Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Invest Dermatol ; 141(9): 2291-2299.e2, 2021 09.
Article in English | MEDLINE | ID: mdl-33773987

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal-dominant disorder characterized by hamartomatous tumors of the skin, kidneys, brain, and lungs. TSC is caused by mutations in the TSC1 and TSC2 genes, which result in hyperactivation of the mTOR, leading to dysregulated cell growth and autophagy. Rapamycin (sirolimus) shrinks TSC tumors, but the clinical benefits of sirolimus are not sustained after its withdrawal. In this study, we studied the cellular processes critical for tumor formation and growth, including cell proliferation and cell size. TSC2-/- and TSC2+/- cells were isolated from TSC skin tumors and normal-appearing skin, respectively. Cells were incubated with sirolimus for 72 hours. Withdrawal of sirolimus from TSC2-/- cells resulted in a highly proliferative phenotype and caused cells to enter the S phase of the cell cycle, with persistent phosphorylation of mTOR, p70 S6 kinase, ribosomal protein S6, and 4EB-P1; decreased cyclin D kinase inhibitors; and transient hyperactivation of protein kinase B. Sirolimus modulated the estrogen- and autophagy-dependent volume of TSC2-/- cells. These results suggest that sirolimus may decrease the size of TSC tumors by reducing TSC2-/- cell volume, altering the cell cycle, and reprogramming TSC2-null cells.


Subject(s)
Angiofibroma/drug therapy , Fibroblasts/physiology , Skin Neoplasms/drug therapy , Skin/pathology , Tuberous Sclerosis Complex 2 Protein/metabolism , Antibiotics, Antineoplastic/pharmacology , Autophagy , Carcinogenesis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Size , Cellular Reprogramming , Estrogens/metabolism , Humans , Mutation, Missense/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics
2.
Am J Respir Crit Care Med ; 188(7): 831-41, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23924348

ABSTRACT

RATIONALE: Alveolar transforming growth factor (TGF)-ß1 signaling and expression of TGF-ß1 target genes are increased in patients with idiopathic pulmonary fibrosis (IPF) and in animal models of pulmonary fibrosis. Internalization and degradation of TGF-ß receptor TßRI inhibits TGF-ß signaling and could attenuate development of experimental lung fibrosis. OBJECTIVES: To demonstrate that after experimental lung injury, human syndecan-2 confers antifibrotic effects by inhibiting TGF-ß1 signaling in alveolar epithelial cells. METHODS: Microarray assays were performed to identify genes differentially expressed in alveolar macrophages of patients with IPF versus control subjects. Transgenic mice that constitutively overexpress human syndecan-2 in macrophages were developed to test the antifibrotic properties of syndecan-2. In vitro assays were performed to determine syndecan-2-dependent changes in epithelial cell TGF-ß1 signaling, TGF-ß1, and TßRI internalization and apoptosis. Wild-type mice were treated with recombinant human syndecan-2 during the fibrotic phase of bleomycin-induced lung injury. MEASUREMENTS AND MAIN RESULTS: We observed significant increases in alveolar macrophage syndecan-2 levels in patients with IPF. Macrophage-specific overexpression of human syndecan-2 in transgenic mice conferred antifibrotic effects after lung injury by inhibiting TGF-ß1 signaling and downstream expression of TGF-ß1 target genes, reducing extracellular matrix production and alveolar epithelial cell apoptosis. In vitro, syndecan-2 promoted caveolin-1-dependent internalization of TGF-ß1 and TßRI in alveolar epithelial cells, which inhibited TGF-ß1 signaling and epithelial cell apoptosis. Therapeutic administration of human syndecan-2 abrogated lung fibrosis in mice. CONCLUSIONS: Alveolar macrophage syndecan-2 exerts antifibrotic effects by promoting caveolin-1-dependent TGF-ß1 and TßRI internalization and inhibiting TGF-ß1 signaling in alveolar epithelial cells. Hence, molecules that facilitate TßRI degradation via endocytosis represent potential therapies for pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis/chemically induced , Macrophages, Alveolar/drug effects , Syndecan-2/therapeutic use , Transforming Growth Factor beta1/drug effects , Animals , Apoptosis , Bleomycin/administration & dosage , Bronchoalveolar Lavage , Caveolin 1/drug effects , Disease Models, Animal , Gene Expression Profiling , Genetic Markers , Humans , Hydroxyproline/analysis , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/genetics , In Vitro Techniques , Mice , Mice, Transgenic , Signal Transduction , Syndecan-2/physiology , Tissue Array Analysis , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
3.
Proc Natl Acad Sci U S A ; 109(36): 14464-9, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22908276

