Lymphocyte cytotoxicity is essential in immune defense. In this issue of Blood, Kurowska and colleagues define a Rab27a/Slp3/kinesin-1 complex that facilitates anterograde microtubule transport of lytic granules, representing a critical step in lymphocyte granule exocytosis and cytotoxicity.1 

Cytotoxic lymphocytes, encompassing cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells, kill pathogen-infected neoplastic as well as certain activated hematopoietic cells through targeted release of perforin-containing lytic granules. Autosomal recessive mutations in several proteins required for cytotoxic lymphocyte granule exocytosis have been associated with fatal diseases.2  The small GTPase Rab27a is one such protein. Rab27a-deficient patients typically present with hyperinflammatory hemophagocytic syndromes due to defects in lytic granule exocytosis, and also display partial albinism owing to a role for Rab27a in the trafficking and secretion of pigment granules from melanocytes.3 

Tug-of-war for cytotoxicity: lytic granule movement at cytotoxic lymphocyte immune synapses. First, engagement of activating receptors induces dynein-dependent retrograde transport of lytic granules on microtubles toward the MTOC, followed by movement of the MTOC with clustered granules to the immune synapse. Second, a novel kinesin-1/Slp3/Rab27a complex mediates anterograde transport of lytic granules away from the MTOC and toward the plasma membrane, before final myosin-dependent exocytosis of granules at the immune synapse.

Tug-of-war for cytotoxicity: lytic granule movement at cytotoxic lymphocyte immune synapses. First, engagement of activating receptors induces dynein-dependent retrograde transport of lytic granules on microtubles toward the MTOC, followed by movement of the MTOC with clustered granules to the immune synapse. Second, a novel kinesin-1/Slp3/Rab27a complex mediates anterograde transport of lytic granules away from the MTOC and toward the plasma membrane, before final myosin-dependent exocytosis of granules at the immune synapse.

Close modal

On target cell recognition by cytotoxic lymphocytes, the prevailing view is that lytic granules first undergo retrograde transport along microtubules to the microtubule organizing center (MTOC).4  Such minus end–directed transport is mediated by the motor protein dynein. Once clustered at the MTOC, granules are then moved toward the interface formed with the target cell, an area termed the immune synapse. This is then followed by a final stage of myosin IIA–dependent movement on actin, facilitating plasma membrane fusion.5  However, almost 20 years ago, Burkhardt and colleagues demonstrated that when lytic granules were isolated and allowed to interact with microtubules in the presence of motor protein–containing supernatants in vitro, movement was strongly biased toward anterograde transport.6  Such anterograde transport was inhibited by antibody-mediated depletion of kinesin. Conventional kinesins, as opposed to dyneins, are motor proteins that mediate anterograde transport of cargo along microtubules and consist of a heterotetramer of 2 heavy chains with motor activity bound to 2 light chains. A role for kinesins and anterograde transport of lytic granules in lymphocyte cytotoxicty has not been investigated.

Prompted by findings in neurons, where Rab27b forms a complex with the synaptotagmin- like protein (Slp)–1 as well as kinesin-1 to facilitate anterograde transport of vesicles,7  Kurowska et al examined the expression of Slps and kinesin subunits in human CTLs.1  At the transcript level, they established that Slp1 and Slp3 represent the major Slp isoforms in CTLs, and that the KIF5B heavy chain and KLC1 light chain are the predominant kinesin-1 subunits. Rab27a can bind multiple effectors, including Slp1, Slp2a, Slp3, Slac2a, and Slac2b.8  Coimmunoprecipitation experiments revealed that KLC1 interacts with Slp3. The interaction was mapped to a linker region on Slp3 that binds the C-terminal tetratricopeptide repeat (TRP) domain of KLC1. Importantly, involvement of kinesin-1 in lymphocyte cytotoxicity was established through the impairment of granule exocytosis on transfection of a KIF5B dominant-negative construct, knockdown of KIF5B using siRNA, and overexpression of the cognate Slp3 linker region. Notably, none of these manipulations impaired granule polarization to the immune synapse. Together, these data reveal that the Rab27a/Slp3/kinesin-1 complex is required for final movement of lytic granules at the immune synapse for exocytosis. Consistent with these data, live cell imaging revealed that lytic granule movement at the plasma membrane is both microtubule- and actin-dependent, whereas movement in the cytoplasm is only dependent on microtubules.9  In this setting, Rab27a-deficiency resulted in a reduced fraction and less mobility of lytic granules at the plasma membrane, concomitant with increased mobility of lytic granules in the cytosol.

Interestingly, Slp1 and Slp2a are also expressed in CTLs and have been implicated in lytic granule exocytosis.10  Kurowska and colleagues found that overexpression of either Slp1 or Slp3 inhibited lytic granule exocytosis, whereas co-overexpression of Rab27a restored exocytosis in the context of Slp3, but not Slp1. This suggests distinct regulation of lytic granule secretion by different Rab27a-Slp complexes. Thus, sequential interactions of different Slp effectors may intricately regulate lytic granule trafficking and exocytosis at the plasma membrane. Further work is required to clarify these issues as well as how granules are transferred to actin filaments.

