Defects of the common gamma chain subunit of the cytokine receptors (γc) or of Jak3, a tyrosine kinase required for γc signal transduction, result in TB+ severe combined immunodeficiency (SCID). However, atypical cases, characterized by progressive development of T lymphocytes, have been also reported. We describe a child with SCID caused by Jak3 gene defects, which strongly but not completely affect Jak3 protein expression and function, who developed a substantial number (>3,000/μL) of autologous CD3+CD4+ T cells. These cells showed a primed/activated phenotype (CD45R0+ Fas+HLA-DR+ CD62Llo), defective secretion of T-helper 1 and T-helper 2 cytokines, reduced proliferation to mitogens, and a high in vitro susceptibility to spontaneous (caused by downregulation of bcl-2 expression) as well as activation-induced cell death. A restricted T-cell receptor repertoire was observed, with oligoclonal expansion within each of the dominant segments. These features resemble those observed in γc-/y and in Jak3−/−mice, in which a population of activated, anergic T cells (predominantly CD4+) also develops with age. These results suggest that residual Jak3 expression and function or other Jak3-independent signals may also permit the generation of CD4+ T cells that undergo in vivo clonal expansion in humans; however, these mechanisms do not allow development of CD8+ T cells, nor do they fully restore the functional properties of CD4+ T lymphocytes.

JAK3, A MEMBER of the Janus family tyrosine kinase,1,2 is an intracellular kinase required for signaling through the common gamma chain (γc),3-5 a subunit of the cytokine receptors for interleukin-2 (IL-2), IL-4, IL-7, IL-9, and IL-15.6 

Functional integrity of the γc/Jak3 system is essential for lymphoid development,7-9 signaling,10 and survival.11 In humans, genetic defects affecting the IL-2RG gene (encoding for γc) or the Jak3 gene result in X-linked12-14 or in autosomal recessive15-19severe combined immunodeficiency (XL-SCID or AR-SCID), respectively, with lack of circulating T cells and a normal or increased number of B lymphocytes that are, however, functionally defective.20 

Disruption of the IL-2RG or the Jak3 genes in mice also results in SCID, but with a distinct biological phenotype, characterized by a severe defect of T-, B-, and natural killer (NK)-cell differentiation, that allows an age-dependent increase of activated and anergic CD4+ T cells in the periphery.13,14,17-19 Jak3 protein expression in the thymus is essential to reconstitute thymocyte development (including γδ and NK cells); furthermore, maintenance of peripheral expression of Jak3 is required to correct the immunological abnormalities (T-cell anergy) observed in Jak3−/− mice.21 

In humans, atypical cases of XL-SCID, characterized by development of T lymphocytes, have been recently described.4,22-24 With the exception of the case reported by Stephan et al, in which development of autologous T lymphocytes resulted from a spontaneous reversion of the genetic defect at the IL-2RG gene locus,24 T cells remained genetically and functionally defective in other patients with atypical XL-SCID.4,22,23 

We report the case of an infant with TB+ SCID caused by Jak3 mutations, which strongly but not completely affect Jak3 protein expression and function, who developed early in life a substantial number of autologous, activated, and oligoclonal CD3+CD4+ T cells that remained genetically and functionally defective.

Case report.

The infant is the only son of nonconsanguineous parents. The diagnosis of SCID was serendipitously obtained at birth, when profound abnormalities of lymphocyte subpopulations (Table 1) were disclosed during blood testing for an episode of intestinal subobstruction that resolved spontaneously at 1 week of age. The infant was admitted to the Bone Marrow Transplantation Unit of the Department of Pediatrics, University of Brescia, at 1 month of age. At that time, chest radiograph examination revealed mild interstitial pneumonia. He was hospitalized in a laminar flow unit, and he was treated with intravenous immunoglobulins, antibiotics, and acyclovir until the age of 3 months, when he received haploidentical bone marrow transplantation (BMT) with T-depleted marrow from the father following conditioning with busulphan and cyclophosphamide. Before BMT, an increase of T-cell count was detected in the periphery (Table 1).