ABSTRACT

Brefeldin A-inhibited guanine nucleotide-exchange protein (BIG)2 activates ADP-ribosylation factors, ∼20-kDa GTPase proteins critical for continuity of intracellular vesicular trafficking by accelerating the replacement of ADP-ribosylation factor-bound GDP with GTP. Mechanisms of additional BIG2 function(s) are less clear. Here, the participation of BIG2 in integrin ß1 cycling through actin dynamics during cell migration was identified using small interfering RNA (siRNA) and difference gel electrophoresis analyses. After a 72-h incubation with BIG2 siRNA, levels of cytosolic Arp2, Arp3, cofilin-1, phosphocofilin, vinculin, and Grb2, known to be involved in the effects of integrin ß1-extracellular matrix interactions on actin function and cell translocation, were increased. Treatment of HeLa cells with BIG2 siRNA resulted in perinuclear accumulation of integrin ß1 and its delayed return to the cell surface. Motility of BIG2-depleted cells was simultaneously decreased, as were actin-based membrane protrusions and accumulations of Arp2, Arp3, cofilin, and phosphocofilin at the leading edges of migrating cells, in wound-healing assays. Taken together, these data reveal a mechanism(s) through which BIG2 may coordinate actin cytoskeleton mechanics and membrane traffic in cell migration via integrin ß1 action and actin functions.


Subject(s)
Actins/physiology , Cell Movement/physiology , Guanine Nucleotide Exchange Factors/metabolism , Integrin beta1/metabolism , DNA Primers/genetics , Electrophoresis , Extracellular Matrix Proteins/metabolism , Fluorescent Antibody Technique , Guanine Nucleotide Exchange Factors/physiology , HeLa Cells , Humans , Image Processing, Computer-Assisted , RNA, Small Interfering/administration & dosage , Real-Time Polymerase Chain Reaction
4.
BMC Med Genomics ; 4: 20, 2011 Feb 28.
Article in English | MEDLINE | ID: mdl-21356094

ABSTRACT

BACKGROUND: The vascular disease in-stent restenosis (ISR) is characterized by formation of neointima and adverse inward remodeling of the artery after injury by coronary stent implantation. We hypothesized that the analysis of gene expression in peripheral blood mononuclear cells (PBMCs) would demonstrate differences in transcript expression between individuals who develop ISR and those who do not. METHODS AND RESULTS: We determined and investigated PBMC gene expression of 358 patients undergoing an index procedure to treat in de novo coronary artery lesions with bare metallic stents, using a novel time-varying intercept model to optimally assess the time course of gene expression across a time course of blood samples. Validation analyses were conducted in an independent sample of 97 patients with similar time-course blood sampling and gene expression data. We identified 47 probesets with differential expression, of which 36 were validated upon independent replication testing. The genes identified have varied functions, including some related to cellular growth and metabolism, such as the NAB2 and LAMP genes. CONCLUSIONS: In a study of patients undergoing bare metallic stent implantation, we have identified and replicated differential gene expression in peripheral blood mononuclear cells, studied across a time series of blood samples. The genes identified suggest alterations in cellular growth and metabolism pathways, and these results provide the basis for further specific functional hypothesis generation and testing of the mechanisms of ISR.


