A girl presented during childhood with a single course of extensive chickenpox and moderate albeit recurrent pneumonia in the presence of idiopathic CD4+ T lymphocytopenia (ICL). Her clinical condition remained stable over the past 10 years without infections, any granulomatous disease, or autoimmunity. Immunophenotyping demonstrated strongly reduced naive T and B cells with intact proliferative capacity. Antibody reactivity on in vivo immunizations was normal. T-cell receptor-Vβ repertoire was polyclonal with a very low content of T-cell receptor excision circles (TRECs). Kappa-deleting recombination excision circles (KRECs) were also abnormal in the B cells. Both reflect extensive in vivo proliferation. Patient-derived CD34+ hematopoietic stem cells could not repopulate RAG2−/−IL2Rγc−/− mice, indicating the lymphoid origin of the defect. We identified 2 novel missense mutations in RAG1 (p.Arg474Cys and p.Leu506Phe) resulting in reduced RAG activity. This report gives the first genetic clue for ICL and extends the clinical spectrum of RAG mutations from severe immune defects to an almost normal condition.

Selective depletion of T lymphocytes is common in both primary and secondary immunodeficiencies. Idiopathic CD4+ T lymphocytopenia (ICL) is defined by an unexplained persistent CD4+ T lymphocyte count of < 300 cells/μL or < 20% of the total T-cell count.1 

Since the discovery of human retroviruses, sporadic ICL patients were recognized with a CD4+ lymphocytopenia not infected by HIV or HTLV-1.2-7  Smith et al reviewed 230179 cases from the CDC AIDS Reporting System and described 47 ICL patients.2,3  Of these cases, only 3 (6%) were asymptomatic. Screening of healthy blood donors confirmed a low prevalence of ICL of 0.2%-0.6%.4,5 

The disease may have a transient nature over the years but mostly persists.3  The immunologic parameters of ICL consist of a prolonged decrease in CD4+ T cell numbers, sometimes with a concomitant decrease in CD8+ T cells and B cells as well. Immunoglobulin levels are normal,2-5  which helps to distinguish ICL from Common Variable Immunodeficiency (CVID).1,6 

Although function declines with age, thymic output is well maintained into late adulthood.7-9  Thymic size correlates with numbers of CD4+CD45RA+ naive T cells. At very young age T lymphocytopenia is often caused by congenital defects resulting in severe combined immunodeficiency syndromes (SCID).10 

Null mutations in RAG1 or RAG2 account for 70% of SCID cases with the classic TBSCID phenotype.10  Hypomorphic mutations with residual RAG activity occur in typical Omenn syndrome,10  or rare cases with lympocytopenia, hypogammaglobulinemia, granulomas in the skin, mucosa, internal organs and viral complications, including EBV-related lymphomas.11,12  In the present paper, we describe hypomorphic RAG1 mutations that corresponds with mild CD4+ T lymphocytopenia, normal in vitro lymphocyte function and in vivo vaccination responses.

Subjects and blood samples

Heparinized venous blood was collected from healthy (age-matched) donors, patient and family members. The study was approved by the institutional medical ethical committee and informed consent for the research purpose described was obtained from the parents of the child and age-matched controls in accordance with standards of the 1964 declaration of Helsinki.

Lymphocyte phenotyping

Absolute numbers of T cells, B cells, and NK cells were determined with Multitest 6-color (FACSCantoII; BD-Biosciences). For T- and B-cell subset analysis directly conjugated monoclonal antibodies (MoAbs) were obtained from BD except for CD45RA-RD1 (Beckman-Coulter) and CD27-FITC (Sanquin).13,14 

Lymphocyte activation, determination of T-cell receptor CDR3 spectratype, and TRECs

Lymphocyte proliferation was performed as described.13  CD45RA+ naive T cells were purified (> 95%) to determine CDR3 patterns in CD4+ and CD8+ T cells or to isolate DNA for Sj–T-cell receptor excision circles (TRECs) analysis.14,15 

KREC assay and Vκ3-20–specific IgκREHMA

Four B-cell subsets were purified from PBMCs using FACS-DiVa cell-sorter.16,17  Somatic hypermutation in these subsets was assessed by the Vκ3-20–specific Igκ-restriction enzyme hot-spot mutation assay (REHMA).16 

RAG gene analysis and in vitro V(D)J recombination assay

The RAG1 and RAG2 genes were amplified by PCR and sequenced.18  The level of recombination activity of the RAG1 mutant proteins were compared with the wt RAG1.