Table 1.

Immunophenotypical Analysis

At Birth At 2.5 Months At 3 MonthsHealthy Controls (1 to 4 mo)
PBL/μL  3,026 4,253  8,890  4,100 ± 1,500  
CD3 (%)  2  24 41  71 ± 7  
CD4 (%)  2  24  41  40 ± 7 
CD8 (%)  1  1  1  25 ± 5  
CD19 (%)  79 48  44  18 ± 6  
CD16 (%)  13  5  11 ± 5 
At Birth At 2.5 Months At 3 MonthsHealthy Controls (1 to 4 mo)
PBL/μL  3,026 4,253  8,890  4,100 ± 1,500  
CD3 (%)  2  24 41  71 ± 7  
CD4 (%)  2  24  41  40 ± 7 
CD8 (%)  1  1  1  25 ± 5  
CD19 (%)  79 48  44  18 ± 6  
CD16 (%)  13  5  11 ± 5 

The molecular defect in the Jak3 gene and the biochemical analysis of the Jak/STAT signaling pathway have been reported in detail elsewhere, together with other Jak3-deficient SCID patients (patient LE25). Briefly, the patient carries distinct mutations on the two Jak3 alleles: an intragenic deletion that results in several mutant splicing products, including an in-frame deleted cDNA form lacking exons 10 to 12, and a de novo A to G transition at the nucleotide 1537, leading to a glutamic acid to glycine substitution at codon 481 (E481G). Both mutant forms of the Jak3 protein were expressed in lymphoblastoid cell lines, albeit in trace amounts, as detected by Western blotting. Furthermore, residual preservation of the function of the E481G Jak3 mutant was shown, based on reduced but detectable phosphorylation of Jak3 and STAT5 proteins in response to IL-2.25 Molecular analysis of the patient's peripheral blood T cells revealed that they were autologous (as detected by highly polymorphic DNA markers D1S80 and DQ alpha) and genetically defective, as shown by mutation analysis at the Jak3 locus (data not shown).

After BMT, the patient developed hemolytic anemia, associated with the presence of autologous and donor-derived T cells in the periphery.

Lymphocyte surface phenotype analysis.

Evaluation of lymphocyte surface membrane expression was performed on heparinized whole blood samples with the following monoclonal antibodies (MoAbs): fluorescein-isothiocyanate (FITC)-conjugated OKT3/CD3, FITC-OKT8/CD8, FITC- or phycoerytrin (PE)-conjugated OKT4/CD4, PE-OKDR/HLA DR (Ortho, Raritan, NJ), PE-Leu11c/CD16, PE-Leu45RO/CD45RO (Becton Dickinson, Mountain View, CA), FITC-B4/CD19, PE-2H4/CD45RA (Coulter Immunology, Hialeah, FL), PE-DREG-56/CD62L, and FITC-DX2/CD95 (Pharmingen, San Diego, CA).

After two washes in phosphate-buffered saline (PBS)-1% fetal calf serum (FCS), samples were analyzed with a flow cytometer equipped with an argon-ion laser (488 nm; Cytoron Absolute, Ortho), using the ABS software. Gates were selected on lymphoid cells as determined by forward and right-angle scatter properties; dead cells were excluded by setting an appropriate threshold trigger on the low forward light scatter parameter; and nonspecific staining was assessed using FITC-conjugated nonimmune mouse IgG (Coulter). Data were acquired and stored in list mode. Fluorescence analysis was performed on a log scale.

Preparation of lymphocytes.

Mononuclear cells (PBMC) were separated from the peripheral blood by Ficoll-Hypaque (Pharmacia Biotech, Piscataway, NJ) density gradient centrifugation and washed three times in RPMI-1640 (GIBCO Laboratories, Grand Island, NY).

Lymphocyte activation.