Subject(s)
Coronary Restenosis/genetics , Stents , Aged , Blood Cells/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Coronary Restenosis/metabolism , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation , Humans , Male , Middle Aged , Repressor Proteins/genetics , Repressor Proteins/metabolism , Software , Time Factors
5.
J Immunol ; 182(3): 1270-7, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155472

ABSTRACT

Lymphangioleiomyomatosis (LAM) is characterized by cystic lung destruction caused by LAM cells (smooth-muscle-like cells) that have mutations in the tumor suppressor genes tuberous sclerosis complex (TSC) 1 or 2 and have the capacity to metastasize. Since chemokines and their receptors function in chemotaxis of metastatic cells, we hypothesized that LAM cells may be recruited by chemokine(s) in the lung. Quantification of 25 chemokines in bronchoalveolar lavage fluid from LAM patients and healthy volunteers revealed that concentrations of CCL2, CXCL1, and CXCL5 were significantly higher in samples from LAM patients than those from healthy volunteers. In vitro, CCL2 or MCP-1 induced selective migration of cells, showing loss of heterozygosity of TSC2 from a heterogeneous population of cells grown from explanted LAM lungs. Additionally, the frequencies of single-nucleotide polymorphisms in the CCL2 gene promoter region differed significantly in LAM patients and healthy volunteers (p = 0.018), and one polymorphism was associated significantly more frequently with the decline of lung function. The presence (i.e., potential functionality) of chemokine receptors was evaluated using immunohistochemistry in lung sections from 30 LAM patients. Expression of chemokines and these receptors varied among LAM patients and differed from that seen in some cancers (e.g., breast cancer and melanoma cells). These observations are consistent with the notion that chemokines such as CCL2 may serve to determine mobility and specify the site of metastasis of the LAM cell.


Subject(s)
Chemokines/physiology , Chemotaxis, Leukocyte/immunology , Genes, Tumor Suppressor , Lymphangioleiomyomatosis/immunology , Lymphangioleiomyomatosis/pathology , Polymorphism, Genetic/immunology , Tumor Suppressor Proteins/genetics , Adult , Case-Control Studies , Cell Line , Cell Line, Tumor , Chemokine CCL2/physiology , Chemokine CCL27/physiology , Chemokines/biosynthesis , Chemokines/genetics , Chemokines, CC/physiology , Chemotaxis, Leukocyte/genetics , Female , Gene Expression Profiling , Humans , Lung/immunology , Lung/metabolism , Lung/pathology , Lymphangioleiomyomatosis/genetics , Middle Aged , Oligonucleotide Array Sequence Analysis , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency
6.
Am J Pathol ; 172(4): 1112-26, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18321997

ABSTRACT

Research suggests that monocytes differentiate into unique lineage-determined macrophage subpopulations in response to the local cytokine environment. The present study evaluated the atherogenic potential of two divergent lineage-determined human monocyte-derived macrophage subpopulations. Monocytes were differentiated for 7 days in the presence of alternative macrophage development cytokines: granulocyte-macrophage colony-stimulating factor to produce granulocyte-macrophage-CSF macrophages (GM-Mac), or macrophage colony-stimulating factor (M-CSF) to produce M-Mac. Gene chip analyses of three monocyte donors demonstrated differential expression of inflammatory and cholesterol homeostasis genes in the macrophage subpopulations. Quantitative PCR confirmed a fivefold elevation in the expression of genes that promote reverse cholesterol transport (PPAR-gamma, LXR-alpha, and ABCG1) and macrophage emigration from lesions (CCR7) in GM-Mac compared to that in M-Mac. Immunocytochemistry confirmed enhanced expression of the proinflammatory marker CD14 in M-Mac relative to GM-Mac. M-Mac spontaneously accumulated cholesterol when incubated with unmodified low-density lipoprotein whereas GM-Mac only accumulated similar levels of cholesterol after protein kinase C activation. Immunostained human coronary arteries showed that macrophages with similar antigen expression to that of M-Mac (CD68(+)/CD14(+)) were predominant within atherosclerotic lesions whereas macrophages with antigen expression similar to GM-Mac (CD68(+)/CD14(-)) were predominant in areas devoid of disease. The identification of macrophage subpopulations with different gene expression patterns and, thus, different potentials for promoting atherosclerosis has important experimental and clinical implications and could prove to be a valuable finding in developing therapeutic interventions in diseases dependent on macrophage function.