Case

The patient was the first child of healthy, nonconsanguineous Dutch parents, born in May 1992. At the age of 5, she contracted chickenpox with numerous large hemorrhaghic skin lesions and VZV-associated pneumonia. Skin lesions healed slowly with cicatriciation. Until the age of approximately 8 years she has suffered from recurrent episodes of fever and (viral) pneumonia for which she regularly received antibiotics. High-resolution CT scanning of neck, thorax, and abdomen around that age showed mild bronchiectasis without enlargement of perihilar lymph nodes or granulomatous lesions in lungs, liver, or spleen; a thymus was not detectable (supplemental Figure 1, available on the Blood Web site; see the Supplemental Materials link at the top of the online article). Dysmorphic features were not observed by clinical geneticists. Chromosomal abnormalities (including 22q11 hemizygosity) were excluded.

She remained disease-free for the past 10 years using oral cotrimoxazole as prophylaxis.

Lymphocyte subpopulations and humoral immune responses

Immunophenotyping of the patient's PBMCs showed low numbers of CD4+ T cells and to a lesser extent, CD8+ T cells. The CD4+ lymphocytopenia was persistent over time with some fluctuations (Figure 1A). The percentage of CD4+ and CD8+ T cells with a CD45RO+ memory phenotype was strongly increased as compared with the naive T cells (Figure 1B).

Figure 1

Absolute numbers of CD4+ and CD8+ T cells, CD19+ B cells and CD3CD16/56+ NK cells over time. (A) Numbers of the patient's lymphocytes are indicated in closed circles; the age-matched control levels for different age categories (ie, 4-10 years; 10-18 years) are indicated in a range of ± 2SD (dotted lines). (B) Immunophenotyping of CD4+ and CD8+ T cells and CD19+CD20+ B cells, according to CD45RA/CD27 and sIgD/CD27, respectively, as compared with a healthy, age-matched control. Proliferative capacity of the patient's T cells. T-cell proliferation of the patient and a healthy control is shown by CFSE dilution after 6 days of culture. Polyclonal proliferation was induced by a combination of CD3/CD28 or IL-15, whereas antigen-specific proliferation was assessed by stimulation with tetanus toxoid (Tet Tox), cytomegalovirus (CMV) or varicella zoster virus (VZV). (C) As expected in the presence of negative serology, CMV antigen did not activate her T cells. Increased proliferation of the peripheral CD4+ T-cell compartment as demonstrated by nuclear Ki67 staining in naive (CD45RA+CD27+) CD4+ and CD8+ T cells, compared with healthy age-matched naive control T cells, as described.13  TRECs in patient's T cells for the early, so-called signal joint (Sj) over a period of 5 years. (D) For T-cell repertoire analysis CDR3 spectratyping in the patient's T cells was analyzed, being representative for 2 separate experiments in triplicate, more than 2 years apart.

Figure 1

Absolute numbers of CD4+ and CD8+ T cells, CD19+ B cells and CD3CD16/56+ NK cells over time. (A) Numbers of the patient's lymphocytes are indicated in closed circles; the age-matched control levels for different age categories (ie, 4-10 years; 10-18 years) are indicated in a range of ± 2SD (dotted lines). (B) Immunophenotyping of CD4+ and CD8+ T cells and CD19+CD20+ B cells, according to CD45RA/CD27 and sIgD/CD27, respectively, as compared with a healthy, age-matched control. Proliferative capacity of the patient's T cells. T-cell proliferation of the patient and a healthy control is shown by CFSE dilution after 6 days of culture. Polyclonal proliferation was induced by a combination of CD3/CD28 or IL-15, whereas antigen-specific proliferation was assessed by stimulation with tetanus toxoid (Tet Tox), cytomegalovirus (CMV) or varicella zoster virus (VZV). (C) As expected in the presence of negative serology, CMV antigen did not activate her T cells. Increased proliferation of the peripheral CD4+ T-cell compartment as demonstrated by nuclear Ki67 staining in naive (CD45RA+CD27+) CD4+ and CD8+ T cells, compared with healthy age-matched naive control T cells, as described.13  TRECs in patient's T cells for the early, so-called signal joint (Sj) over a period of 5 years. (D) For T-cell repertoire analysis CDR3 spectratyping in the patient's T cells was analyzed, being representative for 2 separate experiments in triplicate, more than 2 years apart.

