Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
PLoS Genet ; 12(4): e1005895, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27093186

ABSTRACT

Small cell lung cancer (SCLC) is an aggressive disease with poor survival. A few sequencing studies performed on limited number of samples have revealed potential disease-driving genes in SCLC, however, much still remains unknown, particularly in the Asian patient population. Here we conducted whole exome sequencing (WES) and transcriptomic sequencing of primary tumors from 99 Chinese SCLC patients. Dysregulation of tumor suppressor genes TP53 and RB1 was observed in 82% and 62% of SCLC patients, respectively, and more than half of the SCLC patients (62%) harbored TP53 and RB1 mutation and/or copy number loss. Additionally, Serine/Arginine Splicing Factor 1 (SRSF1) DNA copy number gain and mRNA over-expression was strongly associated with poor survival using both discovery and validation patient cohorts. Functional studies in vitro and in vivo demonstrate that SRSF1 is important for tumorigenicity of SCLC and may play a key role in DNA repair and chemo-sensitivity. These results strongly support SRSF1 as a prognostic biomarker in SCLC and provide a rationale for personalized therapy in SCLC.


Subject(s)
Carcinoma, Small Cell/genetics , Lung Neoplasms/genetics , Oncogene Proteins/genetics , Serine-Arginine Splicing Factors/genetics , Adult , Aged , DNA Copy Number Variations , DNA Damage , Female , Gene Silencing , Humans , Male , Middle Aged , Mutation
2.
Arthritis Rheumatol ; 68(2): 473-83, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26414805

ABSTRACT

OBJECTIVE: Systemic sclerosis (SSc) is a fibrotic disease characterized by an obliterative vasculopathy with thrombosis and impairment of the coagulation-fibrinolysis balance. Plasminogen activator inhibitor 1 (PAI-1) is the major inhibitor of profibrinolytic plasminogen activators (PAs). This study was undertaken to evaluate the contribution of PAI-1 to SSc pathology in the skin. METHODS: PAI-1 was evaluated in skin from patients with diffuse SSc (dSSc) and those with limited SSc (lSSc) by immunohistochemistry. The contribution of PAI-1 to SSc pathology was tested in vivo in murine graft-versus-host disease (GVHD) and bleomycin models of progressive skin fibrosis and in vitro in dermal human microvascular endothelial cells (HMVECs) using a monoclonal antibody that selectively prevents the binding of PAI-1 to PA. RESULTS: Skin from patients with dSSc and those with lSSc showed increased PAI-1 levels in the epidermis and microvessel endothelium. PAI-1 neutralization in the GVHD model led to a dramatic, dose-dependent improvement in clinical skin score, concomitant with vasculopathy resolution, including a reduction in fibrinolysis regulators and vascular injury markers, as well as reduced inflammation. Resolution of vasculopathy and inflammation was associated with resolution of skin fibrosis, as assessed by reduction in collagen content and expression of key profibrotic mediators, including transforming growth factor ß1 and tissue inhibitor of metalloproteinases 1. Similar to the GVHD model, PAI-1 neutralization reduced dermal inflammation and fibrosis in the bleomycin model. PAI-1 neutralization stimulated plasmin-mediated metalloproteinase 1 activation in dermal HMVECs. CONCLUSION: Our findings indicate that neutralization of the antifibrinolytic function of PAI-1 resolves skin fibrosis by limiting the extent of initial vascular injury and connective tissue inflammation. These data suggest that PAI-1 represents an important checkpoint in disease pathology in human SSc.


Subject(s)
Endothelial Cells/metabolism , Graft vs Host Disease/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Scleroderma, Diffuse/metabolism , Scleroderma, Limited/metabolism , Skin/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Bleomycin/toxicity , Case-Control Studies , Disease Models, Animal , Endothelial Cells/drug effects , Fibrosis/chemically induced , Fibrosis/metabolism , Humans , Immunohistochemistry , Matrix Metalloproteinase 1/metabolism , Mice , Plasminogen Activators/metabolism , Real-Time Polymerase Chain Reaction , Skin/drug effects , Skin/pathology
3.
PLoS One ; 10(2): e0115828, 2015.
Article in English | MEDLINE | ID: mdl-25706559

