Download Immunodeficiency Agammaglobulinemia, the First Primary

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Public health genomics wikipedia , lookup

Polycomb Group Proteins and Cancer wikipedia , lookup

Epigenetics in stem-cell differentiation wikipedia , lookup

Microevolution wikipedia , lookup

Artificial gene synthesis wikipedia , lookup

Gene therapy wikipedia , lookup

Point mutation wikipedia , lookup

Neuronal ceroid lipofuscinosis wikipedia , lookup

Site-specific recombinase technology wikipedia , lookup

Designer baby wikipedia , lookup

Mir-92 microRNA precursor family wikipedia , lookup

Vectors in gene therapy wikipedia , lookup

Gene therapy of the human retina wikipedia , lookup

NEDD9 wikipedia , lookup

Transcript
This information is current as
of June 18, 2017.
Colonel Bruton's Kinase Defined the
Molecular Basis of X-Linked
Agammaglobulinemia, the First Primary
Immunodeficiency
Wasif N. Khan
J Immunol 2012; 188:2933-2935; ;
doi: 10.4049/jimmunol.1200490
http://www.jimmunol.org/content/188/7/2933
Subscription
Permissions
Email Alerts
This article cites 41 articles, 14 of which you can access for free at:
http://www.jimmunol.org/content/188/7/2933.full#ref-list-1
Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscription
Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts
The Journal of Immunology is published twice each month by
The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2012 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
Downloaded from http://www.jimmunol.org/ by guest on June 18, 2017
References
Colonel Bruton’s Kinase Defined the Molecular Basis of
X-Linked Agammaglobulinemia, the First
Primary Immunodeficiency
Wasif N. Khan
T
Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33134
Address correspondence and reprint requests to Dr. Wasif N. Khan, Department of
Microbiology and Immunology, Miller School of Medicine, University of Miami,
1600 NW 10th Avenue, 3347A Rosenstiel Medical Science Building, Miami, FL
33134. E-mail address: [email protected]
Abbreviations used in this article: PH, pleckstrin homology; PID, primary immunodeficiency disease; PLC, phospholipase C; TI, T cell independent; XLA, X-linked
agammaglobulinemia.
Copyright Ó 2012 by The American Association of Immunologists, Inc. 0022-1767/12/$16.00
www.jimmunol.org/cgi/doi/10.4049/jimmunol.1200490
the B cell lineage was the source of Igs and that T cells regulated delayed-type hypersensitivity and graft rejection.
Cellular characterization of peripheral blood revealed that
XLA patients have very few or virtually no circulating B cells (3,
7). The peripheral B cell deficiency was found to be due to
a developmental block at the pre-B (CD191cytoplasmic m1)
cell stage in the bone marrow (7–11). However, this defect is
leaky, yielding a few IgM1 immature B cells, which fail to
develop into fully mature B lymphocytes (3, 4). Together,
these studies traced the Ig deficiency to B cell developmental
defects in the bone marrow (pre-B) and in immature B cells in
the periphery. These cellular defects suggested that the protein
encoded by the XLA gene was expressed and critical for B cell
differentiation at multiple stages, from pre-B to plasma cells.
The observation that XLA exclusively affected males indicated that the defective gene was localized to the X chromosome and that the hemizygosity of the X chromosome in males
resulted in expression of the disease phenotype. Because of the
random nature of X-chromosome inactivation in females, Xlinked genes display mosaic expression. The X-linked nature of
the defect was confirmed by X-chromosome inactivation in
XLA patients; only B cells displayed a nonrandom X inactivation (12). Refined gene mapping localized the XLA genetic
defect to the Xq22 region (13–15). Finally, the Bentley group
(16), taking advantage of technical advances in positional
cloning, identified genomic clones spanning the XLA locus
and analyzed the corresponding transcripts for mutations and
disease association. The DXS178 polymorphic DNA marker
was found to be closely linked, and a clone spanning this
marker yielded a transcript that corresponded to a region
altered in all members of an XLA family. The transcript
encoded a novel cytoplasmic tyrosine kinase and was termed
Atk (16). The Witte group, while searching for novel tyrosine
