Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Anal Chem ; 94(9): 3914-3921, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35188385

ABSTRACT

Oligomeric organization of G protein-coupled receptors is proposed to regulate receptor signaling and function, yet rapid and precise identification of the oligomeric status especially for native receptors on a cell membrane remains an outstanding challenge. By using blinking carbon dots (CDs), we now develop a deep learning (DL)-based blinking fingerprint recognition method, named deep-blinking fingerprint recognition (BFR), which allows automatic classification of CD-labeled receptor organizations on a cell membrane. This DL model integrates convolutional layers, long-short-term memory, and fully connected layers to extract time-dependent blinking features of CDs and is trained to a high accuracy (∼95%) for identifying receptor organizations. Using deep blinking fingerprint recognition, we found that CXCR4 mainly exists as 87.3% monomers, 12.4% dimers, and <1% higher-order oligomers on a HeLa cell membrane. We further demonstrate that the heterogeneous organizations can be regulated by various stimuli at different degrees. The receptor-binding ligands, agonist SDF-1α and antagonist AMD3100, can induce the dimerization of CXCR4 to 33.1 and 20.3%, respectively. In addition, cytochalasin D, which inhibits actin polymerization, similarly prompts significant dimerization of CXCR4 to 30.9%. The multi-pathway organization regulation will provide an insight for understanding the oligomerization mechanism of CXCR4 as well as for elucidating their physiological functions.


Subject(s)
Carbon , Deep Learning , Quantum Dots , Receptors, CXCR4 , Benzylamines/chemistry , Benzylamines/pharmacology , Chemokine CXCL12/agonists , Cyclams/chemistry , Cyclams/pharmacology , HeLa Cells , Humans , Receptors, CXCR4/chemistry
2.
Inflammation ; 43(2): 664-672, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31814061

ABSTRACT

The pathogenesis of keloids has not been elucidated, and the disease is thought to be caused by abnormal secretion of proinflammatory mediators and irregular responses to other inflammatory signals mediated by keloid fibroblasts (KFs). In this study, we investigated whether a local increase in interleukin IL-17 in keloid tissues stimulates the production of stromal cell-derived factor-1 (SDF-1) in KFs causing further recruitment of IL-17-producing T helper 17 (Th17) cells, which subsequently creates a positive feedback loop. Histological assessment was performed and the change in the expression of IL-17, IL-1ß, IL-6, and TNF-α which of fibrosis and inflammation associated markers was examined. In addition, fibroblasts were treated with IL-17 in the presence or absence of STAT3 inhibitor STA-21; SDF-1 levels and fibrosis genes were measured. Our results showed that fibrotic reaction and expression of proinflammatory cytokines including IL-17 were most prominent in the growing margin (perilesional area) of keloid tissue and Th17 cells significantly infiltrated the perilesional area. In addition, IL-17 upregulated the expression of SDF-1, collagen, and α-SMA in KFs. Finally, STA-21 decreased SDF-1α expression and the expression of fibrosis genes in KFs even after IL-17 stimulation. Our study demonstrated that a local increase in IL-17 in keloid tissues stimulates the production of SDF-1 in KFs causing further recruitment of IL-17-producing T helper 17 (Th17) cells, which subsequently creates a positive feedback loop. These findings suggest that STAT3 inhibition can be used to treat keloid scars by reversing the vicious cycle between Th17 cells and KFs.


Subject(s)
Chemokine CXCL12/biosynthesis , Fibroblasts/metabolism , Interleukin-17/pharmacology , Keloid/metabolism , STAT3 Transcription Factor/biosynthesis , Skin/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chemokine CXCL12/agonists , Fibroblasts/drug effects , Humans , Keloid/pathology , STAT3 Transcription Factor/agonists , Signal Transduction/drug effects , Signal Transduction/physiology , Skin/drug effects
3.
J Mol Endocrinol ; 59(1): R1-R10, 2017 07.
Article in English | MEDLINE | ID: mdl-28420715