ABSTRACT

Release of endogenous damage associated molecular patterns (DAMPs), including members of the S100 family, are associated with infection, cellular stress, tissue damage and cancer. The extracellular functions of this family of calcium binding proteins, particularly S100A8, S100A9 and S100A12, are being delineated. They appear to mediate their functions via receptor for advanced glycation endproducts (RAGE) or TLR4, but there remains considerable uncertainty over the relative physiological roles of these DAMPs and their pattern recognition receptors. In this study, we surveyed the capacity of S100 proteins to induce proinflammatory cytokines and cell migration, and the contribution RAGE and TLR4 to mediate these responses in vitro. Using adenoviral delivery of murine S100A9, we also examined the potential for S100A9 homodimers to trigger lung inflammation in vivo. S100A8, S100A9 and S100A12, but not the S100A8/A9 heterodimer, induced modest levels of TLR4-mediated cytokine production from human PBMC. In contrast, for most S100s including S100A9, RAGE blockade inhibited S100-mediated cell migration of THP1 cells and major leukocyte populations, whereas TLR4-blockade had no effect. Intranasal administration of murine S100A9 adenovirus induced a specific, time-dependent predominately macrophage infiltration that coincided with elevated S100A9 levels and proinflammatory cytokines in the BAL fluid. Inflammatory cytokines were markedly ablated in the TLR4-defective mice, but unexpectedly the loss of TLR4 signaling or RAGE-deficiency did not appreciably impact the S100A9-mediated lung pathology or the inflammatory cell infiltrate in the alveolar space. These data demonstrate that physiological levels of S100A9 homodimers can trigger an inflammatory response in vivo, and despite the capacity of RAGE and TLR4 blockade to inhibit responses in vitro, the response is predominately independent of both these receptors.


Subject(s)
Calgranulin B/pharmacology , Cell Movement/physiology , Signal Transduction/drug effects , Animals , Cell Line , Cell Movement/drug effects , Humans , Inflammation/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mice , Mice, Knockout , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Toll-Like Receptor 4/metabolism
4.
Arthritis Rheumatol ; 66(4): 1022-33, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24757153

ABSTRACT

OBJECTIVE: The molecular basis of inflammatory myopathies such as dermatomyositis (DM), polymyositis, and inclusion body myositis, which share the characteristics of chronic muscle inflammation and skeletal muscle wasting, are poorly understood. As such, effective targeted treatments for these diseases are lacking, resulting in critical unmet medical needs for these devastating diseases. The purpose of this study was to identify possible new targets for drug development by exploring the mechanism by which inflammation may play a role in the pathology of the inflammatory myopathies. METHODS: We compared expression levels of inflammatory cytokines and microRNAs (miRNAs) between muscle biopsy samples from patients with inflammatory myopathies and those from donors without myositis. In vitro human and mouse model systems were then used to characterize the role of these cytokines and microRNAs on myoblast-to-myocyte differentiation. RESULTS: We observed increased expression of inflammatory cytokines, including tumor necrosis factor α (TNFα), interferon-α (IFNα), IFNß, and interleukin-1ß, in different subtypes of inflammatory myopathies. We observed decreased expression of microRNA-1 (miR-1), miR-133a, and miR-133b in all of the inflammatory myopathy subtypes we evaluated, as well as decreased expression of miR-206 in DM; these miRNAs are essential for adult skeletal muscle differentiation and maintenance. TNFα was significantly inversely correlated with decreased myogenic miRNA expression in the inflammatory myopathy subtypes. In mechanistic studies, TNFα inhibited the expression of myogenic miRNAs and suppressed the differentiation of C2C12 myoblasts to myocytes/myotubes in an NF-κB-dependent manner. This block in differentiation by TNFα was relieved by overexpression of miR-1, miR-206, or miR-133a/b. CONCLUSION: Taken together, these results provide a new mechanistic link between the action of proinflammatory cytokines and the degenerative pathology of inflammatory myopathies, and suggest therapeutic approaches for these diseases.


Subject(s)
Cytokines/metabolism , MicroRNAs/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Myositis/metabolism , Adult , Animals , Cell Differentiation/physiology , Cell Line , Disease Models, Animal , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Mice , MicroRNAs/genetics , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Myositis/genetics , Myositis/pathology , NF-kappa B/metabolism
5.
Cancer Genet ; 206(9-10): 330-9, 2013.
Article in English | MEDLINE | ID: mdl-24200637

