Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Proteomics ; 16(8): 1280-90, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26915739

ABSTRACT

Humoral response in cancer patients can be used for early cancer detection. By screening high-density protein microarrays with sera from colorectal cancer (CRC) patients and controls, we identified 16 tumor-associated antigens (TAAs) exhibiting high diagnostic value. This high number of TAAs requires the development of multiplex assays combining different antigens for a faster and more accurate prediction of CRC. Here, we have developed and optimized a bead-based assay using nine selected TAAs and two controls to provide a multiplex test for early CRC diagnosis. We screened a collection of 307 CRC patients' and control sera with the beads assay to identify and validate the best TAA combination for CRC detection. The multiplex bead-based assay exhibited a similar diagnostic performance to detect the humoral response in comparison to multiple ELISA analyses. After multivariate analysis, a panel composed of GTF2B, EDIL3, HCK, PIM1, STK4, and p53, together with gender and age, was identified as the best combination of TAAs for CRC diagnosis, achieving an AUC of 89.7%, with 66% sensitivity at 90.0% fixed specificity. The model was validated using bootstrapping analysis. In summary, we have developed a novel multiplex bead assay that after validation with a larger independent cohort of sera could be utilized in a high-throughput manner for population screening to facilitate the detection of early CRC patients.


Subject(s)
Antigens, Neoplasm/metabolism , Autoantibodies/metabolism , Colorectal Neoplasms/metabolism , Microspheres , Protein Array Analysis/methods , Aged , Antigens, Neoplasm/immunology , Autoantibodies/blood , Autoantibodies/immunology , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Calcium-Binding Proteins , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cell Adhesion Molecules , Colorectal Neoplasms/blood , Colorectal Neoplasms/diagnosis , Enzyme-Linked Immunosorbent Assay , Female , Humans , Intracellular Signaling Peptides and Proteins , Male , Middle Aged , Multivariate Analysis , Prognosis , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-hck/immunology , Proto-Oncogene Proteins c-hck/metabolism , Proto-Oncogene Proteins c-pim-1/immunology , Proto-Oncogene Proteins c-pim-1/metabolism , Reproducibility of Results , Sensitivity and Specificity , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism
2.
J Immunol ; 195(5): 2383-95, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26232427

ABSTRACT

Myeloid leukocyte recruitment into the lung in response to environmental cues represents a key factor for the induction of lung damage. We report that Hck- and Fgr-deficient mice show a profound impairment in early recruitment of neutrophils and monocytes in response to bacterial LPS. The reduction in interstitial and airway neutrophil recruitment was not due to a cell-intrinsic migratory defect, because Hck- and Fgr-deficient neutrophils were attracted to the airways by the chemokine CXCL2 as wild type cells. However, early accumulation of chemokines and TNF-α in the airways was reduced in hck(-/-)fgr(-/-) mice. Considering that chemokine and TNF-α release into the airways was neutrophil independent, as suggested by a comparison between control and neutrophil-depleted mice, we examined LPS-induced chemokine secretion by neutrophils and macrophages in wild type and mutant cells. Notably, mutant neutrophils displayed a marked deficit in their capability to release the chemokines CXCL1, CXCL2, CCL3, and CCL4 and TNF-α in response to LPS. However, intracellular accumulation of these chemokines and TNF-α, as well as secretion of a wide array of cytokines, including IL-1α, IL-1ß, IL-6, and IL-10, by hck(-/-)fgr(-/-) neutrophils was normal. Intriguingly, secretion of CXCL1, CXCL2, CCL2, CCL3, CCL4, RANTES, and TNF-α, but not IL-1α, IL-1ß, IL-6, IL-10, and GM-CSF, was also markedly reduced in bone marrow-derived macrophages. Consistently, the Src kinase inhibitors PP2 and dasatinib reduced chemokine secretion by neutrophils and bone marrow-derived macrophages. These findings identify Src kinases as a critical regulator of chemokine secretion in myeloid leukocytes during lung inflammation.