Subject(s)
Atherosclerosis/pathology , Macrophages/pathology , Biomarkers/metabolism , Cell Differentiation , Cells, Cultured , Cholesterol/metabolism , Coronary Vessels/pathology , Cytokines/metabolism , Fluorescent Antibody Technique , Gene Expression Profiling , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Inflammation , Lipid Metabolism/genetics , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Phenotype , Polymerase Chain Reaction
7.
Proc Natl Acad Sci U S A ; 105(9): 3539-44, 2008 Mar 04.
Article in English | MEDLINE | ID: mdl-18292222

ABSTRACT

Patients with tuberous sclerosis complex (TSC) develop hamartomas containing biallelic inactivating mutations in either TSC1 or TSC2, resulting in mammalian target of rapamycin (mTOR) activation. Hamartomas overgrow epithelial and mesenchymal cells in TSC skin. The pathogenetic mechanisms for these changes had not been investigated, and the existence or location of cells with biallelic mutations ("two-hit" cells) was unclear. We compared TSC skin hamartomas (angiofibromas and periungual fibromas) with normal-appearing skin of the same patient, and we observed more proliferation and mTOR activation in hamartoma epidermis. Two-hit cells were not detected in the epidermis. Fibroblast-like cells in the dermis, however, exhibited allelic deletion of TSC2, in both touch preparations of fresh tumor samples and cells grown from TSC skin tumors, suggesting that increased epidermal proliferation and mTOR activation were not caused by second-hit mutations in the keratinocytes but by mesenchymal-epithelial interactions. Gene expression arrays, used to identify potential paracrine factors released by mesenchymal cells, revealed more epiregulin mRNA in fibroblast-like angiofibroma and periungual fibroma cells than in fibroblasts from normal-appearing skin of the same patient. Elevation of epiregulin mRNA was confirmed with real-time PCR, and increased amounts of epiregulin protein were demonstrated with immunoprecipitation. Epiregulin stimulated keratinocyte proliferation and phosphorylation of ribosomal protein S6 in vitro. These results suggest that hamartomatous TSC skin tumors are induced by paracrine factors released by two-hit cells in the dermis and that proliferation with mTOR activation of the overlying epidermis is an effect of epiregulin.


Subject(s)
Epidermal Growth Factor/genetics , Epithelium/pathology , Hamartoma/pathology , Mesoderm/pathology , Paracrine Communication , Tuberous Sclerosis/pathology , Cell Proliferation , Epidermal Growth Factor/analysis , Epiregulin , Gene Expression Profiling , Humans , Protein Kinases/metabolism , RNA, Messenger/analysis , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
8.
PLoS One ; 2(6): e544, 2007 Jun 20.
Article in English | MEDLINE | ID: mdl-17579716

ABSTRACT

BACKGROUND: Available blood assays for venous thromboembolism (VTE) suffer from diminished specificity. Compared with single marker tests, such as D-dimer, a multi-marker strategy may improve diagnostic ability. We used direct mass spectrometry (MS) analysis of serum from patients with VTE to determine whether protein expression profiles would predict diagnosis. METHODS AND RESULTS: We developed a direct MS and computational approach to the proteomic analysis of serum. Using this new method, we analyzed serum from inpatients undergoing radiographic evaluation for VTE. In a balanced cohort of 76 patients, a neural network-based prediction model was built using a training subset of the cohort to first identify proteomic patterns of VTE. The proteomic patterns were then validated in a separate group of patients within the cohort. The model yielded a sensitivity of 68% and specificity of 89%, which exceeded the specificity of D-dimer assay tested by latex agglutination, ELISA, and immunoturbimetric methods (sensitivity/specificity of 63.2%/60.5%, 97.4%/21.1%, 97.4%/15.8%, respectively). We validated differences in protein expression between patients with and without VTE using more traditional gel-based analysis of the same serum samples. CONCLUSION: Protein expression analysis of serum using direct MS demonstrates potential diagnostic utility for VTE. This pilot study is the first such direct MS study to be applied to a cardiovascular disease. Differences in protein expression were identified and subsequently validated in a separate group of patients. The findings in this initial cohort can be evaluated in other independent cohorts, including patients with inflammatory conditions and chronic (but not acute) VTE, for the diagnosis of VTE.


