To the editor:

Integrins constitute the major and largest cell adhesion receptor family. These heterodimers undergo dramatic allosteric conformational changes in response to various activation signals acting on their beta integrin subunit tails.1  In hematopoietic cells, these events are critical for platelet aggregation, firm leukocyte adhesiveness to vascular endothelium, and immune synapse formation. Talin1 is the primary cytoskeletal regulator of integrin activation.2  Recent evidence suggests that Kindlins,3  3 structurally related adaptors, cooperate with talin in activating integrins in different cell types.4,5  In contrast to Kindlins1 and 2, Kindlin-3 (FERMT3) expression is restricted to the hematopoietic system and is particularly high in megakaryocytes and spleen.4  Unlike loss of talin1, which is embryonically lethal, deletion of individual Kindlin family members is not, but dramatically affects integrin activation in different hematopoietic cells. Deletion of Kindlin-3 in mice results in severe defects in platelet β3 and β1 integrin activation and osteopetrosis whereas integrin function in non hemtaopoietic cells remained conserved.4 

Upstream of talins and Kindlins, integrin activation is tightly controlled by the Ras-related guanosine triphosphatase (GTPase), Rap-1.6  Deletion of the Rap-1 guanine exchange factor (GEF), CalDAG-GEFI (CDGI, Ras guanine nucleotide releasing protein-2 [RASGRP2]) results in major defects in platelet integrin activation7  strikingly similar to the defects associated with Kindlin-3 deletion.4  CDGI and Rap-1 also control inside out integrin activation in leukocytes.8,9  We have identified impaired expression of CDGI in human hematopoietic cells, a result of a homozygous splice junction mutation in exon 16, which led to a rare autosomal recessive leukocyte adhesion deficiency syndrome, LAD-III, a combined defect in β3, β2 and β1 integrin activation in platelets, neutrophils and lymphocytes.10  These LAD-III cases also suffered from severe osteopetrosis (Kilic, submitted). All of these LAD-III patients were of Turkish origin, children of heterozygote carriers of the same exon 16 CDGI mutation (10  and not shown). Two patients died at age 3 to 6 years and a third infant underwent bone marrow transplantation.

In light of the striking similarity between CDGI and Kindlin-3 knock out platelet dysfunction,4,7  we sequenced Kindlin-3 in our 3 CDGI defective LAD-III cases. Genetic sequencing of Kindlin-3 revealed a homozygous stop codon in R513 (CGA > TGA) of Kindlin-3 in all 3 patients (Figure 1 and Document S1, available on the Blood website; see the Supplemental Materials link at the top of the online article). The patients' parents were all confirmed to be heterozygous carriers of this mutation, which was absent in all 68 control chromosomes tested from Turkish donors (Figure 1 and data not shown). This mutation alters the F3 PTB–containing integrin binding subdomain of Kindlin-3. Notably, Kindlin-3 is expressed in the same locus of CDGI (11q13) and the 2 genes are 503 029 bp apart, whereas other Kindlins are expressed on other chromosomes. None of the patients suffered from nonhematopoietic abnormalities associated with other Kindlin deficiencies.

Figure 1

Multiple alignment of genomic DNA sequence surrounding the putative stop codon mutation within the Kindlin3 F3 region. The reference genome (NCBI human genome database build 36.2, chr11: 63744678-63744716), the mRNA (RefSeq: NM_178443.1), and genomic sequences from one normal control, one parent of a LADIII patient (LAD-A), and 3 LADIII patients (LAD-A, LAD-K, and LAD-N) were aligned surrounding the mutation nt1632c > t. The base of the mutation is highlighted.

Figure 1

Multiple alignment of genomic DNA sequence surrounding the putative stop codon mutation within the Kindlin3 F3 region. The reference genome (NCBI human genome database build 36.2, chr11: 63744678-63744716), the mRNA (RefSeq: NM_178443.1), and genomic sequences from one normal control, one parent of a LADIII patient (LAD-A), and 3 LADIII patients (LAD-A, LAD-K, and LAD-N) were aligned surrounding the mutation nt1632c > t. The base of the mutation is highlighted.

