Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cancer Res ; 83(12): 2052-2065, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37098922

ABSTRACT

Metastases are hard to detect and treat, and they cause most cancer-related deaths. The relative lack of therapies targeting metastases represents a major unmet clinical need. The extracellular matrix (ECM) forms a major component of the tumor microenvironment in both primary and metastatic tumors, and certain ECM proteins can be selectively and abundantly expressed in tumors. Nanobodies against ECM proteins that show selective abundance in metastases have the potential to be used as vehicles for delivery of imaging and therapeutic cargoes. Here, we describe a strategy to develop phage-display libraries of nanobodies against ECM proteins expressed in human metastases, using entire ECM-enriched preparations from triple-negative breast cancer (TNBC) and colorectal cancer metastases to different organs as immunogens. In parallel, LC-MS/MS-based proteomics were used to define a metastasis-associated ECM signature shared by metastases from TNBC and colorectal cancer, and this conserved set of ECM proteins was selectively elevated in other tumors. As proof of concept, selective and high-affinity nanobodies were isolated against an example protein from this signature, tenascin-C (TNC), known to be abundant in many tumor types and to play a role in metastasis. TNC was abundantly expressed in patient metastases and widely expressed across diverse metastatic sites originating from several primary tumor types. Immuno-PET/CT showed that anti-TNC nanobodies bind TNBC tumors and metastases with excellent specificity. We propose that such generic nanobodies against tumors and metastases are promising cancer-agnostic tools for delivery of therapeutics to tumor and metastatic ECM. SIGNIFICANCE: Nanobodies specific for extracellular matrix markers commonly expressed in primary tumors and metastases are promising agents for noninvasive detection of tumors and metastases and potential tools for targeted therapy.


Subject(s)
Colorectal Neoplasms , Single-Domain Antibodies , Triple Negative Breast Neoplasms , Humans , Proteomics/methods , Triple Negative Breast Neoplasms/pathology , Chromatography, Liquid , Tandem Mass Spectrometry , Extracellular Matrix/metabolism , Tenascin/metabolism , Colorectal Neoplasms/pathology , Tumor Microenvironment
2.
Nat Commun ; 13(1): 109, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013154

ABSTRACT

Direct injection of therapies into tumors has emerged as an administration route capable of achieving high local drug exposure and strong anti-tumor response. A diverse array of immune agonists ranging in size and target are under development as local immunotherapies. However, due to the relatively recent adoption of intratumoral administration, the pharmacokinetics of locally-injected biologics remains poorly defined, limiting rational design of tumor-localized immunotherapies. Here we define a pharmacokinetic framework for biologics injected intratumorally that can predict tumor exposure and effectiveness. We find empirically and computationally that extending the tumor exposure of locally-injected interleukin-2 by increasing molecular size and/or improving matrix-targeting affinity improves therapeutic efficacy in mice. By tracking the distribution of intratumorally-injected proteins using positron emission tomography, we observe size-dependent enhancement in tumor exposure occurs by slowing the rate of diffusive escape from the tumor and by increasing partitioning to an apparent viscous region of the tumor. In elucidating how molecular weight and matrix binding interplay to determine tumor exposure, our model can aid in the design of intratumoral therapies to exert maximal therapeutic effect.


Subject(s)
Collagen/genetics , Immunotherapy/methods , Interleukin-2/pharmacology , Melanoma, Experimental/therapy , Receptors, Immunologic/genetics , Skin Neoplasms/therapy , Allografts , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Line, Tumor , Collagen/immunology , Female , Gene Library , Injections, Intralesional , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2/pharmacokinetics , Melanoma, Experimental/diagnostic imaging , Melanoma, Experimental/genetics , Melanoma, Experimental/mortality , Mice , Mice, Inbred C57BL , Peptides/genetics , Peptides/immunology , Positron-Emission Tomography , Protein Binding , Protein Engineering/methods , Receptors, Immunologic/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Serum Albumin/genetics , Serum Albumin/immunology , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Survival Analysis , Tumor Burden/drug effects
3.
PNAS Nexus ; 1(5): pgac244, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36712341