Subject(s)
Biomarkers, Tumor/blood , Proteomics , Venous Thromboembolism/blood , Venous Thromboembolism/diagnosis , Algorithms , Cohort Studies , Diabetes Complications/blood , Electrophoresis, Gel, Two-Dimensional , Enzyme-Linked Immunosorbent Assay , Female , Fibrin Fibrinogen Degradation Products/metabolism , Heart Diseases/blood , Heart Diseases/complications , Humans , Kidney Diseases/blood , Kidney Diseases/complications , Lung Diseases/blood , Lung Diseases/complications , Male , Middle Aged , Neoplasms/blood , Neoplasms/complications , Neural Networks, Computer , Pilot Projects , Prognosis , Sensitivity and Specificity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
Am J Respir Crit Care Med ; 175(11): 1151-7, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17332483

ABSTRACT

RATIONALE: Alveolar macrophages are inflammatory cells that may contribute to the pathogenesis of idiopathic pulmonary fibrosis (IPF), which is characterized by excessive alveolar aggregation of cells and extracellular matrix proteins. OBJECTIVES: To identify potential molecular mechanisms of IPF. METHODS: To examine large-scale gene expression, messenger RNA isolated from alveolar macrophages and peripheral blood mononuclear cells from subjects with IPF and normal volunteers was hybridized to cDNA filters. MEASUREMENTS AND MAIN RESULTS: We showed that in IPF there is global down-regulation of gene expression in alveolar macrophages but not in blood monocytes. Nuclear run-on and pulse-chase studies showed that alveolar macrophages had significantly reduced transcription (p < 0.01). No significant difference in RNA degradation was found between subjects with IPF and normal volunteers. Western blot analyses revealed that concentrations of transcription factor II-H, a general transcription factor, were significantly lower in alveolar macrophages from subjects with IPF than in those from normal volunteers (p = 0.012). CONCLUSIONS: Impaired transcription in IPF is associated with decreased concentrations of transcription factor II-H in alveolar macrophages and may alter the intraalveolar milieu in IPF.


Subject(s)
Macrophages, Alveolar/physiology , Pulmonary Fibrosis/genetics , RNA Stability , RNA, Messenger/genetics , Transcription, Genetic , Biomarkers , Blotting, Northern , Blotting, Western , Disease Progression , Down-Regulation , Humans , Monocytes/physiology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Transcription Factor TFIIH/genetics , Transcription Factor TFIIH/metabolism
10.
J Biol Chem ; 281(44): 33053-65, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-16928680

