We are grateful to Drs Yipp and Kubes for their attention to our recent work and for adding an important new piece to the puzzle of Ly6G and its function in neutrophil-mediated immunity.1,2 

We find no incompatibility between their observations and our own. As is well recognized, the pathways mediating neutrophil migration differ with site and stimulus. In particular, whereas LFA-1 is strictly required for neutrophil migration at the initiation phase of K/BxN arthritis,3,4  other pathways can often compensate for β2 integrin deficiency in the context of bacterial infection.5,6  Because we found that Ly6G ligation attenuated β2 integrin–dependent migration, but not β2 integrin–independent migration,2  it is not clear that an effect would have been expected in the Staphylococcus aureus cellulitis model used by Drs Yipp and Kubes.

However, we recognize that divergent dependence on β2 integrins cannot be the whole story. The Kubes laboratory has employed conjugated anti–Gr-1 as an in vivo tool in several important studies using models of sterile inflammation in which β2 integrins play a demonstrable role.6,7  Further investigation will be required to determine the relevant differences between these experimental systems and those used in our work.

We therefore interpret the findings of Drs Yipp and Kubes as complementary to, rather than in conflict with, our own. Together, these studies raise new and interesting questions. What are the conditions under which Ly6G-binding ligation alters migration? What is the role of Ly6G in the normal course of neutrophil physiology? What does this role of Ly6G tell us about human neutrophil biology? We look forward to working with the Kubes laboratory and others to answer these important questions.

Acknowledgments: This work was supported by grants from the Arthritis National Research Foundation, the Charles H. Hood Foundation, the Harvard Skin Disease Research Center, and the Cogan Family Foundation (to P.A.N).

Contribution: P.A.N. wrote the response, which was circulated among all coauthors of the original article to incorporate their input and secure their approval.

Conflict-of-interest disclosure: The author declares no competing financial interests.

Correspondence: Peter A. Nigrovic, MD, Brigham and Women's Hospital, Division of Rheumatology, Immunology and Allergy, Smith Bldg, Rm 516c, One Jimmy Fund Way, Boston, MA 02115; e-mail: pnigrovic@partners.org.

1
Yipp
 
BG
Kubes
 
P
Antibodies against neutrophil Ly6G do not inhibit leukocyte recruitment in mice in vivo.
Blood
2013
, vol. 
121
 
1
pg. 
239
 
2
Wang
 
JX
Bair
 
AM
King
 
SL
, et al. 
Ly6G ligation blocks recruitment of neutrophils via a beta2-integrin-dependent mechanism.
Blood
2012
, vol. 
120
 
7
(pg. 
1489
-
1498
)
3
Watts
 
GM
Beurskens
 
FJ
Martin-Padura
 
I
, et al. 
Manifestations of inflammatory arthritis are critically dependent on LFA-1.
J Immunol
2005
, vol. 
174
 
6
(pg. 
3668
-
3675
)
4
Monach
 
PA
Nigrovic
 
PA
Chen
 
M
, et al. 
Neutrophils in a mouse model of autoantibody-mediated arthritis: critical producers of Fc receptor gamma, the receptor for C5a, and lymphocyte function-associated antigen 1.
Arthritis Rheum
2010
, vol. 
62
 
3
(pg. 
753
-
764
)
5
Mizgerd
 
JP
Kubo
 
H
Kutkoski
 
GJ
, et al. 
Neutrophil emigration in the skin, lungs, and peritoneum: different requirements for CD11/CD18 revealed by CD18-deficient mice.
J Exp Med
1997
, vol. 
186
 
8
(pg. 
1357
-
1364
)
6
McDonald
 
B
Pittman
 
K
Menezes
 
GB
, et al. 
Intravascular danger signals guide neutrophils to sites of sterile inflammation.
Science
2010
, vol. 
330
 
6002
(pg. 
362
-
366
)
7
Phillipson
 
M
Heit
 
B
Colarusso
 
P
Liu
 
L
Ballantyne
 
CM
Kubes
 
P
Intraluminal crawling of neutrophils to emigration sites: a molecularly distinct process from adhesion in the recruitment cascade.
J Exp Med
2006
, vol. 
203
 
12
(pg. 
2569
-
2575
)
Sign in via your Institution