All experiments were performed in RPMI-1640 medium supplemented with 10% heat-inactivated FCS, 1% glutamin, and antibiotics. Proliferative responses were measured in triplicates of 200 μL containing 1 × 105 total PBMC using various combinations of the following stimulating agents: immobilized anti-CD3 (OKT3) MoAb, 200 ng/mL, with or without IL-2, 20 U/mL (Biosource International, Camarillo, CA); phytoemoagglutinin (PHA), 1.2 μg/mL (Irvine Scientific, Santa Ana, CA); phorbol 12-myristate 13-acetate (PMA), 5 ng/mL (Sigma, St Louis, MO) plus ionomycin, 500 ng/mL (Sigma); and 1 × 105irradiated (2,000 rads) allogeneic PBMC from a pool of healthy donors. After 72 hours of culture (or 120 hours for the mixed lymphocyte culture) at 37°C, 5% CO2, the cells were pulsed overnight with 1 μCi (3H)-thymidine and were obtained subsequently. Incorporated radioactivity was measured by liquid scintillation counting (Beckman, Fullerton, CA), and the results were expressed as mean counts per minute of triplicates.

Apoptosis assay.

PBMC, cultured with or without the stimulating agents in the conditions indicated above, were also evaluated for apoptosis.

After 72 hours of culture, cells were washed and the percentage of hypodiploid nuclei (less than diploid DNA content) was determined with a slight modification of the method of Nicoletti et al,26as described.27 Briefly, 1 × 106 PBMC were fixed in 1 mL of cold 70% ethanol at 4°C for 20 minutes. Cells were then washed, incubated at room temperature for 1 minute with RNase (0.5 mg/mL; Boehringer GmbH, Mannheim, Germany), for 15 minutes with propidium iodide (100 mg/mL; Sigma), and immediately analyzed. The correct threshold was selected experimentally using the model system of apoptosis induced in murine thymocytes by 72 hours culture with dexamethasone 10−7 mol/L. Apoptotic cell nuclei, easily distinguishable from debris owing to the condensation of nuclear chromatin, emitted red fluorescence in the RD-FL channels 46 to 146. As there was no overlap between apoptotic nuclei and debris, the small percentage of residual low-fluorescence detritus (<1% in normal cells) was eliminated by gating the red fluorescence scale at channel 45. All measurements were performed with the same instrument setting.

Analysis of intracellular bcl-2 expression.

A total of 5 × 105 PBMC were permeabilized using Permeafix (Ortho) for 40 minutes, before an incubation with a rabbit polyclonal antibody to bcl-2 (Santa Cruz Biotechnology Inc, Santa Cruz, CA), or with polyclonal nonimmune rabbit IgG. After 30 minutes, cells were washed and incubated with FITC-conjugated goat anti-rabbit IgG antibody (Santa Cruz Biotechnology Inc). Quantitation of positivity for bcl-2 was performed using QuickCal beads (Flow Cytometry Standards Corp, San Juan, PR), and results were expressed as specific molecule equivalents of soluble fluorochrome (MESF), after subtracting the values obtained for nonspecific fluorescence (always <2,000 MESF).

Cytokine production and measurement.

For cytokine production, 1 × 106 PBMC were cultured at 37°C, 5% CO2 in 24-well flat-bottomed plates in a final volume of 1 mL with PMA (5 ng/mL) plus ionomycin (500 ng/mL). After 72 hours, supernatants were collected and frozen at −20°C until use. Sandwich enzyme-linked immunosorbent assay (ELISA) commercial kits were used to measure IL-2 (Genzyme, Cambridge, MA), IL-4 (BioSource International), IL-5 (CytImmune Sciences, College Park, MD), and interferon (IFN)-γ (BioSource International).

Analysis of the T-cell receptor repertoire diversity.