Close modal

Taken together, the LAD-III phenotype reported by us could manifest a combined defect in both upstream and downstream integrin regulatory effectors, namely, CalDAG-GEFI and Kindlin-3, respectively. Our findings predict a potential role for Kindlin-3 in regulating integrin activation in both platelets and leukocytes. Although extremely rare, the presence of identical mutations in 2 functional genes within the same locus in 3 Turkish families may suggest a common ancestor allele carrying both of these mutations. To our knowledge this is a first report of multiple mutations in 2 separate genes which function within a similar signaling axis in a key biologic process. Additional LAD-III cases can serve as ideal model systems to address the cooperative roles of CDGI Rap-1 signaling, talin1, and Kindlin-3 in integrin activation on different hematopoietic cell types.

Approval was obtained from the Rambam Medical Center institutional review board for these studies. Informed consent was obtained in accordance with the Declaration of Helsinki.

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

Correspondence: Amos Etzioni, Rambam Medical Center, Faculty of Medicine, Technion, Haifa, Israel; e-mail: etzioni@rambam.health.gov.il; or Ronen Alon, Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel; e-mail: ronen.alon@weizmann.ac.il.

1
Hynes
 
RO
Integrins: bidirectional, allosteric signaling machines.
Cell
2002
, vol. 
110
 (pg. 
673
-
687
)
2
Critchley
 
DR
Gingras
 
AR
Talin at a glance.
J Cell Sci
2008
, vol. 
121
 (pg. 
1345
-
1347
)
3
Ussar
 
S
Wang
 
HV
Linder
 
S
Fassler
 
R
Moser
 
M
The Kindlins: subcellular localization and expression during murine development.
Exp Cell Res
2006
, vol. 
312
 (pg. 
3142
-
3151
)
4
Moser
 
M
Nieswandt
 
B
Ussar
 
S
Pozgajova
 
M
Fassler
 
R
Kindlin-3 is essential for integrin activation and platelet aggregation.
Nat Med
2008
, vol. 
14
 (pg. 
325
-
330
)
5
Montanez
 
E
Ussar
 
S
Schifferer
 
M
, et al. 
Kindlin-2 controls bidirectional signaling of integrins.
Genes Dev
2008
, vol. 
22
 (pg. 
1325
-
1330
)
6
Kinashi
 
T
Intracellular signalling controlling integrin activation in lymphocytes.
Nat Rev Immunol
2005
, vol. 
5
 (pg. 
546
-
559
)
7
Crittenden
 
JR
Bergmeier
 
W
Zhang
 
Y
, et al. 
CalDAG-GEFI integrates signaling for platelet aggregation and thrombus formation.
Nat Med
2004
, vol. 
10
 (pg. 
982
-
986
)
8
Bergmeier
 
W
Goerge
 
T
Wang
 
H
, et al. 
Mice lacking the signaling molecule CalDAG-GEFI represent a model for leukocyte adhesion deficiency type III.
J Clin Invest
2007
, vol. 
117
 (pg. 
1699
-
1707
)
9
Ghandour
 
H
Cullere
 
X
Alvarez
 
A
Luscinskas
 
FW
Mayadas
 
TN
Essential role for Rap1 GTPase and its guanine exchange factor CalDAG-GEFI in LFA-1 but not VLA-4 integrin mediated human T-cell adhesion.
Blood
2007
, vol. 
110
 (pg. 
3682
-
3690
)
10
Pasvolsky
 
R
Feigelson
 
SW
Kilic
 
SS
, et al. 
A LAD-III syndrome is associated with defective expression of the Rap-1 activator CalDAG-GEFI in lymphocytes, neutrophils, and platelets.
J Exp Med
2007
, vol. 
204
 (pg. 
1571
-
1582
)
Sign in via your Institution