ABSTRACT

Intratumor heterogeneity can confound the results of mutation analyses in oncodriver genes using traditional methods thereby challenging the application of targeted cancer therapy strategies for patients Ultradeep sequencing can detect low frequency and expanded clonal mutations in primary tumors to better inform treatment decisions. KRAS coding exons in 61 treatment-naive colorectal cancer (CRC) tumors and KRAS, EGFR, ALK, and MET in lung tumors from three Chinese non-small cell lung cancer (NSCLC) patients were sequenced using ultradeep sequencing methods. Forty-one percent of CRC patients (25/61) harbored mutations in the KRAS active domain, eight of which (13%) were not detected by Sanger sequencing. Three (of eight) had frequencies less than 10% and one patient harbored more than one mutation. Low frequency KRAS active (G12R) and EGFR kinase domain mutations (G719A) were identified in one NSCLC patient. A second NSCLC patient showed an EML4-ALK fusion with ALK, EGFR, and MET mutations. A third NSCLC patient harbored multiple low frequency mutations in KRAS, EGFR, and MET as well as ALK gene copy number increases. Within the same patient, multiple low frequency mutations occurred within a gene. A complex pattern of intrinsic low frequency driver mutations in well-known tumor oncogenes may exist prior to treatment, resulting in resistance to targeted therapies. Ultradeep sequencing can characterize intratumor heterogeneity and identify such mutations to ultimately affect treatment decisions.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Colorectal Neoplasms/genetics , Lung Neoplasms/genetics , Mutation , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Anaplastic Lymphoma Kinase , Cell Line, Tumor , Child , DNA Mutational Analysis , ErbB Receptors/genetics , Female , Gene Frequency , HCT116 Cells , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins p21(ras) , Receptor Protein-Tyrosine Kinases/genetics , Young Adult
6.
Int J Oncol ; 40(5): 1321-30, 2012 May.
Article in English | MEDLINE | ID: mdl-22327175

ABSTRACT

Angiopoietin 2 (Ang2) is an important regulator of angiogenesis, blood vessel maturation and integrity of the vascular endothelium. The correlation between the dynamic expression of Ang2 in tumors with regions of high angiogenic activity and a poor prognosis in many tumor types makes Ang2 an ideal drug target. We have generated MEDI3617, a human anti-Ang2 monoclonal antibody that neutralizes Ang2 by preventing its binding to the Tie2 receptor in vitro, and inhibits angiogenesis and tumor growth in vivo. Treatment of mice with MEDI3617 resulted in inhibition of angiogenesis in several mouse models including: FGF2-induced angiogenesis in a basement extract plug model, tumor and retinal angiogenesis. In xenograft tumor models, treatment with MEDI3617 resulted in a reduction in tumor angiogenesis and an increase in tumor hypoxia. The administration of MEDI3617 as a single agent to mice bearing human tumor xenografts resulted in tumor growth inhibition against a broad spectrum of tumor types. Combining MEDI3617 with chemotherapy or bevacizumab resulted in a delay in tumor growth and no body weight loss was observed in the combination groups. These results, combined with pharmacodynamic studies, demonstrate that treatment of tumor-bearing mice with MEDI3617 significantly inhibited tumor growth as a single agent by blocking tumor angiogenesis. Together, these data show that MEDI3617 is a robust antiangiogenic agent and support the clinical evaluation and biomarker development of MEDI3617 in cancer patients.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Angiopoietin-2/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Angiogenesis Inhibitors/administration & dosage , Angiopoietin-2/immunology , Angiopoietin-2/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Bevacizumab , Cell Line, Tumor , Corrosion Casting , Dose-Response Relationship, Drug , Female , Fluorescence , HEK293 Cells , Humans , Mice , Mice, Nude , Neoplasms/blood supply , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/drug effects , Paclitaxel/administration & dosage , Phosphorylation , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Retinal Neovascularization/immunology , Retinal Neovascularization/metabolism , Retinal Neovascularization/prevention & control , Time Factors , Transfection , Tumor Burden/drug effects , X-Ray Microtomography , Xenograft Model Antitumor Assays
7.
Infect Immun ; 80(3): 975-81, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22184421

ABSTRACT

Acetic acid bacteria were previously considered nonpathogenic in humans. However, over the past decade, five genera of Acetobacteraceae have been isolated from patients with inborn or iatrogenic immunodeficiencies. Here, we describe the first studies of the interactions of the human innate immune system with a member of this bacterial family, Granulibacter bethesdensis, an emerging pathogen in patients with chronic granulomatous disease (CGD). Efficient phagocytosis of G. bethesdensis by normal and CGD polymorphonuclear leukocytes (CGD PMN) required heat-labile serum components (e.g., C3), and binding of C3 and C9 to G. bethesdensis was detected by immunoblotting. However, this organism survived in human serum concentrations of ≥90%, indicating a high degree of serum resistance. Consistent with the clinical host tropism of G. bethesdensis, CGD PMN were unable to kill this organism, while normal PMN, in the presence of serum, reduced the number of CFU by about 50% after a 24-h coculture. This finding, together with the observations that G. bethesdensis was sensitive to H(2)O(2) but resistant to LL-37, a human cationic antimicrobial peptide, suggests an inherent resistance to O(2)-independent killing. Interestingly, 10 to 100 times greater numbers of G. bethesdensis were required to achieve the same level of reactive oxygen species (ROS) production induced by Escherichia coli in normal PMN. In addition to the relative inability of the organism to elicit production of PMN ROS, G. bethesdensis inhibited both constitutive and FAS-induced PMN apoptosis. These properties of reduced PMN activation and resistance to nonoxidative killing mechanisms likely play an important role in G. bethesdensis pathogenesis.