Close modal

Although B-cell numbers were normal, the distribution of the B-cell subsets was severely disturbed (Figure 1B). The frequency of class-switched CD27+ memory B cells was low (3.4%-9.2%) whereas the frequency of nonswitched sIgD+CD27+ memory B cells showed a high and stable percentage between 58%-72% (controls between 5-15 years: 18.4 ± 7.2%, n = 40). Humoral immunity was intact (supplemental Table 1).

In vitro functionality of T cells, thymic function, and TCR repertoire

T-cell proliferation to general stimuli (ie, CD3/CD28, cytokines), and specific antigens was intact (Figure 1C; data not shown). Cytokine release, CTL or spontaneous NK-cell killing of target cells were normal (data not shown).

In the absence of a detectable thymus, peripheral in vivo T-cell proliferation was expected to be increased to sustain normal T-cell numbers. This was indeed demonstrated in 2 ways. First, lymphocyte expression of the nuclear proliferation marker Ki67 was not different from age-matched control samples (Figure 1D; data not shown). Second, the content of TRECs was reduced compared with controls (Figure 1D), indicative for increased T-cell proliferation. However, despite extensive peripheral T-cell expansion to sustain the number of T cells, no restriction in the TCR repertoire was demonstrated (Figure 1D).

Cellular and molecular properties of the circulating B-cell compartment

B-cell proliferation was found to be normal on BcR-dependent and BcR-independent stimulation (supplemental Figure 2A). The number of cell divisions of the transitional, naive-mature, and nonswitched B cells was strongly increased as compared with healthy controls (supplemental Figure 2B).16,17  Naive B cells were not somatically mutated and the mutation frequency in the expanded fraction of nonswitched natural effector B cells was only 4.7% (healthy controls 15%; n = 5; supplemental Figure 2C). These results indicate that the number of total peripheral B cells is normal because of extensive (antigen-independent) proliferation of the transitional, naive and nonswitched B cells.

Lymphoid origin of the defect and genetic immunodeficiency screening

T-cell maturation data suggested an intrinsic T-cell developmental defect with thymus hypoplasia from early age onward. Using a humanized RAG2−/−IL2Rγc−/− mouse model,19  normal human T-cell reconstitution on intrahepatic injection of 4 × 105 patient-derived CD34+CD38 HSCs from the bone marrow of the patient failed, in contrast to control HSCs. In the blood, no patient T cells could be detected at 6 and 9 weeks, nor in the thymus at 10 weeks (supplemental Table 2), confirming the lymphoid background of the thymus hypoplasia.

RAG defect

Sequence analysis demonstrated 2 novel heterozygous missense mutations in the RAG1 gene (c.1420C > T and c.1516C > T), affecting the evolutionary conserved amino acids Arg474 and Leu506 (Figure 2A). The RAG1 p.Arg474Cys recombinant protein had 25% recombinase activity compared with wild-type RAG1, whereas the p.Leu506Phe mutant did not have any residual activity (Figures 2B-C).

Figure 2

RAG1 gene mutations and residual recombination activity. Sequence alignment of RAG1 protein of different species. Both the arginine (R) at amino acid position 474 and the leucine (L) at 506 are conserved residues. (A) The parents were heterozygous for each of the mutation. (B) Transfection of pDVG93, RAG1 and RAG2 in 3T3 fibroblasts results in an inversion rearrangement of pDVG93, which can be detected by the primers DG89 and DG147. (C) Only the R474C mutation results in residual recombination activity, as assessed by the in vitro recombination assay using pDVG93.

Figure 2

RAG1 gene mutations and residual recombination activity. Sequence alignment of RAG1 protein of different species. Both the arginine (R) at amino acid position 474 and the leucine (L) at 506 are conserved residues. (A) The parents were heterozygous for each of the mutation. (B) Transfection of pDVG93, RAG1 and RAG2 in 3T3 fibroblasts results in an inversion rearrangement of pDVG93, which can be detected by the primers DG89 and DG147. (C) Only the R474C mutation results in residual recombination activity, as assessed by the in vitro recombination assay using pDVG93.