ABSTRACT

Diabetic nephropathy is one of the most frequent, devastating and costly complications of diabetes. The available therapeutic approaches are limited. Dipeptidyl peptidase type 4 (DPP-4) inhibitors represent a new class of glucose-lowering drugs that might also have reno-protective properties. DPP-4 exists in two forms: a plasma membrane-bound form and a soluble form, and can exert many biological actions mainly through its peptidase activity and interaction with extracellular matrix components. The kidneys have the highest DPP-4 expression level in mammalians. DPP-4 expression and urinary activity are up-regulated in diabetic nephropathy, highlighting its role as a potential target to manage diabetic nephropathy. Preclinical animal studies and some clinical data suggest that DPP-4 inhibitors decrease the progression of diabetic nephropathy in a blood pressure- and glucose-independent manner. Many studies reported that these reno-protective effects could be due to increased half-life of DPP-4 substrates such as glucagon-like peptide-1 (GLP-1) and stromal derived factor-1 alpha (SDF-1a). However, the underlying mechanisms are far from being completely understood and clearly need further investigations.


Subject(s)
Diabetic Nephropathies/drug therapy , Dipeptidyl Peptidase 4/genetics , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Hypoglycemic Agents/therapeutic use , Mesangial Cells/drug effects , Podocytes/drug effects , Protective Agents/therapeutic use , Animals , Chemokine CXCL12/agonists , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Clinical Trials as Topic , Diabetic Nephropathies/enzymology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Dipeptidyl Peptidase 4/metabolism , Drug Evaluation, Preclinical , Extracellular Matrix , Gene Expression Regulation , Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Humans , Mesangial Cells/enzymology , Mesangial Cells/pathology , Podocytes/enzymology , Podocytes/pathology
4.
J Tissue Eng Regen Med ; 11(6): 1806-1819, 2017 06.
Article in English | MEDLINE | ID: mdl-26227988

ABSTRACT

Bone has the potential for spontaneous healing. This process, however, often fails in patients with comorbidities. Tissue engineering combining functional cells, biomaterials and osteoinductive cues may provide alternative treatment strategies. We have recently demonstrated that stromal cell-derived factor-1ß (SDF-1ß) works in concert with bone morphogenetic protein-2 (BMP-2) to potentiate osteogenic differentiation of bone marrow-derived mesenchymal stem/stromal cells (BMSCs). Here, we test the hypothesis that SDF-1ß overexpressed in Tet-Off-SDF-1ß BMSCs, delivered on acellular dermal matrix (ADM), synergistically augments BMP-2-induced healing of critical-sized mouse calvarial defects. BMSC therapies alone showed limited bone healing, which was increased with co-delivery of BMP-2. This was further enhanced in Tet-Off-SDF-1ß BMSCs + BMP-2. Only limited BMSC retention on ADM constructs was observed after 4 weeks in vivo, which was increased with BMP-2 co-delivery. In vitro cell proliferation studies showed that supplementing BMP-2 to Tet-Off BMSCs significantly increased the cell number during the first 24 h. Consequently, the increased cell numbers decreased the detectable BMP-2 levels in the medium, but increased cell-associated BMP-2. The data suggest that SDF-1ß provides synergistic effects supporting BMP-2-induced, BMSC-mediated bone formation and appears suitable for optimization of bone augmentation in combination therapy protocols. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Bone Morphogenetic Protein 2 , Cell Differentiation , Chemokine CXCL12 , Extracellular Matrix/chemistry , Fracture Healing , Gene Expression Regulation , Mesenchymal Stem Cells/metabolism , Osteogenesis , Skull , Animals , Bone Morphogenetic Protein 2/agonists , Bone Morphogenetic Protein 2/biosynthesis , Bone Morphogenetic Protein 2/genetics , Chemokine CXCL12/agonists , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Disease Models, Animal , Male , Mesenchymal Stem Cells/pathology , Mice , Skull/injuries , Skull/metabolism , Skull/pathology
5.
J Tissue Eng Regen Med ; 11(11): 3241-3252, 2017 11.
Article in English | MEDLINE | ID: mdl-27943613