Subject(s)
Acetobacteraceae/immunology , Acetobacteraceae/pathogenicity , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Granulomatous Disease, Chronic/immunology , Granulomatous Disease, Chronic/microbiology , Immunity, Innate , Blood Bactericidal Activity , Colony Count, Microbial , Complement System Proteins/immunology , Escherichia coli/immunology , Humans , Microbial Viability , Neutrophils/immunology , Phagocytosis , Reactive Oxygen Species/metabolism
8.
PLoS One ; 6(10): e26177, 2011.
Article in English | MEDLINE | ID: mdl-22046260

ABSTRACT

PURPOSE: Insulin-like growth factor (IGF) signaling through human insulin receptor isoform A (IR-A) contributes to tumorigenesis and intrinsic resistance to anti-IGF1R therapy. In the present study, we (a) developed quantitative TaqMan real time-PCR-based assays (qRT-PCR) to measure human insulin receptor isoforms with high specificity, (b) evaluated isoform expression levels in molecularly-defined breast cancer subtypes, and (c) identified the IR-A:IR-B mRNA ratio as a potential biomarker guiding patient stratification for anti-IGF therapies. EXPERIMENTAL DESIGN: mRNA expression levels of IR-A and IR-B were measured in 42 primary breast cancers and 19 matched adjacent normal tissues with TaqMan qRT-PCR assays. The results were further confirmed in 165 breast cancers. The tumor samples were profiled using whole genome microarrays and subsequently subtyped using the PAM50 breast cancer gene signature. The relationship between the IR-A:IR-B ratio and cancer subtype, as well as markers of proliferation were characterized. RESULTS: The mRNA expression levels of IR-A in the breast tumors were similar to those observed in the adjacent normal tissues, while the mRNA levels of IR-B were significantly decreased in tumors. The IR-A:IR-B ratio was significantly higher in luminal B breast cancer than in luminal A. Strong concordance between the IR-A:IR-B ratio and the composite Oncotype DX proliferation score was observed for stratifying the latter two breast cancer subtypes. CONCLUSIONS: The reduction in IR-B expression is the key to the altered IR-A:IR-B ratio observed in breast cancer. The IR-A:IR-B ratio may have biomarker utility in guiding a patient stratification strategy for an anti-IGF therapeutic.


Subject(s)
Antigens, CD/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Receptor, Insulin/genetics , Antigens, CD/analysis , Biomarkers, Tumor/analysis , Cell Proliferation , Female , Humans , Insulin-Like Growth Factor I/metabolism , Oligonucleotide Array Sequence Analysis , Protein Isoforms/analysis , Protein Isoforms/genetics , RNA, Messenger/analysis , Real-Time Polymerase Chain Reaction , Receptor, Insulin/analysis
9.
Infect Immun ; 77(10): 4337-44, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19635825

ABSTRACT

Chronic granulomatous disease (CGD) patients are susceptible to life-threatening infections by the Burkholderia cepacia complex. We used leukocytes from CGD and healthy donors and compared cell association, invasion, and cytokine induction by Burkholderia multivorans strains. A CGD isolate, CGD1, showed higher cell association than that of an environmental isolate, Env1, which correlated with cell entry. All B. multivorans strains associated significantly more with cells from CGD patients than with those from healthy donors. Similar findings were observed with another CGD pathogen, Serratia marcescens, but not with Escherichia coli. In a mouse model of CGD, strain CGD1 was virulent while Env1 was avirulent. B. multivorans organisms were found in the spleens of CGD1-infected mice at levels that were 1,000 times higher than those found in Env1-infected mice, which was coincident with higher levels of the proinflammatory cytokine interleukin-1beta. Taken together, these results may shed light on the unique susceptibility of CGD patients to specific pathogens.


Subject(s)
Burkholderia/immunology , Burkholderia/pathogenicity , Granulomatous Disease, Chronic/immunology , Granulomatous Disease, Chronic/microbiology , Host-Pathogen Interactions , Animals , Bacterial Adhesion , Burkholderia Infections/immunology , Burkholderia Infections/microbiology , Burkholderia Infections/pathology , Cells, Cultured , Colony Count, Microbial , Cytokines/metabolism , Escherichia coli/immunology , Escherichia coli/pathogenicity , Humans , Leukocytes, Mononuclear/immunology , Mice , Serratia marcescens/immunology , Serratia marcescens/pathogenicity , Spleen/microbiology , Virulence
10.
J Immunol ; 181(6): 4004-9, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18768855