ABSTRACT

Confining cytokine exposure to the tumors would greatly enhance cancer immunotherapy safety and efficacy. Immunocytokines, cytokines fused to tumor-targeting antibodies, have been developed with this intention, but without significant clinical success to date. A critical limitation is uptake by receptor-expressing cells in the blood, that decreases the dose at the tumor and engenders toxicity. Small-format immunocytokines, constructed with antibody fragments, are hypothesized to improve tumor specificity due to rapid systemic clearance. However, effective design criteria for small-format immunocytokines need further examination. Here, we engineer small interleukin-2 (IL-2) immunocytokines fused to nanobodies with nanomolar to picomolar affinities for the tumor-specific EIIIB domain of fibronectin (also known as EDB). Upon intravenous delivery into immunocompetent mice, such immunocytokines led to similar tumor growth delay as size-matched untargeted IL-2. Intratumoral (i.t.) delivery imparted improved survival dependent on affinity to EIIIB. I.t. administration offers a promising avenue to deliver small-format immunocytokines, given effective affinity for the tumor microenvironment.

4.
Arterioscler Thromb Vasc Biol ; 41(1): e18-e32, 2021 01.
Article in English | MEDLINE | ID: mdl-33207933

ABSTRACT

OBJECTIVE: Exposure of the arterial endothelium to low and disturbed flow is a risk factor for the erosion and rupture of atherosclerotic plaques and aneurysms. Circulating and locally produced proteins are known to contribute to an altered composition of the extracellular matrix at the site of lesions, and to contribute to inflammatory processes within the lesions. We have previously shown that alternative splicing of FN (fibronectin) protects against flow-induced hemorrhage. However, the impact of alternative splicing of FN on extracellular matrix composition remains unknown. Approach and Results: Here, we perform quantitative proteomic analysis of the matrisome of murine carotid arteries in mice deficient in the production of FN splice isoforms containing alternative exons EIIIA and EIIIB (FN-EIIIAB null) after exposure to low and disturbed flow in vivo. We also examine serum-derived and endothelial-cell contributions to the matrisome in a simplified in vitro system. We found flow-induced differences in the carotid artery matrisome that were impaired in FN-EIIIAB null mice. One of the most interesting differences was reduced recruitment of FBLN1 (fibulin-1), abundant in blood and not locally produced in the intima. This defect was validated in our in vitro assay, where FBLN1 recruitment from serum was impaired by the absence of these alternatively spliced segments. CONCLUSIONS: Our results reveal the extent of the dynamic alterations in the matrisome in the acute response to low and disturbed flow and show how changes in the splicing of FN, a common response in vascular inflammation and remodeling, can affect matrix composition.


Subject(s)
Alternative Splicing , Carotid Arteries/metabolism , Carotid Stenosis/metabolism , Extracellular Matrix/metabolism , Fibronectins/metabolism , Vascular Remodeling , Animals , Carotid Arteries/pathology , Carotid Arteries/physiopathology , Carotid Stenosis/pathology , Carotid Stenosis/physiopathology , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Extracellular Matrix/pathology , Fibronectins/deficiency , Fibronectins/genetics , Mechanotransduction, Cellular , Mice, Knockout , Protein Isoforms , Regional Blood Flow , Stress, Mechanical
5.
Proc Natl Acad Sci U S A ; 116(28): 14181-14190, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31068469

ABSTRACT

Extracellular matrix (ECM) deposition is a hallmark of many diseases, including cancer and fibroses. To exploit the ECM as an imaging and therapeutic target, we developed alpaca-derived libraries of "nanobodies" against disease-associated ECM proteins. We describe here one such nanobody, NJB2, specific for an alternatively spliced domain of fibronectin expressed in disease ECM and neovasculature. We showed by noninvasive in vivo immuno-PET/CT imaging that NJB2 detects primary tumors and metastatic sites with excellent specificity in multiple models of breast cancer, including human and mouse triple-negative breast cancer, and in melanoma. We also imaged mice with pancreatic ductal adenocarcinoma (PDAC) in which NJB2 was able to detect not only PDAC tumors but also early pancreatic lesions called pancreatic intraepithelial neoplasias, which are challenging to detect by any current imaging modalities, with excellent clarity and signal-to-noise ratios that outperformed conventional 2-fluorodeoxyglucose PET/CT imaging. NJB2 also detected pulmonary fibrosis in a bleomycin-induced fibrosis model. We propose NJB2 and similar anti-ECM nanobodies as powerful tools for noninvasive detection of tumors, metastatic lesions, and fibroses. Furthermore, the selective recognition of disease tissues makes NJB2 a promising candidate for nanobody-based therapeutic applications.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/diagnostic imaging , Extracellular Matrix/drug effects , Pancreatic Neoplasms/diagnostic imaging , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Fibrosis/pathology , Humans , Male , Mice , Pancreatic Neoplasms/pathology , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Radiopharmaceuticals/pharmacology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/pharmacology , Pancreatic Neoplasms
6.
Proc Natl Acad Sci U S A ; 116(16): 7624-7631, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30936321