ABSTRACT

Here we investigated the impact of hypoxic environment on the angiogenic properties of early-outgrowth endothelial progenitor cells (EPCs), with particular focus on the role of secreted vascular endothelial growth factor-A (VEGF-A) and stromal derived factor-1 (SDF-1) in mediating these effects. We found that cultured EPCs secreted factors with paracrine effects on chemotaxis, migration, proliferation and tube formation of mature endothelial cells (ECs), and these properties were not affected by hypoxia. Depletion of VEGF-A did not change the ability of EPC-conditioned medium (CM) to promote EC migration and tube formation in vitro, suggesting that the pro-angiogenic paracrine effects of EPCs did not totally rely on the presence of VEGF-A. These findings were confirmed by in vivo experiments, on a mouse model of hind limb ischaemia, which showed that VEGF-depleted EPC-CM sustained tissue perfusion at the same level as complete EPC-CM. However, concomitant deletion of VEGF-A and SDF-1 in EPC-CM impaired the pro-angiogenic properties of EPC-CM, by inhibition of EC spreading in culture, tube-like structure formation on Matrigel support, in vivo neovessels formation and ischaemic hind limb regeneration. Taken together, our data demonstrate that: (i) hypoxia does not affect the capacity of EPCs to support the angiogenic process; (ii) the absence of either VEGF-A or SDF-1 from EPC-CM can be rescued by the presence of the other one, so that the overall angiogenic effects remain unchanged; and (iii) and the concomitant deletion of VEGF-A and SDF-1 from EPC-CM impairs its pro-angiogenic effect, both in vitro and in vivo. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Chemokine CXCL12 , Endothelial Progenitor Cells/metabolism , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A , Animals , Chemokine CXCL12/agonists , Chemokine CXCL12/metabolism , Hindlimb/blood supply , Hindlimb/metabolism , Humans , Ischemia/metabolism , Ischemia/therapy , Mice , Vascular Endothelial Growth Factor A/agonists , Vascular Endothelial Growth Factor A/metabolism
6.
Ann Hematol ; 95(12): 1979-1988, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27542958

ABSTRACT

Dysregulation of B cell receptor (BCR) signalling is a hallmark of chronic lymphocytic leukaemia (CLL) pathology, and targeting BCR pathway kinases has brought great therapeutic advances. Activation of the BCR in lymphoid organs has been associated with CLL cell proliferation and survival, leading to progressive disease. While these responses are mediated predominantly by IgM, the role of IgD is less clear. Seeking to uncover downstream consequences of individual and combined stimulation of the two BCR isotypes, we found an amplification of IgD expression and IgD-mediated calcium signalling by previous stimulation of IgM in CLL. Furthermore, no heterologous downmodulation of the isotypes, as observed in healthy donors, was present. Only marginal downregulation of the expression of various chemokine receptors by α-IgM and α-IgD stimulation was found as compared to normal B cells. Consistently, calcium responses of CLL cells to different chemokines were only weakly affected by preceding BCR activation. In contrast, migration towards the two homeostatic chemokines CXCL12 and CCL21 was differentially regulated by IgM and IgD. While IgM activation reduced migration of CLL cells towards CXCL12, but not CCL21, IgD activation predominantly impacted on CCL21 but not CXCL12-mediated chemotaxis. This indicates that the preference for one chemokine over the other may depend on the functional presence of the two isotypes in CLL. Inhibitors against the kinases Syk, Lyn, and Btk antagonised both BCR- and chemokine-induced calcium signals.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Chemokine CCL21/metabolism , Chemokine CXCL12/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Receptors, Antigen, B-Cell/metabolism , Chemokine CCL21/agonists , Chemokine CXCL12/agonists , Chemokines/agonists , Chemokines/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Receptors, Antigen, B-Cell/agonists , Tumor Cells, Cultured
7.
J Mol Cell Cardiol ; 91: 72-80, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26739213