ABSTRACT

We have devised an ex vivo culture system which generates large numbers of eosinophils at high purity (>90%) from unselected mouse bone marrow progenitors. In response to 4 days of culture with recombinant mouse FLT3-L and recombinant mouse stem cell factor followed by recombinant mouse IL-5 alone thereafter, the resulting bone marrow-derived eosinophils (bmEos) express immunoreactive major basic protein, Siglec F, IL-5R alpha-chain, and transcripts encoding mouse eosinophil peroxidase, CCR3, the IL-3/IL-5/GM-CSF receptor common beta-chain, and the transcription factor GATA-1. BmEos are functionally competent: they undergo chemotaxis toward mouse eotaxin-1 and produce characteristic cytokines, including IFN-gamma, IL-4, MIP-1alpha, and IL-6. The rodent pathogen pneumonia virus of mice replicates in bmEos and elevated levels of IL-6 are detected in supernatants of bmEos cultures in response to active infection. Finally, differentiating bmEos are readily transfected with lentiviral vectors, suggesting a means for rapid production of genetically manipulated cells.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Differentiation/immunology , Eosinophils/cytology , Eosinophils/immunology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/virology , Cell Communication/immunology , Cell Separation , Cells, Cultured , Chemotaxis, Leukocyte/immunology , Cytokines/metabolism , Cytokines/physiology , Eosinophils/metabolism , Eosinophils/virology , Mice , Mice, Inbred BALB C , Murine pneumonia virus/immunology , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , Stem Cells/virology
11.
Eukaryot Cell ; 7(2): 258-67, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18065654

ABSTRACT

S-adenosylmethionine (AdoMet) synthetase (EC 2.5.1.6) is the enzyme that catalyzes the synthesis of AdoMet, a molecule important for all cellular organisms. We have cloned and characterized an AdoMet synthetase gene (sam1) from Pneumocystis spp. This gene was transcribed primarily as an approximately 1.3-kb mRNA which encodes a protein containing 381 amino acids in P. carinii or P. murina and 382 amino acids in P. jirovecii. sam1 was also transcribed as part of an apparent polycistronic transcript of approximately 5.6 kb, together with a putative chromatin remodeling protein homologous to Saccharomyces cerevisiae, CHD1. Recombinant Sam1, when expressed in Escherichia coli, showed functional enzyme activity. Immunoprecipitation and confocal immunofluorescence analysis using an antipeptide antibody showed that this enzyme is expressed in P. murina. Thus, Pneumocystis, like other organisms, can synthesize its own AdoMet and may not depend on its host for the supply of this important molecule.


Subject(s)
Gene Expression Regulation, Fungal , Methionine Adenosyltransferase/genetics , Methionine Adenosyltransferase/metabolism , Pneumocystis carinii/enzymology , Amino Acid Sequence , Base Sequence , Blotting, Northern , Blotting, Southern , Cloning, Molecular , Immunoprecipitation , Molecular Sequence Data , Pneumocystis carinii/genetics , RNA, Messenger , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid
12.
BMC Biochem ; 8: 18, 2007 Sep 19.
Article in English | MEDLINE | ID: mdl-17880698

ABSTRACT

BACKGROUND: Heterotrimeric guanine nucleotide-binding regulatory proteins (G proteins), composed of G alpha, G beta, and G gamma subunits, are positioned at the inner face of the plasma membrane and relay signals from activated G protein-coupled cell surface receptors to various signaling pathways. G beta 5 is the most structurally divergent G beta isoform and forms tight heterodimers with regulator of G protein signalling (RGS) proteins of the R7 subfamily (R7-RGS). The subcellular localization of G beta 5/R7-RGS protein complexes is regulated by the palmitoylation status of the associated R7-binding protein (R7BP), a recently discovered SNARE-like protein. We investigate here whether R7BP controls the targeting of G beta 5/R7-RGS complexes to lipid rafts, cholesterol-rich membrane microdomains where conventional heterotrimeric G proteins and some effector proteins are concentrated in neurons and brain. RESULTS: We show that endogenous G beta 5/R7-RGS/R7BP protein complexes are present in native neuron-like PC12 cells and that a fraction is targeted to low-density, detergent-resistant membrane lipid rafts. The buoyant density of endogenous raft-associated G beta 5/R7-RGS protein complexes in PC12 cells was similar to that of lipid rafts containing the palmitoylated marker proteins PSD-95 and LAT, but distinct from that of the membrane microdomain where flotillin was localized. Overexpression of wild-type R7BP, but not its palmitoylation-deficient mutant, greatly enriched the fraction of endogenous G beta 5/R7-RGS protein complexes in the lipid rafts. In HEK-293 cells the palmitoylation status of R7BP also regulated the lipid raft targeting of co-expressed G beta 5/R7-RGS/R7BP proteins. A fraction of endogenous G beta 5/R7-RGS/R7BP complexes was also present in lipid rafts in mouse brain. CONCLUSION: A fraction of G beta 5/R7-RGS/R7BP protein complexes is targeted to low-density, detergent-resistant membrane lipid rafts in PC12 cells and brain. In cultured cells, the palmitoylation status of R7BP regulated the lipid raft targeting of endogenous or co-expressed G beta 5/R7-RGS proteins. Taken together with recent evidence that the kinetic effects of the G beta 5 complex on GPCR signaling are greatly enhanced by R7BP palmitoylation through a membrane-anchoring mechanism, our data suggest the targeting of the G beta 5/R7-RGS/R7BP complex to lipid rafts in neurons and brain, where G proteins and their effectors are concentrated, may be central to the G protein regulatory function of the complex.