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has been successful in clinical trials against hematological cancers, but has experienced challenges in the treatment of solid tumors. One of the main difficulties lies in a paucity of tumor-specific targets that can serve as CAR recognition domains. We therefore focused on developing VHH-based, single-domain antibody (nanobody) CAR T cells that target aspects of the tumor microenvironment conserved across multiple cancer types. Many solid tumors evade immune recognition through expression of checkpoint molecules, such as PD-L1, that down-regulate the immune response. We therefore targeted CAR T cells to the tumor microenvironment via the checkpoint inhibitor PD-L1 and observed a reduction in tumor growth, resulting in improved survival. CAR T cells that target the tumor stroma and vasculature through the EIIIB+ fibronectin splice variant, which is expressed by multiple tumor types and on neovasculature, are likewise effective in delaying tumor growth. VHH-based CAR T cells can thus function as antitumor agents for multiple targets in syngeneic, immunocompetent animal models. Our results demonstrate the flexibility of VHH-based CAR T cells and the potential of CAR T cells to target the tumor microenvironment and treat solid tumors.


Subject(s)
Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/metabolism , Single-Domain Antibodies/pharmacology , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents/pharmacology , Mice , Neoplasms, Experimental , Xenograft Model Antitumor Assays
7.
Data Brief ; 17: 604-609, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29552609

ABSTRACT

The Rb protein is a tumor suppressor protein that regulates the key G1S checkpoint consequently blocking the progression of cell cycle into S-phase. Despite its pertinent role in cell cycle regulation, comprehensive information on its interacting partners across cell cycle progression is lacking. Here, we aim to submit a comprehensive set of Rb interactors as the cell progresses from G0 through G1 and S into G2 phase in HEK 293 cell line. Affinity purification of HA-tagged Rb protein along with its interactors was analyzed by mass spectrometry (AP-MS). SILAC labeling enabled differentiation of Rb interactors in different cell cycle stages as well as their quantification - G0 cells were labeled with light labels of lysine and arginine (K0R0), cells in G1S transition were labeled with heavy labels (K8R10) while the G2 cells were labeled with medium labels (K6R6). LC-MS/MS analysis resulted in 6 wiff files which were submitted to protein pilot software for peptide identification and quantification. Here we submit the dataset which clearly captures the changing interacting partners of the Rb protein as the cell cycle progressed from G0 through G1S checkpoint into G2 phase. Data is publicly available via ProteomeXchange with identifier PXD007708.

8.
Sci Rep ; 8(1): 1303, 2018 01 22.
Article in English | MEDLINE | ID: mdl-29358593

ABSTRACT

Cell growth and proliferation are two diverse processes yet always linked. Akt1, a serine/threonine kinase, is a multi-functional protein implicated in regulation of cell growth, survival and proliferation. Though it has a role in G1/S progression, the manner by which Akt1 controls cell cycle and blends cell growth with proliferation is not well explored. In this study, we characterize the Akt1 interactome as the cell cycle progresses from G0 to G1/S and G2 phase. For this, Akt1-overexpressing HEK293 cells were subjected to AP-MS. To distinguish between individual cell cycle stages, cells were cultured in the light, medium and heavy labelled SILAC media. We obtained 213 interacting partners of Akt1 from these studies. GO classification revealed that a significant number of proteins fall into functional classes related to cell growth or cell cycle processes. Of these, 32 proteins showed varying association with Akt1 in different cell cycle stages. Further analyses uncovered a subset of proteins showing counteracting effects so as to tune stage-specific progression through the cycle. Thus, our study provides some novel perspectives on Akt1-mediated regulation of the cell cycle and offers the framework for a detailed resolution of the downstream cellular mechanisms that are mediated by this kinase.