ABSTRACT

AIMS: Dipeptidyl peptidase-4 (DPP-4) inhibitors are reported to have protective effects on various cells but it is unclear how DPP-4 inhibitors have cardioprotective effects. Our aim was to study the mechanisms of cardioprotective effects by DPP-4 inhibition. METHODS AND RESULTS: C57BL/6 mice and DPP-4 knockout (DPP-4KO) mice were subjected to left coronary artery ligation to produce acute myocardial infarction (MI). C57BL/6 mice were then treated with vehicle or DPP-4 inhibitor. Left ventricular function, infarct size, the number of vessels, and myocardial ischemia were assessed at 5days after MI. The treatment with DPP-4 inhibitor significantly improved cardiac function and decreased the infarct size. DPP-4 inhibitor increased the ratio of endothelial cell numbers to a cardiomyocyte. The extent of myocardial ischemia and the number of TUNEL-positive cells in the border area were significantly decreased by DPP-4 inhibitor. Stromal cell-derived factor-1α (SDF-1α) level in myocardium was significantly increased by DPP-4 inhibitor. Those cardioprotective effects after MI were also recognized in DPP-4KO mice. DPP-4 protein was expressed on rat neonatal cardiomyocytes and DPP-4 inhibitor significantly reduced hypoxia-induced apoptosis in the cardiomyocytes. However, this effect was abolished by the pretreatment with a CXCR4 antagonist or a signal transducer and activator of transcription 3 (STAT3) inhibitor. The beneficial effects of DPP-4 inhibitor on heart failure after MI were abolished by cardiomyocyte-specific deletion of STAT3. CONCLUSIONS: DPP-4 inhibition may have direct protective effects on the post-MI heart by inducing an antiapoptotic effect and inhibiting a decrease in vessel number through the SDF-1α/CXCR4-mediated STAT3 signaling pathway.


Subject(s)
Chemokine CXCL12/genetics , Dipeptidyl Peptidase 4/genetics , Heart Failure/prevention & control , Myocardial Infarction/drug therapy , Receptors, CXCR4/genetics , STAT3 Transcription Factor/genetics , Animals , Animals, Newborn , Apoptosis/drug effects , Cardiotonic Agents/pharmacology , Chemokine CXCL12/agonists , Chemokine CXCL12/metabolism , Dipeptidyl Peptidase 4/deficiency , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction , Ventricular Function, Left/drug effects
8.
Stem Cell Rev Rep ; 12(1): 121-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26400757

ABSTRACT

Successful clinical outcomes from transplantation of hematopoietic stem cells (HSCs) depend upon efficient HSC homing to bone marrow (BM), subsequent engraftment, and, finally, BM repopulation. Homing of intravenously administered HSCs from peripheral blood (PB) through the circulation to the BM stem cell niches, which is the first critical step that precedes their engraftment, is enforced by chemotactic factors released in the BM microenvironment that chemoattract HSCs. These chemotactic factors include α-chemokine stromal-derived factor 1 (SDF-1), the bioactive phosphosphingolipids sphingosine-1-phosphate (S1P) and ceramid-1-phosphate (C1P), and the extracellular nucleotides ATP and UTP. Stem cells may also respond to a Ca(2+) or H(+) gradient by employing calcium- or proton-sensing receptors, respectively. In this review, we will present emerging strategies based on ex vivo manipulation of graft HSCs that are aimed at enhancing the responsiveness of HSCs to BM-secreted chemoattractants and/or promoting HSC adhesion and seeding efficiency in the BM microenvironment.


Subject(s)
Chemotactic Factors/pharmacology , Graft Survival/drug effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Stem Cell Niche/drug effects , Adenosine Triphosphate/agonists , Adenosine Triphosphate/immunology , Bone Marrow/drug effects , Bone Marrow/immunology , Ceramides/agonists , Ceramides/immunology , Ceramides/pharmacology , Chemokine CXCL12/agonists , Chemokine CXCL12/immunology , Chemokine CXCL12/pharmacology , Chemotactic Factors/agonists , Chemotactic Factors/immunology , Chemotaxis/drug effects , Dinoprostone/therapeutic use , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Lysophospholipids/agonists , Lysophospholipids/immunology , Lysophospholipids/pharmacology , Membrane Microdomains/drug effects , Receptors, CXCR4/agonists , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Sphingosine/agonists , Sphingosine/analogs & derivatives , Sphingosine/immunology , Sphingosine/pharmacology , Stem Cell Niche/immunology , Uridine Triphosphate/agonists , Uridine Triphosphate/immunology , Valproic Acid/therapeutic use
10.
Curr Med Chem ; 18(4): 497-512, 2011.
Article in English | MEDLINE | ID: mdl-21143114

ABSTRACT

The seven-spanning transmembrane G-protein coupled receptor CXCR4, which specifically binds to the chemokine CXCL12, is expressed on many cell types, including various types of tumour cells. CXCR4 plays a crucial role in organ-specific metastasis, directing migration of malignant cells expressing this receptor toward microenvironments where the cognate ligand is secreted. CXCL12 has a direct growth and survival-promoting effect for various cancer cells and enhances moreover tumour angiogenesis by recruiting endothelial progenitor cells to tumours. Drugs which modulate the CXCL12/CXCR4 axis are therefore promising candidates in anti-cancer therapies. CXCR4 is also a coreceptor for human immunodeficiency virus type 1 (HIV-1) X4 virus and, as such, plays an important role in virus entry into target cells. Hence, antiviral agents that bind to CXCR4 are expected to inhibit HIV-1 entry. Here we review the structure, mechanism of action and biological activity of the main CXCR4 antagonists (peptide inhibitors, non-peptide antagonists, neutralizing antibodies, modified analogues of CXCL12) and agonists (CXCL12 peptide analogues) and discuss the CXCL12/CXCR4 axis as an important target in development of anti-tumoral and anti-HIV-1 therapies.


Subject(s)
Chemokine CXCL12/agonists , HIV Infections/drug therapy , HIV-1 , Neoplasms/drug therapy , Receptors, CXCR4/agonists , Receptors, CXCR4/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Chemokine CXCL12/metabolism , Humans , Peptides/chemistry , Peptides/therapeutic use , Receptors, CXCR4/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use
11.
Steroids ; 74(13-14): 1015-24, 2009.
Article in English | MEDLINE | ID: mdl-19665469

ABSTRACT

Endometriosis, defined as the presence of endometrial glands and stroma at extra-uterine sites, is a gynecological condition that affects women of reproductive age. Consistent with its uterine origins, endometriotic lesions and resulting symptoms are hormonally responsive. To investigate Progesterone Receptor (PR)-based therapies, we measured physiological endpoints and gene expression in rat models of uterine cell estrogenic activity. Estrogen-induced ELT-3 rat leiomyoma cell proliferation was significantly inhibited by progesterone (P4), while the antiprogestin RU486 or the Selective PR Modulator (SPRM) asoprisnil, did not block proliferation. Stromal cell-derived factor-1 (SDF-1/Cxcl12) gene expression was induced by estrogen, and was repressed by the Selective Estrogen Receptor Modulators (SERMs), the antiestrogen ICI 182,780, and P4, but not by RU486 or the ERbeta-selective ligand ERB-041. In ELT-3 cells, asoprisnil demonstrated partial PR agonism on SDF-1 gene repression. Magnetic Resonance Imaging was used to monitor development of ectopic cysts in a rat surgical model of endometriosis. SERMs and P4 significantly decreased cyst volumes comparably by approximately 60%. However, ERB-041 and asoprisnil had no effect on cyst volume, and RU486 increased cyst volume by 20%. SDF-1 expression was modestly, but significantly, increased in the cyst compared to eutopic uterus, and P4 and raloxifene could repress the expression. We showed that SDF-1 was similarly regulated in human cells. These data suggest that transcriptional regulation of SDF-1 is a surrogate marker of estrogenic activities via ERalpha in rat uterine cells, and that SDF-1 repression by PR agonists can predict the ability to oppose the actions of estrogen in vivo.