Subject(s)
Brain/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Membrane Microdomains/metabolism , Neurons/metabolism , RGS Proteins/metabolism , Signal Transduction , Animals , Kinetics , Mice , PC12 Cells , Palmitic Acid/metabolism , Protein Binding , Rats
14.
J Immunol ; 179(3): 1693-9, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17641035

ABSTRACT

A critical role for eosinophils in remodeling of allergic airways was observed in vivo upon disruption of the dblGATA enhancer that regulates expression of GATA-1, which resulted in an eosinophil-deficient phenotype in the DeltadblGATA mouse. We demonstrate here that bone marrow progenitors isolated from DeltadblGATA mice can differentiate into mature eosinophils when subjected to cytokine stimulation ex vivo. Cultured DeltadblGATA eosinophils contain cytoplasmic granules with immunoreactive major basic protein and they express surface Siglec F and transcripts encoding major basic protein, eosinophil peroxidase, and GATA-1, -2, and -3 to an extent indistinguishable from cultured wild-type eosinophils. Fibroblast coculture and bone marrow cross-transplant experiments indicate that the in vivo eosinophil deficit is an intrinsic progenitor defect, and remains unaffected by interactions with stromal cells. Interestingly, and in contrast to those from the wild type, a majority of the GATA-1 transcripts from cultured DeltadblGATA progenitors express a variant GATA-1 transcript that includes a first exon (1E(B)), located approximately 3700 bp downstream to the previously described first exon found in hemopoietic cells (1E(A)) and approximately 42 bp upstream to another variant first exon, 1E(C). These data suggest that cultured progenitors are able to circumvent the effects of the DeltadblGATA ablation by using a second, more proximal, promoter and use this mechanism to generate quantities of GATA-1 that will support eosinophil growth and differentiation.


Subject(s)
Bone Marrow Cells/immunology , Cell Differentiation/genetics , Cell Lineage/genetics , Enhancer Elements, Genetic , Eosinophils/metabolism , GATA1 Transcription Factor/genetics , Hematopoietic Stem Cells/immunology , Promoter Regions, Genetic/immunology , Animals , Binding Sites/genetics , Binding Sites/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/immunology , Cell Lineage/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Eosinophils/cytology , Eosinophils/immunology , Eosinophils/transplantation , GATA1 Transcription Factor/physiology , GATA2 Transcription Factor/genetics , GATA3 Transcription Factor/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Interleukin-5/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Th2 Cells/immunology
15.
J Virol ; 81(11): 5594-606, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17376901

ABSTRACT

Simian immunodeficiency virus (SIV) infection of macaques and human immunodeficiency virus type 1 (HIV-1) infection of humans result in variable but generally fatal disease outcomes. Most SIV-infected macaques progress to AIDS over a period of 1 to 3 years, in the face of robust SIV-specific immune responses (conventional progressors [CP]). A small number of SIV-inoculated macaques mount transient immune responses and progress rapidly to AIDS (rapid progressors [RP]). We speculated that the underlying pathogenic mechanisms may differ between RP and CP macaques. We compared the pathological lesions, virus loads, and distribution of virus and target cells in SIVsmE660- or SIVsmE543-infected RP and CP rhesus macaques at terminal disease. RP macaques developed a wasting syndrome characterized by severe SIV enteropathy in the absence of opportunistic infections. In contrast, opportunistic infections were commonly observed in CP macaques. RP and CP macaques showed distinct patterns of CD4(+) T-cell depletion, with a selective loss of memory cells in RP macaques and a generalized (naive and memory) CD4 depletion in CP macaques. In situ hybridization demonstrated higher levels of virus expression in lymphoid tissues (P < 0.001) of RP macaques and a broader distribution to include many nonlymphoid tissues. Finally, SIV was preferentially expressed in macrophages in RP macaques whereas the primary target cells in CP macaques were T lymphocytes at end stage disease. These data suggest distinct pathogenic mechanisms leading to the deaths of these two groups of animals, with CP macaques being more representative of HIV-induced AIDS in humans.