kinases involved in B cell development, identified a cDNA
clone that also mapped close to the DXS178 marker. This
clone encoded a tyrosine kinase and was termed bpk (17).
The findings of the two groups were later reconciled, and the
causative gene for XLA was named Bruton’s agammaglobulinemia tyrosine kinase (BTK ). The seminal research articles
by Vetrie et. al. (16) and Tsukada et. al. (17) describing the
underlying genetic defect in XLA are selected for the Pillars of
Immunology series for this issue of The Journal of Immunology.
Within 1 y of BTK ’s discovery, the Xid locus in the CBA/
N mouse stain was mapped to the Btk gene (18, 19). A naturally occurring mutation (R28C) was found in the pleckstrin homology (PH) domain (see below) of the Btk gene.
Downloaded from http://www.jimmunol.org/ by guest on June 18, 2017
he importance of pathogen-specific humoral immunity was recognized from the early days of immunology. However, that integrity of humoral immunity is
essential to protect against life-threatening infections was established with the discovery of the first primary immunodeficiency disease (PID). In the spring of 1952, Colonel Ogden C.
Bruton (1) reported an 8-y-old boy with extreme susceptibility to infections. He had suffered from $19 episodes of
clinical pneumococcal sepsis within the past 4 y. Electrophoretic analysis revealed a lack of gammaglobulins (IgG) in
the patient’s serum. Remarkably, furnishing IgG controlled
the life-threatening infections. This was the first description
of a PID and its IgG replacement therapy (1). Janeway and
colleagues’ observations with an additional five males corroborated Bruton’s findings (2). Based on the X-linked pattern
of inheritance, the disease later became known as X-linked
agammaglobulinemia (XLA), and IgG replacement remains
an effective treatment for XLA today. XLA, a prototype forB cell developmental defects and Ab deficiency, is the most
frequent PID and is responsible for disease in 85% of earlyonset Ab-deficient patients (3).
The XLA defect is primarily characterized by a failure to
clear infections, particularly those involving encapsulated
bacteria. This defect is accompanied by a severe reduction in
the basal serum Igs, as well as a failure to produce Ag-induced
Ab responses against infectious agents or immunizations (3, 4).
Thus, XLA is a selective Ab defect that leaves cellular immunity relatively intact. This phenotype suggested that the immune system is composed of specialized compartments for
humoral and cellular immune responses. These insights were
highly significant, because, at this time, the basis for cellular
and humoral immunity was not known. The selective Ab
defect in XLA contributed to the concept of specialized lymphocyte lineages and the discovery by Cooper et al. (5, 6) that
2934
activation of PLC-g2 provided a missing link between BCR
and the generation of lipid second messengers. Consistently,
BTK-deficient B cells are impaired in activating the protein
kinase C- and Ca21-regulated transcription factors NF-kB
and NFAT (31–35).
Recent studies demonstrate an involvement of BTK function in many signaling pathways in addition to that downstream of the BCR. These include pathways induced by
stimulation of cytokine receptors, G protein-coupled receptors, receptors of the TNFR family, and most TLRs (31,
36–38). Recent studies also suggest that TLR and BAFF
receptors play an important role in regulating TI Ab responses. It remains to be explored whether defective BTK
signaling in the TLR and BAFF receptor pathways contributes
to the defective TI immune responses in XLA and Xid. A role
for BTK in these signaling pathways is also relevant for immune tolerance and autoimmunity. Recently, a novel function of BTK in the full activation of TLR signaling was
described in which it forms a complex with nonconventional
endosomal MHC class II, CD40, and MyD88 (39). This type
of signaling in B cells can enhance B cell survival and activation and can play a role in TLR7/9-dependent development
of autoimmunity. The physiological significance of these
newly discovered BTK signaling mechanisms in B cell development, survival, and immune responses both in health and
autoimmune disease remains to be explored.
The identification of BTK has allowed detection of female
carriers of the defective XLA allele and corroboration of