Subject(s)
Chemokines/immunology , Lipopolysaccharides/immunology , Lung/immunology , Myeloid Cells/immunology , Proto-Oncogene Proteins c-hck/immunology , Proto-Oncogene Proteins/immunology , src-Family Kinases/immunology , Animals , Cells, Cultured , Chemokines/metabolism , Dasatinib/immunology , Dasatinib/pharmacology , Immunohistochemistry , Lipopolysaccharides/pharmacology , Lung/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
3.
Oncotarget ; 6(18): 15752-71, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26087188

ABSTRACT

The hematopoietic cell kinase (HCK) is a member of the SRC family of cytoplasmic tyrosine kinases (SFKs), and is expressed in cells of the myeloid and B-lymphocyte cell lineages. Excessive HCK activation is associated with several types of leukemia and enhances cell proliferation and survival by physical association with oncogenic fusion proteins, and with functional interactions with receptor tyrosine kinases. Elevated HCK activity is also observed in many solid malignancies, including breast and colon cancer, and correlates with decreased patient survival rates. HCK enhances the secretion of growth factors and pro-inflammatory cytokines from myeloid cells, and promotes macrophage polarization towards a wound healing and tumor-promoting alternatively activated phenotype. Within tumor associated macrophages, HCK stimulates the formation of podosomes that facilitate extracellular matrix degradation, which enhance immune and epithelial cell invasion. By virtue of functional cooperation between HCK and bona fide oncogenic tyrosine kinases, excessive HCK activation can also reduce drug efficacy and contribute to chemo-resistance, while genetic ablation of HCK results in minimal physiological consequences in healthy mice. Given its known crystal structure, HCK therefore provides an attractive therapeutic target to both, directly inhibit the growth of cancer cells, and indirectly curb the source of tumor-promoting changes in the tumor microenvironment.


Subject(s)
Neoplasms/immunology , Neoplasms/therapy , Proto-Oncogene Proteins c-hck/immunology , Animals , Cell Proliferation/physiology , Humans , Signal Transduction
4.
J Biol Chem ; 288(15): 10395-405, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23439650

ABSTRACT

A critical step in the induction of adaptive mucosal immunity is antigen transcytosis, in which luminal antigens are transported to organized lymphoid tissues across the follicle-associated epithelium (FAE) of Peyer's patches. However, virtually nothing is known about intracellular signaling proteins and transcription factors that regulate apical-to-basolateral transcytosis. The FAE can transcytose a variety of luminal contents, including inert particles, in the absence of specific opsonins. Furthermore, it expresses receptors for secretory immunoglobulin A (SIgA), the main antibody in mucosal secretions, and uses them to efficiently transcytose SIgA-opsonized particles present in the lumen. Using a human FAE model, we show that the tyrosine kinase HCK regulates apical-to-basolateral transcytosis of non-opsonized and SIgA-opsonized particles. We also show that, in cultured intestinal epithelial cells, ectopic expression of the transcription factor SPIB or EHF is sufficient to activate HCK-dependent apical-to-basolateral transcytosis of these particles. Our results provide the first molecular insights into the intracellular regulation of antigen sampling at mucosal surfaces.


Subject(s)
DNA-Binding Proteins/immunology , Intestinal Mucosa/immunology , Models, Immunological , Peyer's Patches/immunology , Proto-Oncogene Proteins c-hck/immunology , Transcription Factors/immunology , Transcytosis/immunology , Animals , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Peyer's Patches/cytology , Peyer's Patches/metabolism , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
J Immunol ; 190(2): 531-8, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23248261

ABSTRACT

IL-33 is elevated in afflicted tissues of patients with mast cell (MC)-dependent chronic allergic diseases. Based on its acute effects on mouse MCs, IL-33 is thought to play a role in the pathogenesis of allergic disease through MC activation. However, the manifestations of prolonged IL-33 exposure on human MC function, which best reflect the conditions associated with chronic allergic disease, are unknown. In this study, we found that long-term exposure of human and mouse MCs to IL-33 results in a substantial reduction of MC activation in response to Ag. This reduction required >72 h exposure to IL-33 for onset and 1-2 wk for reversion following IL-33 removal. This hyporesponsive phenotype was determined to be a consequence of MyD88-dependent attenuation of signaling processes necessary for MC activation, including Ag-mediated calcium mobilization and cytoskeletal reorganization, potentially as a consequence of downregulation of the expression of phospholipase Cγ(1) and Hck. These findings suggest that IL-33 may play a protective, rather than a causative, role in MC activation under chronic conditions and, furthermore, reveal regulated plasticity in the MC activation phenotype. The ability to downregulate MC activation in this manner may provide alternative approaches for treatment of MC-driven disease.