Subject(s)
Acquired Immunodeficiency Syndrome/pathology , Acquired Immunodeficiency Syndrome/virology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Animals , Disease Progression , HIV-1/pathogenicity , Humans , Macaca mulatta , Simian Immunodeficiency Virus/pathogenicity
16.
Mol Cancer Res ; 5(2): 183-93, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17314275

ABSTRACT

Parafibromin is a tumor suppressor protein encoded by HRPT2, a gene recently implicated in the hereditary hyperparathyroidism-jaw tumor syndrome, parathyroid cancer, and a subset of kindreds with familial isolated hyperparathyroidism. Human parafibromin binds to RNA polymerase II as part of a PAF1 transcriptional regulatory complex. The mechanism by which loss of parafibromin function can lead to neoplastic transformation is poorly understood. Because the subcellular localization of parafibromin is likely to be critical for its function with the nuclear PAF1 complex, we sought to experimentally define the nuclear localization signal (NLS) of parafibromin and examine its potential role in parafibromin function. Using site-directed mutagenesis, we define a dominant bipartite NLS and a secondary NLS, both in the NH(2)-terminal region of parafibromin whose combined mutation nearly abolishes nuclear targeting. The NLS-mutant parafibromin is significantly impaired in its association with endogenous Paf1 and Leo1. We further report that overexpression of wild-type but not NLS-mutant parafibromin induces apoptosis in transfected cells. Inhibition of endogenous parafibromin expression by RNA interference inhibits the basal rate of apoptosis and apoptosis resulting from DNA damage induced by camptothecin, a topoisomerase I inhibitor. These experiments identify for the first time a proapoptotic activity of endogenous parafibromin likely to be important in its role as a tumor suppressor and show a functional role for the NLS of parafibromin in this activity.


Subject(s)
Cell Nucleus/metabolism , Hyperparathyroidism/metabolism , Jaw Neoplasms/metabolism , Nuclear Localization Signals/genetics , Tumor Suppressor Proteins/genetics , Amino Acid Sequence , Animals , Apoptosis , Cell Line, Tumor , Humans , Hyperparathyroidism/genetics , Jaw Neoplasms/genetics , Mice , Molecular Sequence Data , Transfection , Tumor Suppressor Proteins/metabolism
17.
J Biol Chem ; 281(35): 25622-34, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16803891

ABSTRACT

In this study, we explore the evolution and function of two closely related RNase A ribonucleases from the chicken, Gallus gallus. Separated by approximately 10 kb on chromosome 6, the coding sequences of RNases A-1 and A-2 are diverging under positive selection pressure (dN > dS) but remain similar to one another (81% amino acid identity) and to the mammalian angiogenins. Immunoreactive RNases A-1 and A-2 (both approximately 16 kDa) were detected in peripheral blood granulocytes and bone marrow. Recombinant proteins are ribonucleolytically active (kcat = 2.6 and 0.056 s(-1), respectively), and surprisingly, both interact with human placental ribonuclease inhibitor. RNase A-2, the more cationic (pI 11.0), is both angiogenic and bactericidal; RNase A-1 (pI 10.2) has neither activity. We demonstrated via point mutation of the catalytic His110 that ablation of ribonuclease activity has no impact on the bactericidal activity of RNase A-2. We determined that the divergent domains II (amino acids 71-76) and III (amino acids 89-104) of RNase A-2 are both important for bactericidal activity. Furthermore, we demonstrated that these cationic domains can function as independent bactericidal peptides without the tertiary structure imposed by the RNase A backbone. These results suggest that ribonucleolytic activity may not be a crucial constraint limiting the ongoing evolution of this gene family and that the ribonuclease backbone may be merely serving as a scaffold to support the evolution of novel, nonribonucleolytic proteins.


Subject(s)
Leukocytes/enzymology , Ribonuclease, Pancreatic/chemistry , Amino Acid Sequence , Animals , Bone Marrow Cells/metabolism , Chickens , Evolution, Molecular , Humans , Kinetics , Leukocytes/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Phylogeny , Protein Structure, Tertiary , Sequence Homology, Amino Acid
18.
Exp Hematol ; 33(1): 73-84, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15661400