ABSTRACT

The identification of ABCA1 as a key transporter responsible for cellular lipid efflux has led to considerable interest in defining its role in cholesterol metabolism and atherosclerosis. In this study, the effect of overexpressing ABCA1 in the liver of LDLr-KO mice was investigated. Compared with LDLr-KO mice, ABCA1-Tg x LDLr-KO (ABCA1-Tg) mice had significantly increased plasma cholesterol levels, mostly because of a 2.8-fold increase in cholesterol associated with a large pool of apoB-lipoproteins. ApoB synthesis was unchanged but the catabolism of (125)I-apoB-VLDL and -LDL were significantly delayed, accounting for the 1.35-fold increase in plasma apoB levels in ABCA1-Tg mice. We also found rapid in vivo transfer of free cholesterol from HDL to apoB-lipoproteins in ABCA1-Tg mice, associated with a significant 2.7-fold increase in the LCAT-derived cholesteryl linoleate content found primarily in apoB-lipoproteins. ABCA1-Tg mice had 1.4-fold increased hepatic cholesterol concentrations, leading to a compensatory 71% decrease in de novo hepatic cholesterol synthesis, as well as enhanced biliary cholesterol, and bile acid secretion. CAV-1, CYP2b10, and ABCG1 were significantly induced in ABCA1-overexpressing livers; however, no differences were observed in the hepatic expression of CYP7alpha1, CYP27alpha1, or ABCG5/G8 between ABCA1-Tg and control mice. As expected from the pro-atherogenic plasma lipid profile, aortic atherosclerosis was increased 10-fold in ABCA1-Tg mice. In summary, hepatic overexpression of ABCA1 in LDLr-KO mice leads to: 1) expansion of the pro-atherogenic apoB-lipoprotein cholesterol pool size via enhanced transfer of HDL-cholesterol to apoB-lipoproteins and delayed catabolism of cholesterol-enriched apoB-lipoproteins; 2) increased cholesterol concentration in the liver, resulting in up-regulated hepatobiliary sterol secretion; and 3) significantly enhanced aortic atherosclerotic lesions.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Lipoproteins/metabolism , Liver/metabolism , Receptors, LDL/deficiency , Receptors, LDL/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Animals , Aorta/metabolism , Aorta/pathology , Atherosclerosis/genetics , Biliary Tract/metabolism , Cholesterol/blood , Disease Progression , Feces , Female , Gene Expression Regulation , Hemostasis , Humans , Intestinal Mucosa/metabolism , Lipid Metabolism , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Organ Specificity , Receptors, LDL/genetics , Sterols/metabolism
11.
PLoS Biol ; 2(12): e423, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15550989

ABSTRACT

Murine leukemia virus (MLV)-derived vectors are widely used for hematopoietic stem cell (HSC) gene transfer, but lentiviral vectors such as the simian immunodeficiency virus (SIV) may allow higher efficiency transfer and better expression. Recent studies in cell lines have challenged the notion that retroviruses and retroviral vectors integrate randomly into their host genome. Medical applications using these vectors are aimed at HSCs, and thus large-scale comprehensive analysis of MLV and SIV integration in long-term repopulating HSCs is crucial to help develop improved integrating vectors. We studied integration sites in HSCs of rhesus monkeys that had been transplanted 6 mo to 6 y prior with MLV- or SIV-transduced CD34(+)cells. Unique MLV (491) and SIV (501) insertions were compared to a set of in silico-generated random integration sites. While MLV integrants were located predominantly around transcription start sites, SIV integrants strongly favored transcription units and gene-dense regions of the genome. These integration patterns suggest different mechanisms for integration as well as distinct safety implications for MLV versus SIV vectors.


Subject(s)
Genetic Vectors , Genome , Hematopoietic Stem Cells/virology , Leukemia Virus, Murine/metabolism , Simian Immunodeficiency Virus/metabolism , Stem Cells/virology , Animals , Antigens, CD34/biosynthesis , Binding Sites , Cell Line , Cloning, Molecular , Cluster Analysis , DNA Primers/chemistry , Gene Transfer Techniques , Macaca mulatta , Molecular Sequence Data , Mutation , Polymerase Chain Reaction , Retroviridae/genetics , Time Factors , Transcription, Genetic
12.
Blood ; 103(1): 325-32, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14504100

ABSTRACT

An immune pathophysiology for acquired aplastic anemia (AA) has been inferred from the responsiveness of the patients to immunosuppressive therapies and experimental laboratory data. To address the transcriptome of hematopoietic cells in AA, we undertook GeneChip analysis of the extremely limited numbers of progenitor and stem cells in the marrow of patients with this disease. We pooled total RNA from highly enriched bone marrow CD34 cells of 36 patients with newly diagnosed AA and 12 healthy volunteers for analysis on oligonucleotide chips. A large number of genes implicated in apoptosis and cell death showed markedly increased expression in AA CD34 cells, and negative proliferation control genes also had increased activity. Conversely, cell cycle progress-enhancing genes showed low expression in AA. Cytokine/chemokine signal transducer genes, stress response genes, and defense/immune response genes were up-regulated, as anticipated from other evidence of the heightened immune activity in AA patients' marrow. In summary, detailed genetic analysis of small numbers of hematopoietic progenitor cells is feasible even in marrow failure states where such cells are present in very small numbers. The gene expression profile of primary human CD34 hematopoietic stem cells from AA was consistent with a stressed, dying, and immunologically activated target cell population. Many of the genes showing differential expression in AA deserve further detailed analysis, including comparison with other marrow failure states and autoimmune disease.