Subject(s)
Immunosuppression Therapy , Interleukins/immunology , Mast Cells/immunology , Mast Cells/metabolism , Phenotype , Actins/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Humans , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/pharmacology , Mast Cells/drug effects , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Phospholipase C gamma/genetics , Phospholipase C gamma/immunology , Phospholipase C gamma/metabolism , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/immunology , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology
6.
J Immunol ; 187(11): 6043-51, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22021612

ABSTRACT

The TLRs play a key role in host defense against infection and injury, and mounting evidence suggests that these receptors may also play a role in diseases such autoimmunity, atherosclerosis, and cancer. Activation of TLRs on macrophages results in the production of multiple soluble mediators including the key inflammatory cytokines, TNF and IL-6. Thus, the intracellular signaling mechanism by which TLRs signal is a subject of great interest. As well as activating the NF-κB and MAPK pathways, TLR engagement leads to tyrosine kinase activation within minutes. Src family kinases (SFKs) are the largest nonreceptor tyrosine kinase family with nine members: Src, Hck, Lyn, Fyn, Fgr, Blk, Lck, Yes, and Ylk. The role of the SFKs in TLR signaling has been an area of much controversy, with conflicting findings between studies using chemical inhibitors and knockout mice. Using primary human macrophages in combination with adenoviral overexpression and small interfering RNA knockdown studies, we show that the SFK, Hck, has a pre-eminent role in LPS/TLR4-induced TNF and IL-6 production. Hck kinase mediates TLR4-induced transcription of both TNF and IL-6 by a mechanism that involves neither the NF-κB nor the MAPK pathways, but rather leads to AP-1 binding with a complex of c-fos and JunD. These data highlight the importance of Hck as an active component in LPS-induced TLR signaling and suggest the possibility of targeting this kinase for the alleviation of excessive inflammation.


Subject(s)
Interleukin-6/biosynthesis , Proto-Oncogene Proteins c-hck/metabolism , Toll-Like Receptor 4/metabolism , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Blotting, Western , Cells, Cultured , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Macrophages/immunology , Macrophages/metabolism , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/immunology , RNA Interference , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
7.
J Immunol ; 185(10): 5993-6002, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20944008

ABSTRACT

Mast cell activation via FcεRI involves activation of the Src family kinases (SFKs) Lyn, Fyn, and Hck that positively or, in the case of Lyn, negatively regulate cellular responses. Little is known of upstream activators of these SFKs in FcεRI-dependent signaling. We investigated the role of receptor protein tyrosine phosphatase (PTP)α, a well-known activator of SFKs in diverse signaling systems, FcεRI-mediated mast cell activation, and IgE-dependent allergic responses in mice. PTPα(-/-) bone marrow-derived mast cells hyperdegranulate and exhibit increased cytokine and cysteinyl leukotriene secretion, and PTPα(-/-) mice display enhanced IgE-dependent anaphylaxis. At or proximal to FcεRI, PTPα(-/-) cells have reduced IgE-dependent activation of Lyn and Fyn, as well as reduced FcεRI and SHIP phosphorylation. In contrast, Hck and Syk activation is enhanced. Syk hyperactivation correlated with its increased phosphorylation at positive regulatory sites and defective phosphorylation at a negative regulatory site. Distal to FcεRI, we observed increased activation of PI3K and MAPK pathways. These findings demonstrate that PTPα activates the FcεRI-coupled kinases Lyn and Fyn and suppresses Hck activity. Furthermore, the findings indicate that hyperactivation of PTPα(-/-) mast cells and enhanced IgE-dependent allergic responses of PTPα(-/-) mice are due to the ablated function of PTPα as a critical regulator of Lyn negative signaling.


Subject(s)
Mast Cells/immunology , Proto-Oncogene Proteins c-fyn/immunology , Proto-Oncogene Proteins c-hck/immunology , Receptor-Like Protein Tyrosine Phosphatases, Class 4/immunology , src-Family Kinases/immunology , Animals , Cell Degranulation/immunology , Hypersensitivity/immunology , Hypersensitivity/metabolism , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-fyn/metabolism , Proto-Oncogene Proteins c-hck/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Receptors, IgE/immunology , Receptors, IgE/metabolism , Signal Transduction/immunology , src-Family Kinases/metabolism
8.
J Immunol ; 184(12): 7030-9, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20488787

ABSTRACT

Macrophages are a major target of HIV-1 infection. HIV-1-infected macrophages form multinucleated giant cells (MGCs) using poorly elucidated mechanisms. In this study, we show that MGC formation was reduced when human macrophages were infected with nef-deleted HIV-1. Moreover, expression of Nef, an HIV-1 protein required in several aspects of AIDS, was sufficient to trigger the formation of MGCs in RAW264.7 macrophages. Among Nef molecular determinants, myristoylation was dispensable, whereas the polyproline motif was instrumental for this phenomenon. Nef has been shown to activate hematopoietic cell kinase (Hck), a Src tyrosine kinase specifically expressed in phagocytes, through a well-described polyproline-SH3 interaction. Knockdown approaches showed that Hck is involved in Nef-induced MGC formation. Hck is expressed as two isoforms located in distinct subcellular compartments. Although both isoforms were activated by Nef, only p61Hck mediated the effect of Nef on macrophage fusion. This process was abolished in the presence of a p61Hck kinase-dead mutant or when p61Hck was redirected from the lysosome membrane to the cytosol. Finally, lysosomal proteins including vacuolar adenosine triphosphatase and proteases participated in Nef-induced giant macrophage formation. We conclude that Nef participates in HIV-1-induced MGC formation via a p61Hck- and lysosomal enzyme-dependent pathway. This work identifies for the first time actors of HIV-1-induced macrophage fusion, leading to the formation of MGCs commonly found in several organs of AIDS patients.


Subject(s)
Gene Products, nef/metabolism , Giant Cells/virology , HIV Infections/metabolism , Macrophages/virology , Proto-Oncogene Proteins c-hck/metabolism , Animals , Cell Line , Gene Products, nef/immunology , Giant Cells/immunology , Giant Cells/metabolism , HIV Infections/immunology , Humans , Immunoblotting , Isoenzymes , Macrophages/immunology , Macrophages/metabolism , Mice , Microscopy, Fluorescence , Proto-Oncogene Proteins c-hck/immunology
9.
Curr Mol Med ; 9(1): 69-85, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19199943

ABSTRACT

The activity of tyrosine kinases is central to many cellular processes, and accumulating evidence suggests that their role in inflammation is no less profound. Three main tyrosine kinase families, the Src, Tec and Syk kinase families are intimately involved in TLR signalling, the critical first step in cellular recognition of invading pathogens and tissue damage. Their activity results in changes in gene expression in affected cells. Key amongst these genes are the cytokines, which orchestrate both the duration and extent of inflammation. Tyrosine kinases also play important roles in cytokine function, and are implicated in signalling through both pro- and anti-inflammatory cytokines such as TNF, IL-6 and IL-10. Thus, strategies to modulate tyrosine kinase activity have significant therapeutic potential in combating the chronic inflammatory state that is typical of many major health issues that face us today, including Rheumatoid Arthritis, Cardiovascular disease and cancer. Here we review current knowledge of the role of tyrosine kinases in inflammation with particular emphasis on their role in TLR signalling.


Subject(s)
Inflammation/immunology , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism , Signal Transduction/immunology , Adjuvants, Immunologic/metabolism , Animals , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/metabolism , Cell Movement/immunology , Chronic Disease , Cytokines/biosynthesis , Cytokines/immunology , Cytokines/metabolism , Focal Adhesion Kinase 2/chemistry , Focal Adhesion Kinase 2/immunology , Focal Adhesion Kinase 2/metabolism , Gene Expression/immunology , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Janus Kinases/chemistry , Janus Kinases/immunology , Janus Kinases/metabolism , Mice , Protein-Tyrosine Kinases/chemistry , Proto-Oncogene Proteins c-hck/immunology , Proto-Oncogene Proteins c-hck/metabolism , Syk Kinase , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , src-Family Kinases/chemistry , src-Family Kinases/immunology , src-Family Kinases/metabolism
10.
J Immunol ; 178(6): 3874-85, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17339487