ABSTRACT

OBJECTIVE: Activated platelets participate in inflammatory and microbicidal processes by upregulation of surface selectins, shedding of CD40 ligand, and release of platelet microbicidal proteins and microparticles. Given their myeloid lineage, we hypothesized that platelets express functional N-formyl peptide receptors and respond to the bacterially derived chemotactic peptide N-formyl peptide with gradient-driven chemotaxis. METHODS AND RESULTS: Here we show specific binding of N-formyl peptides to the surface of activated platelets. Platelet expression and function of the formyl peptide receptor, FPR, was verified by RT-PCR of the differentiated megakaryocyte MEG-01 cell line, immunoblotting of platelet proteins, and calcium mobilization in platelets with formyl peptide binding. Furthermore, we demonstrate gradient-driven chemotaxis of platelets by video microscopy and transwell migration toward formyl peptides. We also show that endogenous formyl peptides, released by eukaryotic mitochondria from necrotic cells, induce chemotaxis using formyl peptide receptors expressed by thrombin-activated platelets. Conversely, supernatants from cells undergoing apoptotic cell death do not induce platelet chemotaxis. Platelet chemotaxis to formyl peptides was blocked with FPR-specific antibody as well as by pertussis toxin inhibition of the formyl peptide G-coupled receptor. CONCLUSION: These data establish a new role for platelets in host defense and suggest reexamination of their active function in microbicidal and other host defense activities.


Subject(s)
Blood Platelets/physiology , Chemotaxis , Receptors, Formyl Peptide/physiology , Aorta , Blood Platelets/metabolism , Calcium Signaling , Cell Line , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Humans , Microscopy, Video , Mitochondria/metabolism , Necrosis , Protein Binding , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism
19.
Exp Hematol ; 32(2): 135-9, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15102473

ABSTRACT

The danger model of immunity and tolerance holds that antigen-presenting cells (APCs), activated by stress, injury, or necrosis, but not by physiological (apoptotic) cell death, initiate adaptive immune responses. APC activation is fundamentally associated with binding of CD40 to its ligand CD154. Platelets express CD154 upon activation and are thus potential primal danger signals linking the homeostatic response to trauma to activation of the acquired immune system. Previously, we showed that platelets can undergo gradient-driven migration, or chemotaxis, toward supernatants from cells injured by repeated freeze/thaws, UV light, or ischemia/reperfusion. Herein, we demonstrate that platelet-derived CD154 induces immature dendritic cell maturation with upregulation of costimulatory molecules and IL-12p40 production. Overall, these results provide a mechanism for platelet activation of APC facilitating the induction of adaptive immunity in environments of cell injury.


Subject(s)
Antigen-Presenting Cells/physiology , Blood Platelets/physiology , CD40 Ligand/physiology , Humans , Immunity, Innate , Interleukin-12/analysis , Interleukin-12 Subunit p40 , Protein Subunits/analysis
20.
Neurobiol Aging ; 23(5): 777-85, 2002.
Article in English | MEDLINE | ID: mdl-12392781

ABSTRACT

The mechanism linking the APOE4 gene with increased susceptibility for Alzheimer's disease (AD) and poorer outcomes following closed head injury and stroke is unknown. One potential link is activation of the innate immune system in the CNS. Our previously published data demonstrated that apolipoprotein E regulates production of nitric oxide, a critical cytoactive factor released by immune active macrophages. To determine if immune regulation is different in the presence of apolipoprotein E4 compared to apolipoprotein E3, we have measured NO production by peritoneal and CNS macrophages (microglia) cultured from transgenic mice that only express the human apoE4 or apoE3 protein isoform. Significantly more NO was produced in APOE4 mice compared to APOE3 transgenic mice that only express human apoE3 protein. Similarly, monocyte derived macrophages from humans carrying APOE4 gene alleles also produce significantly greater NO than those individuals with APOE3. The mechanism for this isoform-specific difference in NO production is not known and multiple sites in the NO production pathway may be affected. Expression of inducible nitric oxide synthase (iNOS) mRNA and protein are not significantly different between the APOE3 and APOE4 mice, suggesting that induction of iNOS is not a primary cause of the increased NO production in APOE4 animals. One alternative regulatory mechanism that demonstrates isoform specificity is arginine transport, which is greater in microglia from APOE4 transgenic mice compared to microglia from APOE3 mice. Increased transport is consistent with an increased production of NO and may reflect a direct or indirect effect of the APOE genotype on microglial arginine uptake and microglial activation in general. Overall, greater NO production in APOE4 carriers where characteristically high levels of oxidative/nitrosative stress are found in diseases such as AD provides a mechanism that potentially explains the genetic association between APOE4 and human diseases.


Subject(s)
Alzheimer Disease/metabolism , Apolipoproteins E/metabolism , Microglia/metabolism , Nitric Oxide/metabolism , Oxidative Stress/genetics , Alzheimer Disease/epidemiology , Alzheimer Disease/genetics , Animals , Apolipoprotein E3 , Apolipoprotein E4 , Apolipoproteins E/genetics , Arginine/metabolism , Cells, Cultured , Genetic Predisposition to Disease/epidemiology , Humans , Macrophages, Peritoneal/metabolism , Mice , Microglia/cytology , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL