scholarly journals Distinct Role of ZAP-70 and Src Homology 2 Domain-Containing Leukocyte Protein of 76 kDa in the Prolonged Activation of Extracellular Signal-Regulated Protein Kinase by the Stromal Cell-Derived Factor-1α/CXCL12 Chemokine

2003 ◽  
Vol 171 (1) ◽  
pp. 360-367 ◽  
Author(s):  
Kimberly N. Kremer ◽  
Troy D. Humphreys ◽  
Ashok Kumar ◽  
Nan-Xin Qian ◽  
Karen E. Hedin
2001 ◽  
Vol 360 (1) ◽  
pp. 57-66 ◽  
Author(s):  
Michael D. SCHALLER ◽  
Erik M. SCHAEFER

Paxillin is a focal-adhesion-associated, tyrosine-phosphorylated protein. In cells transformed by the src, crk or BCR-Abl oncogenes, the phosphotyrosine content of paxillin is elevated. In normal cells paxillin functions in signalling following integrin-dependent cell adhesion or exposure to a number of stimuli, including growth factors and neuropeptides. These stimuli induce tyrosine phosphorylation of paxillin, regulating the association of Src homology 2 domain-containing signalling molecules with paxillin. There are multiple sites of tyrosine phosphorylation on paxillin. To elucidate the role of paxillin in transducing signals in response to various stimuli, it is essential to identify all of the sites of phosphorylation on paxillin and to define which residues are phosphorylated in response to distinct stimuli. We describe two new sites of tyrosine phosphorylation on paxillin, residues at positions 40 and 88. Using paxillin variants with phenylalanine substitutions at phosphorylation sites and phospho-specific paxillin antibodies, tyrosine phosphorylation of paxillin in response to distinct stimuli was examined. The results demonstrate that Tyr31 and Tyr118, which are binding sites for Crk, are major sites of tyrosine phosphorylation following cell adhesion or stimulation with platelet-derived growth factor or angiotensin II. Thus multiple stimuli may elicit similar signalling events downstream of paxillin.


2000 ◽  
Vol 193 (1) ◽  
pp. 61-72 ◽  
Author(s):  
Dianne Cox ◽  
Benjamin M. Dale ◽  
Masaki Kashiwada ◽  
Cheryl D. Helgason ◽  
Steven Greenberg

The Src homology 2 domain–containing inositol 5′-phosphatase (SHIP) is recruited to immunoreceptor tyrosine-based inhibition motif (ITIM)–containing proteins, thereby suppressing phosphatidylinositol 3-kinase (PI 3-kinase)–dependent pathways. The role of SHIP in phagocytosis, a PI 3-kinase–dependent pathway, is unknown. Overexpression of SHIP in macrophages led to an inhibition of phagocytosis mediated by receptors for the Fc portion of IgG (FcγRs). In contrast, macrophages expressing catalytically inactive SHIP or lacking SHIP expression demonstrated enhanced phagocytosis. To determine whether SHIP regulates phagocytosis mediated by receptors that are not known to recruit ITIMs, we determined the effect of SHIP expression on complement receptor 3 (CR3; CD11b/CD18; αMβ2)–dependent phagocytosis. Macrophages overexpressing SHIP demonstrated impaired CR3-mediated phagocytosis, whereas macrophages expressing catalytically inactive SHIP demonstrated enhanced phagocytosis. CR3-mediated phagocytosis in macrophages derived from SHIP−/− mice was up to 2.5 times as efficient as that observed in macrophages derived from littermate controls. SHIP was localized to FcγR- and CR3-containing phagocytic cups and was recruited to the cytoskeleton upon clustering of CR3. In a transfected COS cell model of activation-independent CR3-mediated phagocytosis, catalytically active but not inactive SHIP also inhibited phagocytosis. We conclude that PI 3-kinase(s) and SHIP regulate multiple forms of phagocytosis and that endogenous SHIP plays a role in modulating β2 integrin outside-in signaling.


Blood ◽  
2002 ◽  
Vol 99 (12) ◽  
pp. 4318-4325 ◽  
Author(s):  
Marit Inngjerdingen ◽  
Knut Martin Torgersen ◽  
Azzam A. Maghazachi

Stromal cell–derived factor 1α (CXCL12) induces chemotaxis of lymphocytes through its receptor CXCR4. We examined the role of nonreceptor tyrosine kinases in CXCL12-induced chemotaxis of T cells and natural killer (NK) cells. Damnacanthal, a specific Lck inhibitor, but not the Syk inhibitor piceatannol, inhibited CXCL12-induced chemotaxis of both lymphocyte subsets. Similarly, damnacanthal was shown to inhibit CXCL12-induced chemotaxis of the Jurkat T-cell line. Stimulating T and NK cells with CXCL12 increased both the tyrosine phosphorylation and the kinase activity of Lck. A direct involvement of Lck in CXCL12-induced chemotaxis was demonstrated in the Lck-deficient Jurkat-derived cell line JCaM1.6. Although JCaM1.6 cells express CXCR4, no significant migration was detected after CXCL12 stimulation. Reconstitution with wild-type Lck restored both CXCL12-induced chemotaxis and Lck activation. Furthermore, cotransfection of wild-type Lck with C-terminal Src kinase (Csk) into JCaM1.6 failed to restore the chemotactic response induced by CXCL12. Finally, by targeting critical residues in the Src homology–2 (SH2) or SH3 domains of Lck, we observed that the SH3 domain is important for the function of Lck in CXCL12-mediated chemotaxis. Together, these results suggest a role for Lck in CXCL12-induced signaling pathways leading to lymphocyte chemotaxis.


Sign in / Sign up

Export Citation Format

Share Document