clinical diagnosis of XLA patients with specific mutations in
the BTK gene. The discovery of BTK also has implications for
prenatal screening for the PIDs. Given that XLA is the most
common monogenetic immunodeficiency in humans, the potential for gene replacement and correction therapy needs
to be explored. Indeed, Btk gene replacement has been successful in mouse models; thus, human XLA gene therapy
remains a potential goal (40, 41). A successful human XLA
gene therapy may require precise genetic manipulations, such
as targeting the endogenous BTK locus in stem cells. The
origins of the clinical usefulness and potential for BTK gene
therapy lie in the discovery of XLA and the underlying defect
in the gene encoding Colonel Bruton’s kinase.
Acknowledgments
I thank Drs. Max D. Cooper and Emily S. Clark for helpful discussions
and critical reading of the manuscript.
Disclosures
The author has no financial conflicts of interest.
References
1. Bruton, O. C. 1952. Agammaglobulinemia. Pediatrics 9: 722–728.
2. Bruton, O. C., L. Apt, D. Gitlin, and C. A. Janeway. 1952. Absence of serum
gamma globulins. AMA Am. J. Dis. Child. 84: 632–636.
3. Conley, M. E. 1985. B cells in patients with X-linked agammaglobulinemia. J.
Immunol. 134: 3070–3074.
4. Conley, M. E., O. Parolini, J. Rohrer, and D. Campana. 1994. X-linked agammaglobulinemia: new approaches to old questions based on the identification of the
defective gene. Immunol. Rev. 138: 5–21.
5. Cooper, M. D., D. A. Raymond, R. D. Peterson, M. A. South, and R. A. Good.
1966. The functions of the thymus system and the bursa system in the chicken. J.
Exp. Med. 123: 75–102.
6. Cooper, M. D., R. D. Peterson, and R. A. Good. 1965. Delineation of the Thymic
and Bursal Lymphoid Systems in the Chicken. Nature 205: 143–146.
7. Cooper, M. D., and A. R. Lawton. 1972. Circulating B-cells in patients with
immunodeficiency. Am. J. Pathol. 69: 513–528.
Downloaded from http://www.jimmunol.org/ by guest on June 18, 2017
Soon thereafter, gene-targeted mice with a Btk-null mutation
unequivocally demonstrated that Xid is caused by a defect in
the Btk gene (20–22).The Xid mouse strain has been a model
for B cell and Ab immunodeficiency in mice since 1975 (23).
Although Xid mice display a much milder phenotype than do
XLA patients, perhaps due to a compensatory effect of the Tec
kinase (24), they do display B cell developmental defects at
the pro- to pre-B and the immature to mature B cell stages
(22, 23, 25, 26). Xid mice also displayed a severely defective
response to T cell-independent (TI) type 2 and a reduced
response to TI type 1 polysaccharide Ags (22, 23, 25, 26).
This observation strongly suggests that Btk function is required for TI immune responses in the mouse. Furthermore,
this defect is reminiscent of the unusual susceptibility of
XLA patients to encapsulated bacterial infections and suggests that this kinase is a critical signaling molecule in the
regulation of TI Ab responses both in mouse and man. It remains to be investigated whether the lack of mature B cells in
XLA or the lack of functional BTK in mature B cells is
responsible for the TI Ab response defect. A failure to produce
TI Ab responses is dangerous, because Abs facilitate complement-mediated lysis of bacteria and eliminate pathogens more
rapidly than does a delayed T cell-mediated immune response.
After the discovery that defects in the BTK gene caused
XLA, it was expected that it would be possible to correlate
gene mutations with the severity of disease phenotype, which
would allow a better understanding of the functional significance of the BTK protein domains. More than 800 mutations
have been identified that are scattered throughout the BTK
gene and include missense, nonsense, and insertion/deletion
mutations; however, no straightforward structure–function
correlation has been found (http://bioinf.uta.fi/BTKbase).
This is due, in part, to variability of the phenotype among
XLA patients, even those bearing the same mutation. Further,
different mutations differentially affect the stability of the
BTK protein, and studies in mice suggest that Btk protein
levels are critical for its function at distinct B cell developmental stages and during activation processes (27, 28).
Studies of BTK revealed that it belongs to a unique family
of cytoplasmic tyrosine kinases, termed the Tec family, and