Subject(s)
Cell Cycle , Protein Interaction Maps , Proto-Oncogene Proteins c-akt/metabolism , HEK293 Cells , Humans , Protein Binding , Proteome/metabolism
9.
Data Brief ; 13: 187-191, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28603765

ABSTRACT

Akt1 is a multi-functional protein, implicated in multiple human solid tumors. Pertaining to its key role in cell survival, Akt1 is under focus for development of targeted therapies. Functional diversity of Akt1 is a result of its interactions with other proteins; which changes with changing context. This investigation was designed to capture the dynamics of Akt1 Interactome as a function of its active state. Delineating dynamic changes in association of Akt1 with its interactors could help us comprehend how it changes as a function of inhibition of its active form. Similar information on changes in Akt1 interactome as of now is not well explored. Akt1 expressing HEK293 cells were cultured in light and heavy labeled SILAC media. Normal lysine and arginine were incorporated as light labels while for heavy labeling the isotopes were 8 and 10 Da heavier. Light labeled cells represented the indigenous state of Akt1 interactome while heavy labeled cells represented Akt1 interactome in presence of its allosteric inhibitor, MK-2206. Equal number of cells from both conditions were pooled, lysed and subjected to Affinity Purification coupled to Mass Spectroscopy (AP-MS). Additionally, SILAC labeling aided in quantitative estimation of changing association of a number of proteins which were common to the two experimental conditions, with Akt1. Data are available via ProteomeXchange with identifier PXD005976.

10.
Proc Natl Acad Sci U S A ; 114(28): E5625-E5634, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28652369

ABSTRACT

The extracellular microenvironment is an integral component of normal and diseased tissues that is poorly understood owing to its complexity. To investigate the contribution of the microenvironment to lung fibrosis and adenocarcinoma progression, two pathologies characterized by excessive stromal expansion, we used mouse models to characterize the extracellular matrix (ECM) composition of normal lung, fibrotic lung, lung tumors, and metastases. Using quantitative proteomics, we identified and assayed the abundance of 113 ECM proteins, which revealed robust ECM protein signatures unique to fibrosis, primary tumors, or metastases. These analyses indicated significantly increased abundance of several S100 proteins, including Fibronectin and Tenascin-C (Tnc), in primary lung tumors and associated lymph node metastases compared with normal tissue. We further showed that Tnc expression is repressed by the transcription factor Nkx2-1, a well-established suppressor of metastatic progression. We found that increasing the levels of Tnc, via CRISPR-mediated transcriptional activation of the endogenous gene, enhanced the metastatic dissemination of lung adenocarcinoma cells. Interrogation of human cancer gene expression data revealed that high TNC expression correlates with worse prognosis for lung adenocarcinoma, and that a three-gene expression signature comprising TNC, S100A10, and S100A11 is a robust predictor of patient survival independent of age, sex, smoking history, and mutational load. Our findings suggest that the poorly understood ECM composition of the fibrotic and tumor microenvironment is an underexplored source of diagnostic markers and potential therapeutic targets for cancer patients.


Subject(s)
Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Proteomics/methods , Tenascin/physiology , Adenocarcinoma/metabolism , Animals , Annexin A2/metabolism , CRISPR-Cas Systems , Disease Progression , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Male , Mice , Mice, Inbred C57BL , Multivariate Analysis , Neoplasm Metastasis , Prognosis , S100 Proteins/metabolism , Thyroid Nuclear Factor 1/metabolism , Treatment Outcome , Tumor Microenvironment
11.
Data Brief ; 11: 252-257, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28243621

