosine Diphosphate chemistry, Adenosine Triphosphatases chemistry, Adenosine Triphosphatases metabolism, Adenosine Triphosphate chemistry, Binding Sites, Cell Division, Chromatography, Gel, Dose-Response Relationship, Drug, Drosophila Proteins metabolism, Humans, Hydrolysis, Kinesins metabolism, Kinetics, Microtubule-Associated Proteins, Microtubules metabolism, Models, Chemical, Phosphocreatine chemistry, Protein Binding, Protein Structure, Tertiary, Saccharomyces cerevisiae Proteins, Time Factors, Ultracentrifugation, Kinesins chemistry, Mitosis
- Abstract
Eg5 is a slow, plus-end-directed microtubule-based motor of the BimC kinesin family that is essential for bipolar spindle formation during eukaryotic cell division. We have analyzed two human Eg5/KSP motors, Eg5-367 and Eg5-437, and both are monomeric based on results from sedimentation velocity and sedimentation equilibrium centrifugation as well as analytical gel filtration. The steady-state parameters were: for Eg5-367: k(cat) = 5.5 s(-1), K(1/2,Mt) = 0.7 microm, and K(m,ATP) = 25 microm; and for Eg5-437: k(cat) = 2.9 s(-1), K(1/2,Mt) = 4.5 microm, and K(m,ATP) = 19 microm. 2'(3')-O-(N-Methylanthraniloyl)-ATP (mantATP) binding was rapid at 2-3 microm(-1)s(-1), followed immediately by ATP hydrolysis at 15 s(-1). ATP-dependent Mt.Eg5 dissociation was relatively slow and rate-limiting at 8 s(-1) with mantADP release at 40 s(-1). Surprisingly, Eg5-367 binds microtubules more effectively (11 microm(-1)s(-1)) than Eg5-437 (0.7 microm(-1)s(-1)), consistent with the steady-state K(1/2,Mt) and the mantADP release K(1/2,Mt). These results indicate that the ATPase pathway for monomeric Eg5 is more similar to conventional kinesin than the spindle motors Ncd and Kar3, where ADP product release is rate-limiting for steady-state turnover.
- Published
- 2004
- Full Text
- View/download PDF
23. Cross-inhibition of SR-BI- and ABCA1-mediated cholesterol transport by the small molecules BLT-4 and glyburide.
- Author
-
Nieland TJ, Chroni A, Fitzgerald ML, Maliga Z, Zannis VI, Kirchhausen T, and Krieger M
- Subjects
- ATP Binding Cassette Transporter 1, ATP-Binding Cassette Transporters metabolism, Apolipoprotein A-I metabolism, Biological Transport drug effects, CD36 Antigens, Humans, Inhibitory Concentration 50, Lipoproteins, HDL metabolism, Protein Binding drug effects, Receptors, Immunologic metabolism, Receptors, Scavenger, Scavenger Receptors, Class B, ATP-Binding Cassette Transporters antagonists & inhibitors, Cholesterol metabolism, Glyburide pharmacology, Naphthalenes pharmacology, Receptors, Immunologic antagonists & inhibitors, Urea analogs & derivatives, Urea pharmacology
- Abstract
Scavenger receptor class B type I (SR-BI) and ABCA1 are structurally dissimilar cell surface proteins that play key roles in HDL metabolism. SR-BI is a receptor that binds HDL with high affinity and mediates both the selective lipid uptake of cholesteryl esters from lipid-rich HDL to cells and the efflux of unesterified cholesterol from cells to HDL. ABCA1 mediates the efflux of unesterified cholesterol and phospholipids from cells to lipid-poor apolipoprotein A-I (apoA-I). The activities of ABCA1 and other ATP binding cassette superfamily members are inhibited by the drug glyburide, and SR-BI-mediated lipid transport is blocked by small molecule inhibitors called BLTs. Here, we show that one BLT, [1-(2-methoxy-phenyl)-3-naphthalen-2-yl-urea] (BLT-4), blocked ABCA1-mediated cholesterol efflux to lipid-poor apoA-I at a potency similar to that for its inhibition of SR-BI (IC(50) approximately 55-60 microM). Reciprocally, glyburide blocked SR-BI-mediated selective lipid uptake and efflux at a potency similar to that for its inhibition of ABCA1 (IC(50) approximately 275-300 microM). As is the case with BLTs, glyburide increased the apparent affinity of HDL binding to SR-BI. The reciprocal inhibition of SR-BI and ABCA1 by BLT-4 and glyburide raises the possibility that these proteins may share similar or common steps in their mechanisms of lipid transport.
- Published
- 2004
- Full Text
- View/download PDF
24. Evidence that monastrol is an allosteric inhibitor of the mitotic kinesin Eg5.
- Author
-
Maliga Z, Kapoor TM, and Mitchison TJ
- Subjects
- Adenosine Diphosphate metabolism, Adenosine Triphosphate metabolism, Allosteric Regulation, Animals, Cells, Cultured, Chlorocebus aethiops, Enzyme Inhibitors chemistry, Enzyme Inhibitors metabolism, Fluorescent Dyes, Humans, Hydrolysis, Kidney cytology, Kidney metabolism, Kinetics, Microtubules physiology, Models, Biological, Protein Structure, Tertiary, Pyrimidines chemistry, Spectrometry, Fluorescence, Stereoisomerism, Thiones chemistry, Enzyme Inhibitors pharmacology, Kinesins antagonists & inhibitors, Pyrimidines pharmacology, Thiones pharmacology, Xenopus Proteins antagonists & inhibitors
- Abstract
Monastrol, a cell-permeable inhibitor of the kinesin Eg5, has been used to probe the dynamic organization of the mitotic spindle. The mechanism by which monastrol inhibits Eg5 function is unknown. We found that monastrol inhibits both the basal and the microtubule-stimulated ATPase activity of the Eg5 motor domain. Unlike many ATPase inhibitors, monastrol does not compete with ATP binding to Eg5. Monastrol appears to inhibit microtubule-stimulated ADP release from Eg5 but does not compete with microtubule binding, suggesting that monastrol binds a novel allosteric site in the motor domain. Finally, we established that (S)-monastrol, as compared to the (R)-enantiomer, is a more potent inhibitor of Eg5 activity in vitro and in vivo. Future structural studies should help in designing more potent Eg5 inhibitors for possible use as anticancer drugs and cell biological reagents.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.