Close modal

Hypomorphic mutations may result in typical Omenn patients with T cells showing an oligoclonal TCR repertoire, and poor development of precursor B cells,18,20  which may contribute to hyperinflammation/autoimmunity because of low-affinity autoantibody production.21,22  These features were absent in our patient.

Epicrise

V(D)J recombination activity of the RAG1 mutant proteins was very low. Even this residual RAG1 activity provides a low level of T- and B-cell production for effective host immunity. The functional immune system in terms of the mild clinical course in the absence of a detectable thymus could be explained by cytokine-driven proliferation, as reflected by the maintenance of a polyclonal repertoire.23  The persistent lymphocyte activation could be compatible with cytokine-driven homeostasis (Figure 2A; supplemental Table 3).

The thymus hypoplasia associated with an immune defect similar to ICL and few clinical symptoms was identified to be caused by novel heterozygous RAG1 mutations with very low RAG activity. Instead of SCID, Omenn syndrome or extensive granulomatous disease, our case study adds to the complexity of hypomorphic mutations in the RAG genes.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank Paul Baars for technical support and Prof Jacques van Dongen for the help in genetic analyses. We are grateful to Profs Hergen Spits and Frank Miedema for critically reading the manuscript and the useful comments and research suggestions over the years.

Contribution: T.W.K. performed the diagnosis and treatment, designed research, and wrote the paper; H.I. performed research, cloning and expression of RAG variants, and analysis of data; E.M.M.v.L. contributed to the design of research and data analysis; M.H.J. and M.D.H. performed research and data analysis; K.C.W. performed vital mouse studies; R.A.W.v.L. contributed to the design of research and wrote the manuscript; and M.v.d.B. contributed to the design of study and wrote the manuscript.

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

Correspondence: T. W. Kuijpers, AMC (location G8-205), Meibergdreef 9, 1105 AZ Amsterdam; The Netherlands; e-mail: t.w.kuijpers@amc.nl.

1
Primary immunodeficiency diseases: Report of an IUIS scientific committee
International Union of Immunological Societies.
Clin Exp Immunol
1999
, vol. 
118
 (pg. 
S1
-
S28
)
2
Smith
 
D
Neal
 
JJ
Holmberg
 
SD
the Centers for Disease Control Idiopathic CD4+ T-lymphocytopenia Task Force
Unexplained opportunistic infections and CD4+ T-lymphocytopenia without HIV infection.
N Engl J Med
1993
, vol. 
328
 
6
(pg. 
373
-
379
)
3
Zonios
 
DI
Falloon
 
J
Bennett
 
JE
, et al. 
Idiopathic CD4+ lymphocytopenia: natural history and prognostic factors.
Blood
2008
, vol. 
112
 
2
(pg. 
287
-
294
)
4
Aledort
 
LM
Operskalski
 
EA
Dietrich
 
SL
, et al. 
Low CD4+ counts in a study of transfusion safety.
N Engl J Med
1993
, vol. 
328
 
6
(pg. 
441
-
442
)
5
Girotto
 
M
Verani
 
DA
Pagliaro
 
PP
Idiopathic CD4+ T-lymphocytopenia in blood donors: cohort study.
Transfusion
1994
, vol. 
34
 
10
(pg. 
935
-
936
)
6
Spickett
 
GP
Farrant
 
J
North
 
ME
Zhang
 
JG
Morgan
 
L
Webster
 
AD
Common variable immunodeficiency: how many diseases?
Immunol Today
1997
, vol. 
18
 
7
(pg. 
325
-
328
)
7
Mackall
 
CL
Fleisher
 
TA
Brown
 
MR
, et al. 
Age, thymopoiesis, and CD4+ T-lymphocyte regeneration after intensive chemotherapy.
N Engl J Med
1995
, vol. 
332
 
3
(pg. 
143
-
149
)
8
Douek
 
CD
McFarland
 
DR
Keiser
 
PH
, et al. 
Changes in thymic function with age and during the treatment of HIV infection.
Nature
1998
, vol. 
396
 