The findings by Kurowska and coworkers provide further insights into the intriguing biology of lymphocyte cytotoxicity. After dynein-dependent retrograde microtubule transport of granules to the MTOC and movement of the MTOC to the immune synapse, they propose a switch to kinesin-1–dependent anterograde microtubule transport to position granules near the plasma membrane (see figure), before final myosin IIA–driven movement on actin for plasma membrane fusion. The Rab27a/Slp3/kinesin-1 complex is clearly a critical facilitator of lymphocyte cytotoxicity, as Rab27a deficiency abrogates granule exocytosis, causing immunodeficiency. Notably, the pattern of granule exocytosis at cytotoxic lymphocyte immune synapses may depend on the nature of receptors engaged,11  suggesting different modes of spatio-temporal regulation of lytic granule trafficking and fusion by distinct receptors. Thus, further experiments are needed to address how dynein and kinesin motor proteins are recruited to regulate the tug-of-war facilitating granule movement along microtubules.

In conclusion, a critical molecular complex for cytotoxic lymphocyte granule movement and exocytosis has been identified. Besides Rab27a, mutations in the gene encoding Slp3, SYTL3, could be associated with immunodeficiencies. Further work will hopefully address how different motor proteins for lymphocyte cytotoxicity are regulated spatially and temporally.

Conflict-of-interest disclosure: The authors declare no competing financial interests. ■

1
Kurowska
 
M
Goudin
 
N
Nehme
 
NT
, et al. 
Terminal transport of lytic granules to the immune synapse is mediated by the kinesin-1/Slp3/Rab27a complex.
Blood
2012
, vol. 
119
 
17
(pg. 
3879
-
3889
)
2
Pachlopnik Schmid
 
J
Cote
 
M
Menager
 
MM
, et al. 
Inherited defects in lymphocyte cytotoxic activity.
Immunol Rev
2010
, vol. 
235
 
1
(pg. 
10
-
23
)
3
Menasche
 
G
Pastural
 
E
Feldmann
 
J
, et al. 
Mutations in RAB27A cause Griscelli syndrome associated with haemophagocytic syndrome.
Nat Genet
2000
, vol. 
25
 
2
(pg. 
173
-
176
)
4
Stinchcombe
 
JC
Majorovits
 
E
Bossi
 
G
Fuller
 
S
Griffiths
 
GM
Centrosome polarization delivers secretory granules to the immunological synapse.
Nature
2006
, vol. 
443
 
7110
(pg. 
462
-
465
)
5
Andzelm
 
MM
Chen
 
X
Krzewski
 
K
Orange
 
JS
Strominger
 
JL
Myosin IIA is required for cytolytic granule exocytosis in human NK cells.
J Exp Med
2007
, vol. 
204
 
10
(pg. 
2285
-
2291
)
6
Burkhardt
 
JK
McIlvain
 
JM
Sheetz
 
MP
Argon
 
Y
Lytic granules from cytotoxic T cells exhibit kinesin-dependent motility on microtubules in vitro.
J Cell Sci
1993
, vol. 
104
 
Pt 1
(pg. 
151
-
162
)
7
Arimura
 
N
Kimura
 
T
Nakamuta
 
S
, et al. 
Anterograde transport of TrkB in axons is mediated by direct interaction with Slp1 and Rab27.
Dev Cell
2009
, vol. 
16
 
5
(pg. 
675
-
686
)
8
Kuroda
 
TS
Fukuda
 
M
Ariga
 
H
Mikoshiba
 
K
The Slp homology domain of synaptotagmin-like proteins 1-4 and Slac2 functions as a novel Rab27A binding domain.
J Biol Chem
2002
, vol. 
277
 
11
(pg. 
9212
-
9218
)
9
Liu
 
D
Meckel
 
T
Long
 
EO
Distinct role of rab27a in granule movement at the plasma membrane and in the cytosol of NK cells.
PloS One
2010
, vol. 
5
 
9
pg. 
e12870
 
10
Holt
 
O
Kanno
 
E
Bossi
 
G
, et al. 
Slp1 and Slp2-a localize to the plasma membrane of CTL and contribute to secretion from the immunological synapse.
Traffic
2008
, vol. 
9
 
4
(pg. 
446
-
457
)
11
Liu
 
D
Bryceson
 
YT
Meckel
 
T
Vasiliver-Shamis
 
G
Dustin
 
ML
Long
 
EO
Integrin-dependent organization and bidirectional vesicular traffic at cytotoxic immune synapses.
Immunity
2009
, vol. 
31
 
1
(pg. 
99
-
109
)
Sign in via your Institution