ABSTRACT

Akt1 is a multi-functional protein implicated in key cellular processes including regulation of proliferation, survival, metabolism and protein synthesis. Its functional diversity results through interactions with other proteins which change with changing context. This study was designed to capture proteins, which interact with Akt1 as the cell cycle progresses from G0 to G1S and then G2 phase. Such an insight might help us understand the role of Akt1 in cell cycle, which as of now is not well explored. Akt1 expressing HEK 293 cells were cultured in light, medium and heavy labeled SILAC media. Normal lysine and arginine were incorporated as light labels; 6 Da (Dalton) heavier isotopes of the same amino acids were used as medium labels; while for heavy labeling the isotopes were 8 and 10 Da heavier. Light labeled cells were arrested in G0 phase while medium and heavy labeled cells were arrested in G2 and G1S phases, respectively. Equal number of cells from each phase was pooled, lysed and subjected to Affinity Purification coupled to Mass Spectroscopy (AP-MS). The obtained Akt1 protein partners were observed to change as the cell cycle progressed from G0 to G1S and then to G2 phase. Additionally, SILAC labeling aided in quantitative estimation of changing association of a number of proteins which were common to two or more phases, with Akt1. Data are available via ProteomeXchange with identifier PXD005557.

12.
Curr Opin Genet Dev ; 23(6): 649-57, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24287335

ABSTRACT

Classical 'one-gene/one-disease' models cannot fully reconcile with the increasingly appreciated prevalence of complicated genotype-to-phenotype associations in human disease. Genes and gene products function not in isolation but as components of intricate networks of macromolecules (DNA, RNA, or proteins) and metabolites linked through biochemical or physical interactions, represented in 'interactome' network models as 'nodes' and 'edges', respectively. Accordingly, mechanistic understanding of human disease will require understanding of how disease-causing mutations affect systems or interactome properties. The study of 'edgetics' uncovers specific loss or gain of interactions (edges) to interpret genotype-to-phenotype relationships. We review how distinct genetic variants, the genotype, lead to distinct phenotypic outcomes, the phenotype, through edgetic perturbations in interactome networks altogether representing the 'edgotype'.


Subject(s)
Gene Regulatory Networks , Genetic Association Studies/methods , Models, Genetic , Protein Interaction Domains and Motifs/genetics , Genetic Predisposition to Disease/genetics , Genotype , Humans , Metabolic Networks and Pathways/genetics , Mutation , Phenotype
13.
Genome Res ; 21(12): 2067-81, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21865350

ABSTRACT

Drug development efforts against cancer are often hampered by the complex properties of signaling networks. Here we combined the results of an RNAi screen targeting the cellular signaling machinery, with graph theoretical analysis to extract the core modules that process both mitogenic and oncogenic signals to drive cell cycle progression. These modules encapsulated mechanisms for coordinating seamless transition of cells through the individual cell cycle stages and, importantly, were functionally conserved across different cancer cell types. Further analysis also enabled extraction of the core signaling axes that progressively guide commitment of cells to the division cycle. Importantly, pharmacological targeting of the least redundant nodes in these axes yielded a synergistic disruption of the cell cycle in a tissue-type-independent manner. Thus, the core elements that regulate temporally distinct stages of the cell cycle provide attractive targets for the development of multi-module-based chemotherapeutic strategies.


Subject(s)
Cell Cycle , Models, Biological , Neoplasms/metabolism , Signal Transduction , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/therapy
14.
Anticancer Agents Med Chem ; 11(1): 64-77, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21214508

ABSTRACT

Coordinated coupling of biochemical reactions involving protein phosphorylation and dephosphorylation represents the hallmark of the intracellular signal transduction machinery. Distinct classes of enzymes known as kinases and phosphatases respectively drive these reactions. Alterations in activity of such signaling intermediates, either due to mutations in the corresponding genes or epigenetic modulation of their expression levels, is often the cause of many cancers. The role of kinases during signal transduction has been extensively investigated over the past several decades and the consensus view is that subsets of kinases form distinct cascades of signaling pathways. Further, the extensive crosstalk that exists between these cascades leads to a complex network configuration for the signaling machinery. Inhibitors of many of these kinases are now being exploited in cancer therapy. In contrast to this, regulation by cellular phosphatases has generally been considered to occur through isolated interactions between a given phosphatase and its target substrate. Emerging evidence, however, is beginning to suggest that phosphatases also inter-regulate each other, and that such interactions can lead to the formation of discrete phosphatase-specific cascades. A phosphatase cascade may be defined broadly as a series of successive dephosphorylation reactions that occur within a cell and are catalyzed by phosphatases which are activated sequentially. In general, the term phosphatase cascade refers to cascades that include two or more phosphatase members. The crosstalk between such regulatory axes of phosphatases and kinase cascades provides for complex modes of regulation, with non-linear signal input/output relationships. This review discusses the implications of such phosphatase-constituted regulatory elements for both signal processing and transmission. Further, we also explore the potential that insights on the functioning of phosphatase cascades offers, for the development of new and selective strategies for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Neoplasms/enzymology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Animals , Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Humans , Neoplasms/metabolism , Structure-Activity Relationship
15.
BMC Syst Biol ; 4: 169, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-21143896