Subject(s)
Anemia, Aplastic/genetics , Anemia, Aplastic/immunology , Antigens, CD34/metabolism , Adolescent , Adult , Aged , Anemia, Aplastic/pathology , Base Sequence , Case-Control Studies , Cell Cycle/genetics , Cell Division/genetics , Child , DNA, Complementary/genetics , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis
13.
Proteins ; 53(2): 307-19, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14517981

ABSTRACT

The potential for therapeutic specificity in regulating diseases and for reduced side effects has made cannabinoid (CB) receptors one of the most important G-protein-coupled receptor (GPCR) targets for drug discovery. The cannabinoid (CB) receptor subtype CB2 is of particular interest due to its involvement in signal transduction in the immune system and its increased characterization by mutational and other studies. However, our understanding of their mode of action has been limited by the absence of an experimental receptor structure. In this study, we have developed a 3D model of the CB2 receptor based on the recent crystal structure of a related GPCR, bovine rhodopsin. The model was developed using multiple sequence alignment of homologous receptor sub-types in humans and mammals, and compared with other GPCRs. Alignments were analyzed with mutation scores, pairwise hydrophobicity profiles and Kyte-Doolittle plots. The 3D model of the transmembrane segment was generated by mapping the CB2 sequence onto the homologous residues of the rhodopsin structure. The extra- and intracellular loop regions of the CB2 were generated by searching for homologous C(alpha) backbone sequences in published structures in the Brookhaven Protein Databank (PDB). Residue side chains were positioned through a combination of rotamer library searches, simulated annealing and minimization. Intermediate models of the 7TM helix bundles were analyzed in terms of helix tilt angles, hydrogen-bond networks, conserved residues and motifs, possible disulfide bonds. The amphipathic cytoplasmic helix domain was also correlated with biological and site-directed mutagenesis data. Finally, the model receptor-binding cavity was characterized using solvent-accessible surface approach.


Subject(s)
Models, Molecular , Receptor, Cannabinoid, CB2/chemistry , Amino Acid Sequence , Amino Acids/chemistry , Disulfides/chemistry , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/chemistry , Sequence Alignment
14.
Nat Med ; 9(4): 431-8, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12612546

ABSTRACT

High-dose intravenous immunoglobulin (IVIG) prevents immune damage by scavenging complement fragments C3b and C4b. We tested the hypothesis that exogenous immunoglobulin molecules also bind anaphylatoxins C3a and C5a, thereby neutralizing their pro-inflammatory effects. Single-cell calcium measurements in HMC-1 human mast cells showed that a rise in intracellular calcium caused by C3a and C5a was inhibited in a concentration-dependent manner by IVIG, F(ab)2-IVIG and irrelevant human monoclonal antibody. C3a- and C5a-induced thromboxane (TXB2) generation and histamine release from HMC-1 cells and whole-blood basophils were also suppressed by exogenous immunoglobulins. In a mouse model of asthma, immunoglobulin treatment reduced cellular migration to the lung. Lethal C5a-mediated circulatory collapse in pigs was prevented by pretreatment with F(ab)2-IVIG. Molecular modeling, surface plasmon resonance (SPR) and western blot analyses suggested a physical association between anaphylatoxins and the constant region of F(ab)2. This binding could interfere with the role of C3a and C5a in inflammation.


Subject(s)
Complement C3a/antagonists & inhibitors , Complement C5a/antagonists & inhibitors , Immunoglobulins, Intravenous/pharmacology , gamma-Globulins/pharmacology , Animals , Asthma/metabolism , Blood Pressure/drug effects , Calcium , Cell Line , Cell Migration Inhibition , Complement C3a/metabolism , Complement C5a/metabolism , Dose-Response Relationship, Drug , Histamine Release/drug effects , Humans , Mast Cells/metabolism , Mice , Respiratory Distress Syndrome/prevention & control , Swine , Thromboxane B2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...