6712
(pg. 
690
-
695
)
9
McCune
 
JM
Loftus
 
R
Schmidt
 
DK
, et al. 
High prevalence of thymic tissue in adults with human immunodeficiency virus-1 infection.
J Clin Invest
1998
, vol. 
101
 
11
(pg. 
2301
-
2308
)
10
Fischer
 
A
Cavazzana-Calvo
 
M
De Saint Basile
 
G
, et al. 
Naturally occurring primary deficiencies of the immune system.
Annu Rev Immunol
1997
, vol. 
15
 (pg. 
93
-
124
)
11
Schuetz
 
C
Huck
 
K
Gudowius
 
S
, et al. 
An immunodeficiency disease with RAG mutations and granulomas.
N Engl J Med
2008
, vol. 
358
 
19
(pg. 
2030
-
2038
)
12
De Ravin
 
SS
Cowen
 
EW
Zarember
 
KA
, et al. 
Hypomorphic Rag mutations can cause destructive midline granulomatous disease.
Blood
2010
, vol. 
116
 
8
(pg. 
1263
-
1271
)
13
Kuijpers
 
TW
Bende
 
RJ
Baars
 
PA
, et al. 
CD20 deficiency in humans results in impaired T cell-independent antibody responses.
J Clin Invest
2010
, vol. 
120
 
1
(pg. 
214
-
222
)
14
Hazenberg
 
MD
Stuart
 
JW
Otto
 
SA
, et al. 
T cell division in human immunodeficiency virus (HIV)-1 infection is mainly due to immune activation: a longitudinal analysis in patients before and during highly active anti-retroviral therapy.
Blood
2000
, vol. 
95
 
1
(pg. 
249
-
255
)
15
van Dongen
 
JJ
Langerak
 
AW
Brüggemann
 
M
, et al. 
Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: report of the BIOMED-2 Concerted Action BMH4-CT98–3936.
Leukemia
2003
, vol. 
17
 
12
(pg. 
2257
-
2317
)
16
van Zelm
 
MC
Szczepanski
 
T
van der Burg
 
M
van Dongen
 
JJ
Replication history of B lymphocytes reveals homeostatic proliferation and extensive antigen-induced B cell expansion.
J Exp Med
2007
, vol. 
204
 
3
(pg. 
645
-
655
)
17
van der Burg
 
M
Weemaes
 
CM
Preijers
 
F
, et al. 
B-cell recovery after stem cell transplantation of Artemis-deficient SCID requires elimination of autologous bone marrow precursor-B-cells.
Haematologica
2006
, vol. 
91
 
12
(pg. 
1705
-
1709
)
18
Noordzij
 
JG
Verkaik
 
NS
Hartwig
 
NG
de Groot
 
R
van Gent
 
DC
van Dongen
 
JJ
N-terminal truncated human RAG1 proteins can direct T-cell receptor but not immunoglobulin gene rearrangements.
Blood
2000
, vol. 
96
 
1
(pg. 
203
-
209
)
19
Gimeno
 
R
Weijer
 
K
Voordouw
 
A
, et al. 
Monitoringthe effect of gene silencing by RNA interference in human CD34+ cells injected into newborn RAG2-/- gammac-/- mice: functional inactivation of p53 in developing T cells.
Blood
2004
, vol. 
104
 
13
(pg. 
3886
-
3893
)
20
De
 
P
Rodgers
 
KK
Putting the pieces together: identification and characterization of structural domains in the V(D)J recombination protein RAG1.
Immunol Rev
2004
, vol. 
200
 (pg. 
70
-
82
)
21
Walter
 
JE
Rucci
 
F
Patrizi
 
L
, et al. 
Expansion of immunoglobulin-secreting cells and defects in B cell tolerance in Rag-dependent immunodeficiency.
J Exp Med
2010
, vol. 
207
 
7
(pg. 
1541
-
1554
)
22
Cassani
 
B
Poliani
 
PL
Marrella
 
V
, et al. 
Homeostatic expansion of autoreactive immunoglobulin-secreting cells in the Rag2 mouse model of Omenn syndrome.
J Exp Med
2010
, vol. 
207
 
7
(pg. 
1525
-
1540
)
23
Surh
 
CD
Sprent
 
J
Homeostasis of naive and memory T cells.
Immunity
2008
, vol. 
29
 
6
(pg. 
848
-
862
)
Sign in via your Institution