ABSTRACT

BACKGROUND: Engagement of the antigen receptor on immature B-lymphocytes leads to cell cycle arrest, and subsequent apoptosis. This is an essential process for eliminating self reactive B cells during its different stages of development. However, the mechanism by which it is achieved is not completely understood. RESULTS: Here we employed a systems biology approach that combined extensive experimentation with in silico methodologies to chart the network of receptor-activated pathways that mediated the arrest of immature B cells in the G1 phase of the cell cycle. Interestingly, we found that only a sparse network of signaling intermediates was recruited upon engagement of the antigen receptor. This then led to the activation of a restricted subset of transcription factors, with the consequent induction of genes primarily involved in the cell death pathway. Subsequent experiments revealed that the weak initiation of intracellular signaling pathways derived from desensitization of the receptor-proximal protein tyrosine kinase Lyn, to receptor-dependent activation. Intriguingly, the desensitization was a result of the constitutive activation of this kinase in unstimulated cells, which was likely maintained through a regulatory feedback loop involving the p38 MAP kinase. The high basal activity then attenuated the ability of the antigen receptor to recruit Lyn, and thereby also the downstream signaling intermediates. Finally, integration of these results into a mathematical model provided further substantiation to the novel finding that the ground state of the intracellular signaling machinery constitutes an important determinant of the outcome of receptor-induced cellular responses. CONCLUSIONS: Our results identify the global events leading to the G1 arrest and subsequent apoptosis in immature B cells upon receptor activation.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Cycle , Receptors, Antigen, B-Cell/metabolism , Systems Biology/methods , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , B-Lymphocytes/drug effects , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Feedback, Physiological/drug effects , Gene Expression Profiling , Imidazoles/pharmacology , Mice , Pyridines/pharmacology , Receptors, Antigen, B-Cell/genetics , Signal Transduction/drug effects , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases/metabolism
16.
FEBS J ; 275(4): 739-52, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18190531

ABSTRACT

Spo0B is an important component of the phosphorelay signal transduction pathway, the pathway involved in the initiation of sporulation in Bacillus subtilis. Bioinformatic, phylogenetic and biochemical studies showed that Spo0B of Bacillus anthracis has evolved from citrate/malate kinases. During the course of evolution, Spo0B has retained the characteristic histidine kinase boxes H, N, F, G(1) and G(2), and has acquired nucleotide-binding domains, Walker A and Walker B, of ATPases. Owing to the presence of these domains, autophosphorylation and ATPase activity was observed in Spo0B of B. anthracis. Mutational studies showed that among the six histidine residues, His13 of the H-box is involved in the autophosphorylation activity of Spo0B, whereas Lys33 of the Walker A domain is associated with the ATPase activity of the protein. Thermodynamic and binding studies of the binding of Mg-ATP to Spo0B using isothermal titration calorimetry (ITC) suggested that the binding is driven by favorable entropy changes and that the reaction is exothermic, with an apparent dissociation constant (K(d)) equal to 0.02 mm. The value of the dissociation constant (K(d) = 0.05 mm) determined by the intrinsic fluorescence of trytophan of Spo0B was similar to that obtained by ITC studies. The purified Spo0B of B. anthracis also showed nucleoside diphosphate kinase-like activity of phosphate transfer from nucleoside triphosphate to nucleoside diphosphate. This is the first evidence for Spo0B of B. anthracis as an enzyme with histidine kinase and ATPase activities, which may have important roles to play in sporulation and pathogenesis.


Subject(s)
Bacillus anthracis/metabolism , Bacterial Proteins/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Bacillus anthracis/classification , Bacillus anthracis/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Dimerization , Molecular Sequence Data , Mutagenesis , Phosphorylation , Phylogeny , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...