ABSTRACT

The chemotactic peptide formyl-methionyl-leucyl-phenilalanine (fMLP) triggers intracellular protein tyrosine phosphorylation leading to neutrophil activation. Deficiency of the Src family kinases Hck and Fgr have previously been found to regulate fMLP-induced degranulation. In this study, we further investigate fMLP signaling in hck-/-fgr-/- neutrophils and find that they fail to activate a respiratory burst and display reduced F-actin polymerization in response to fMLP. Additionally, albeit migration of both hck-/-fgr-/-mouse neutrophils and human neutrophils incubated with the Src family kinase inhibitor 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2) through 3-microm pore size Transwells was normal, deficiency, or inhibition, of Src kinases resulted in a failure of neutrophils to migrate through 1-microm pore size Transwells. Among MAPKs, phosphorylation of ERK1/2 was not different, phosphorylation of p38 was only partially affected, and phosphorylation of JNK was markedly decreased in fMLP-stimulated hck-/-fgr-/- neutrophils and in human neutrophils incubated with PP2. An increase in intracellular Ca(2+) concentration and phosphorylation of Akt/PKB occurred normally in fMLP-stimulated hck-/-fgr-/- neutrophils, indicating that activation of both phosphoinositide-specific phospholipase C and PI3K is independent of Hck and Fgr. In contrast, phosphorylation of the Rho/Rac guanine nucleotide exchange factor Vav1 and the Rac target p21-activated kinases were markedly reduced in both hck-/-fgr-/- neutrophils and human neutrophils incubated with a PP2. Consistent with these findings, PP2 inhibited Rac2 activation in human neutrophils. We suggest that Hck and Fgr act within a signaling pathway triggered by fMLP receptors that involves Vav1 and p21-activated kinases, leading to respiratory burst and F-actin polymerization.


Subject(s)
Cell Degranulation/immunology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophil Activation/immunology , Neutrophils/immunology , Proto-Oncogene Proteins c-hck/immunology , Proto-Oncogene Proteins/immunology , src-Family Kinases/immunology , Animals , Cell Degranulation/drug effects , Cell Degranulation/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell Movement/immunology , Female , Humans , Male , Mice , Mice, Knockout , Neutrophil Activation/drug effects , Neutrophil Activation/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-hck/antagonists & inhibitors , Proto-Oncogene Proteins c-hck/deficiency , Proto-Oncogene Proteins c-vav/immunology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/deficiency
11.
Immunol Cell Biol ; 84(2): 154-65, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16519733

ABSTRACT

The structures, molecular interactions and functions of CD4 in a subset of T lymphocytes have been well characterized. The CD4 receptors of other cell types have, however, been poorly documented. We have previously shown that lymphocytes and monocytes/macrophages differ in their expression of CD4 monomers and dimers. In the present study, we have shown further significant differences. Variability in the blocking of CD4 mAb binding by sulfated polyanions indicated differences in exofacial CD4 structures. In contrast to the well-documented 55 kDa monomers in lymphocytic cells, monocytic cells were found to coexpress two monomer isoforms: the 55 kDa form and a novel 59 kDa species. Experimental uncoupling of CD4 disulfides indicated that the oxidized 55 kDa monomer could be converted to the 59 kDa form. This was achieved by chemical reduction of purified native or recombinant CD4, or in cell transfection experiments by mutation of cysteine to alanine in domain 1 (D1) (Cys16 or Cys84) and in domain 4 (D4) (Cys303 or Cys345). All of these modifications promote CD4 distension on SDS-PAGE analysis and indicate that, when CD4 inter-beta-sheet disulfides in the D1 and D4 Ig folds are disrupted, there is an unravelling of the oxidized form to an extended 59 kDa unfolded state. We hypothesize that this may be a transition-state, structural-intermediate in the formation of disulfide-linked homodimers. Also identified were CD4-tyrosine kinase dissimilarities in which lymphocyte CD4 associated with Lck, but monocyte CD4 associated with HcK. These findings show that there is complex heterogeneity in structures and interactions in the CD4 of T lymphocytes and monocytes.


Subject(s)
CD4 Antigens/immunology , Cell Communication/immunology , Macrophages/immunology , T-Lymphocyte Subsets/immunology , Amino Acid Substitution , CD4 Antigens/genetics , Cell Communication/genetics , Cell Line , Dimerization , Gene Expression , Humans , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins c-hck/immunology , Structure-Activity Relationship , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...