plays a role in BCR signaling distinct from the Src family kinase
Lyn and the spleen tyrosine kinase Syk (29). The Tec kinases
have unique features, including a PH domain (mutated in Xid)
at the N terminus. Two of the Tec kinases, Tec and Itk, were
known at the time of BTK discovery. However, discovery of
the significance of BTK in a human disease brought this family
of kinases to the attention of the wider scientific community.
In addition to the PH domain, BTK contains a Tec homology
domain followed by SH3, SH2, and kinase domains. BTK
activation and function are intertwined with lipid metabolism
and calcium signaling (30). In response to BCR engagement,
BTK activation requires phosphorylation by the Src kinase
Lyn in the catalytic domain and membrane recruitment by
interaction of the PH domain with phosphatidylinositol 3,4,5triphosphate, a PI3K product. Active BTK autophosphorylates
its SH3 domain and ignites downstream pathways by directly
phosphorylating phospholipase C (PLC)-g2. Active PLC-g2
hydrolyzes phosphatidylinositol-4,5-bisphosphate into inositol
triphosphate and diacylglycerol, mobilizing intracellular Ca21
and activating protein kinase C and RasGRP (24, 31). BTK
PILLARS OF IMMUNOLOGY
The Journal of Immunology
24. Ellmeier, W., S. Jung, M. J. Sunshine, F. Hatam, Y. Xu, D. Baltimore, H. Mano,
and D. R. Littman. 2000. Severe B cell deficiency in mice lacking the tec kinase
family members Tec and Btk. J. Exp. Med. 192: 1611–1624.
25. Khan, W. N. 2001. Regulation of B lymphocyte development and activation by
Bruton’s tyrosine kinase. Immunol. Res. 23: 147–156.
26. Sideras, P., and C. I. Smith. 1995. Molecular and cellular aspects of
X-linked agammaglobulinemia. Adv. Immunol. 59: 135–223.
27. Broides, A., W. Yang, and M. E. Conley. 2006. Genotype/phenotype correlations
in X-linked agammaglobulinemia. Clin. Immunol. 118: 195–200.
28. Satterthwaite, A. B., F. Willis, P. Kanchanastit, D. Fruman, L. C. Cantley,
C. D. Helgason, R. K. Humphries, C. A. Lowell, M. Simon, M. Leitges, et al. 2000.
A sensitized genetic system for the analysis of murine B lymphocyte signal transduction pathways dependent on Bruton’s tyrosine kinase. Proc. Natl. Acad. Sci. USA
97: 6687–6692.
29. Lindvall, J. M., K. E. Blomberg, J. Väliaho, L. Vargas, J. E. Heinonen, A. Berglöf,
A. J. Mohamed, B. F. Nore, M. Vihinen, and C. I. Smith. 2005. Bruton’s tyrosine
kinase: cell biology, sequence conservation, mutation spectrum, siRNA modifications, and expression profiling. Immunol. Rev. 203: 200–215.
30. Rawlings, D. J. 1999. Bruton’s tyrosine kinase controls a sustained calcium signal
essential for B lineage development and function. Clin. Immunol. 91: 243–253.
31. Kurosaki, T. 2002. Regulation of B cell fates by BCR signaling components. Curr.
Opin. Immunol. 14: 341–347.
32. Antony, P., J. B. Petro, G. Carlesso, N. P. Shinners, J. Lowe, and W. N. Khan.
2003. B cell receptor directs the activation of NFAT and NF-kappaB via distinct
molecular mechanisms. Exp. Cell Res. 291: 11–24.
33. Hoek, K. L., P. Antony, J. Lowe, N. Shinners, B. Sarmah, S. R. Wente, D. Wang,
R. M. Gerstein, and W. N. Khan. 2006. Transitional B cell fate is associated with
developmental stage-specific regulation of diacylglycerol and calcium signaling upon
B cell receptor engagement. J. Immunol. 177: 5405–5413.
34. Bajpai, U. D., K. Zhang, M. Teutsch, R. Sen, and H. H. Wortis. 2000. Bruton’s
tyrosine kinase links the B cell receptor to nuclear factor kappaB activation. J. Exp.
Med. 191: 1735–1744.
35. Petro, J. B., S. M. Rahman, D. W. Ballard, and W. N. Khan. 2000. Bruton’s tyrosine kinase is required for activation of IkappaB kinase and nuclear factor kappaB
in response to B cell receptor engagement. J. Exp. Med. 191: 1745–1754.
36. Doyle, S. L., and L. A. O’Neill. 2006. Toll-like receptors: from the discovery of
NFkappaB to new insights into transcriptional regulations in innate immunity.
Biochem. Pharmacol. 72: 1102–1113.
37. Khan, W. N. 2009. B cell receptor and BAFF receptor signaling regulation of B cell
homeostasis. J. Immunol. 183: 3561–3567.
38. Qiu, Y., and H. J. Kung. 2000. Signaling network of the Btk family kinases. Oncogene 19: 5651–5661.
39. Liu, X., Z. Zhan, D. Li, L. Xu, F. Ma, P. Zhang, H. Yao, and X. Cao. 2011.
Intracellular MHC class II molecules promote TLR-triggered innate immune
responses by maintaining activation of the kinase Btk. Nat. Immunol. 12: 416–