Subject(s)
Chemokine CXCL12/antagonists & inhibitors , Endometriosis/drug therapy , Progesterone/therapeutic use , Progestins/therapeutic use , Receptors, Progesterone/agonists , Selective Estrogen Receptor Modulators/therapeutic use , Uterus/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL12/agonists , Chemokine CXCL12/metabolism , Cysts/drug therapy , Cysts/metabolism , Cysts/pathology , Disease Models, Animal , Endometriosis/metabolism , Endometriosis/pathology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrenes/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Female , Fulvestrant , Gene Expression , Hormone Antagonists/pharmacology , Humans , Mifepristone/pharmacology , Oxazoles/pharmacology , Oximes/pharmacology , Progesterone/pharmacology , Progestins/pharmacology , Rats , Selective Estrogen Receptor Modulators/pharmacology , Uterus/metabolism
12.
BMC Immunol ; 9: 26, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18534032

ABSTRACT

BACKGROUND: Activation of integrins in response to inside-out signaling serves as a basis for leukocyte arrest on endothelium, and migration of immune cells. Integrin-dependent adhesion is controlled by the conformational state of the molecule (i.e. change in the affinity for the ligand and molecular unbending (extension)), which is regulated by seven-transmembrane Guanine nucleotide binding Protein-Coupled Receptors (GPCRs). alpha4beta1-integrin (CD49d/CD29, Very Late Antigen-4, VLA-4) is expressed on leukocytes, hematopoietic stem cells, hematopoietic cancer cells, and others. Affinity and extension of VLA-4 are both rapidly up-regulated by inside-out signaling through several Galphai-coupled GPCRs. The goal of the current report was to study the effect of Galphas-coupled GPCRs upon integrin activation. RESULTS: Using real-time fluorescent ligand binding to assess affinity and a FRET based assay to probe alpha4beta1-integrin unbending, we show that two Galphas-coupled GPCRs (H2-histamine receptor and beta2-adrenergic receptor) as well as several cAMP agonists can rapidly down modulate the affinity of VLA-4 activated through two Galphai-coupled receptors (CXCR4 and FPR) in U937 cells and primary human peripheral blood monocytes. This down-modulation can be blocked by receptor-specific antagonists. The Galphas-induced responses were not associated with changes in the expression level of the Galphai-coupled receptors. In contrast, the molecular unbending of VLA-4 was not significantly affected by Galphas-coupled GPCR signaling. In a VLA-4/VCAM-1-specific myeloid cell adhesion system, inhibition of the VLA-4 affinity change by Galphas-coupled GPCR had a statistically significant effect upon cell aggregation. CONCLUSION: We conclude that Galphas-coupled GPCRs can rapidly down modulate the affinity state of VLA-4 binding pocket through a cAMP dependent pathway. This plays an essential role in the regulation of cell adhesion. We discuss several possible implications of this described phenomenon.


Subject(s)
Cell Adhesion/immunology , Integrin alpha4beta1/chemistry , Integrin alpha4beta1/metabolism , Receptor Cross-Talk/immunology , Signal Transduction , Adrenergic beta-Agonists/pharmacology , Cell Adhesion/drug effects , Chemokine CXCL12/agonists , Chemokine CXCL12/pharmacology , Down-Regulation , Histamine Agonists/pharmacology , Humans , Integrin alpha4beta1/immunology , Isoproterenol/pharmacology , Leukocytes, Mononuclear , Oligopeptides/pharmacology , Phenylurea Compounds/pharmacology , Protein Binding/drug effects , Protein Conformation , Receptors, Adrenergic, beta-2/immunology , Receptors, Adrenergic, beta-2/metabolism , Receptors, CXCR4/agonists , Receptors, Histamine H2/immunology , Receptors, Histamine H2/metabolism , Recombinant Proteins/agonists , Thiazoles/pharmacology , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...