424.
40. Kerns, H. M., B. Y. Ryu, B. V. Stirling, B. D. Sather, A. Astrakhan, S. HumbletBaron, D. Liggitt, and D. J. Rawlings. 2010. B cell-specific lentiviral gene therapy
leads to sustained B-cell functional recovery in a murine model of X-linked agammaglobulinemia. Blood 115: 2146–2155.
41. Ng, Y. Y., M. R. Baert, K. Pike-Overzet, M. Rodijk, M. H. Brugman,
A. Schambach, C. Baum, R. W. Hendriks, J. J. van Dongen, and F. J. Staal.
2010. Correction of B-cell development in Btk-deficient mice using lentiviral vectors with codon-optimized human BTK. Leukemia 24: 1617–1630.
Downloaded from http://www.jimmunol.org/ by guest on June 18, 2017
8. Geha, R. S., F. S. Rosen, and E. Merler. 1973. Identification and characterization of
subpopulations of lymphocytes in human peripheral blood after fractionation on
discontinuous gradients of albumin. The cellular defect in X-linked agammaglobulinemia. J. Clin. Invest. 52: 1726–1734.
9. Pearl, E. R., L. B. Vogler, A. J. Okos, W. M. Crist, A. R. Lawton, III, and
M. D. Cooper. 1978. B lymphocyte precursors in human bone marrow: an analysis
of normal individuals and patients with antibody-deficiency states. J. Immunol. 120:
1169–1175.
10. Siegal, F. P., B. Pernis, and H. G. Kunkel. 1971. Lymphocytes in human immunodeficiency states: a study of membrane-associated immunoglobulins. Eur. J.
Immunol. 1: 482–486.
11. Vogler, L. B., E. R. Pearl, W. E. Gathings, A. R. Lawton, and M. D. Cooper. 1976.
Letter: B lymphocyte precursors in bone-marrow in immunoglobulin deficiency
diseases. Lancet 2: 376.
12. Conley, M. E. 1992. Molecular approaches to analysis of X-linked immunodeficiencies. Annu. Rev. Immunol. 10: 215–238.
13. Kwan, S. P., L. Kunkel, G. Bruns, R. J. Wedgwood, S. Latt, and F. S. Rosen. 1986.
Mapping of the X-linked agammaglobulinemia locus by use of restriction fragmentlength polymorphism. J. Clin. Invest. 77: 649–652.
14. Lovering, R., H. R. Middleton-Price, M. A. O’Reilly, S. A. Genet, M. Parkar,
A. K. Sweatman, L. D. Bradley, L. A. Alterman, S. Malcolm, G. Morgan, et al.
1993. Genetic linkage analysis identifies new proximal and distal flanking markers
for the X-linked agammaglobulinemia gene locus, refining its localization in Xq22.
Hum. Mol. Genet. 2: 139–141.
15. Parolini, O., J. F. Hejtmancik, R. C. Allen, J. W. Belmont, G. L. Lassiter,
M. J. Henry, D. F. Barker, and M. E. Conley. 1993. Linkage analysis and physical
mapping near the gene for X-linked agammaglobulinemia at Xq22. Genomics 15:
342–349.
16. Vetrie, D., I. Vorechovský, P. Sideras, J. Holland, A. Davies, F. Flinter,
L. Hammarström, C. Kinnon, R. Levinsky, M. Bobrow, et al. 1993. The gene
involved in X-linked agammaglobulinaemia is a member of the src family of protein-tyrosine kinases. Nature 361: 226–233.
17. Tsukada, S., D. C. Saffran, D. J. Rawlings, O. Parolini, R. C. Allen, I. Klisak,
R. S. Sparkes, H. Kubagawa, T. Mohandas, S. Quan, et al. 1993. Deficient expression of a B cell cytoplasmic tyrosine kinase in human X-linked agammaglobulinemia. Cell 72: 279–290.
18. Rawlings, D. J., D. C. Saffran, S. Tsukada, D. A. Largaespada, J. C. Grimaldi,
L. Cohen, R. N. Mohr, J. F. Bazan, M. Howard, N. G. Copeland, et al. 1993.
Mutation of unique region of Bruton’s tyrosine kinase in immunodeficient XID
mice. Science 261: 358–361.
19. Thomas, J. D., P. Sideras, C. I. Smith, I. Vorechovský, V. Chapman, and
W. E. Paul. 1993. Colocalization of X-linked agammaglobulinemia and X-linked
immunodeficiency genes. Science 261: 355–358.
20. Hendriks, R. W., M. F. de Bruijn, A. Maas, G. M. Dingjan, A. Karis, and
F. Grosveld. 1996. Inactivation of Btk by insertion of lacZ reveals defects in B cell
development only past the pre-B cell stage. EMBO J. 15: 4862–4872.
21. Kerner, J. D., M. W. Appleby, R. N. Mohr, S. Chien, D. J. Rawlings,
C. R. Maliszewski, O. N. Witte, and R. M. Perlmutter. 1995. Impaired expansion
of mouse B cell progenitors lacking Btk. Immunity 3: 301–312.
22. Khan, W. N., F. W. Alt, R. M. Gerstein, B. A. Malynn, I. Larsson, G. Rathbun,
L. Davidson, S. Müller, A. B. Kantor, L. A. Herzenberg, et al. 1995. Defective B cell
development and function in Btk-deficient mice. Immunity 3: 283–299.
23. Scher, I. 1982. The CBA/N mouse strain: an experimental model illustrating the
influence of the X-chromosome on immunity. Adv. Immunol. 33: 1–71.
2935