One microgram of the total RNA, prepared with the guanidinium thiocyanate-phenol-chloroform method from PBMC, was used to synthesize the first strand of TCRB chain cDNA using a specific TCRBC primer (βcDNA: 5′ GGG CTG CTC CTT GAG GGG CTG CGG 3′) and the RiboClone cDNA Synthesis System (Promega Corporation, Madison, WI). cDNA was subjected to enzymatic amplification using a second human TCRBC primer (βAI: 5′ CCC ACT GTG CAC CTC CTT CC 3′) and a TCRBV degenerated primer (Vβd: 5′ ACG TGA ATT CT(GT) T(ACT)(CT) TGG TA(CT) (AC)(AG)(AT) CA 3′). Forty cycles of polymerase chain reaction (PCR) were performed under the following conditions: denaturation at 94°C for 1 minute, annealing at 52°C for 1 minute, and extension at 72°C for 1 minute; the last cycle extension was performed at 72°C for 7 minutes. The specificity of the total amplified products was analyzed using a colorimetric method and biotinylated TCRBV-specific probes.28 Subsequently, the TCRBV chains of interest were amplified by 35 cycles of PCR, using TCRBV-specific family primers (TCRBV2: 5′ TCA TCA ACC ATG CAA GCC TGA CCT 3′; TCRBV4: 5′ GCC CAA ACC TAA CAT TCT CAA CTC 3′; TCRBV5S1: 5′ ATA CTT CAG TGA GAC ACA GAG AAA 3′; TCRBV6: 5′ AGG CCT GAG GGA TCC GTC TC 3′; and TCRBV14: 5′ GTC TCT CGA AAA GAG AAG AGG AAT 3′) and the TCRBC oligonucleotide βAI. For heteroduplex analysis, 20 μL of TCRBV-specific products were heated to 95°C for 5 minutes and cooled to 50°C for 1 hour. The annealed samples were kept on ice and then run for 5 to 6 hours at 200 V, at room temperature, on a 12% nondenaturing polyacrylamide gel (PAGE; 29:1 acrylamide/bisacrylamide) performed in 1× TBE buffer (0.089 mol/L Tris-borate and 0.002 mol/L EDTA, pH 8.0). DNA Molecular Weight V (Boehringer Mannheim) was used as size marker. The gel was stained for 30 to 60 minutes, at room temperature, in the dark, in a solution containing 0.75 μg/mL ethidium bromide in 200 mL of 1× TBE and photographed under ultraviolet light. TCRBV8-specific amplified products from the T-cell line J77 and from PBMC stimulated with an anti-TCRBV8 MoAb were used as monoclonal and polyclonal controls, respectively.29 TCRBV2 and TCRBV5S1 products, obtained from a new amplification performed with specific primers, were purified, cloned, and sequenced as recently described.30 Sequences were compared with published data relative to TCRBV, TCRBD, TCRBJ, and TCRBC segments.31,32 

Immunophenotypical and molecular studies.

Cytofluorimetric analysis performed on PBMC collected from the patient at the time of birth revealed a typical TB+ SCID phenotype, with a severely reduced proportion of peripheral blood T lymphocytes and an increased proportion of B cells (Table 1).

Interestingly, at 3 months of age the patient developed autologous (as assessed by HLA-typing) CD3+CD4+ cells that increased to 41% just before undergoing haploidentical BMT from his father (Table 1). However, these lymphocytes were phenotypically abnormal, as they predominantly coexpressed the activation marker HLA-DR, were almost exclusively CD45R0+ and CD95/Fas+, and showed a reduced density of CD62L (Fig 1). These features are typical of primed/activated T cells.

Fig. 1.

Two-color analysis of activation/memory markers expression by peripheral CD4+ lymphocytes from the patient at 3 months of age and from one representative age-matched healthy control. Data are presented as relative log fluorescence intensity.

Fig. 1.

Two-color analysis of activation/memory markers expression by peripheral CD4+ lymphocytes from the patient at 3 months of age and from one representative age-matched healthy control. Data are presented as relative log fluorescence intensity.

Close modal

Normal expression of the γc was found on the lymphocyte surface, as detected with the rat TUGh4 MoAb (a kind gift of Dr K. Sugamura, Department of Microbiology, Tohoku University School of Medicine, Japan; data not shown).

TCRBV repertoire diversity.

The analysis of the TCRBV usage was performed on patient's PBMC at 3 months of age and compared with that of age-matched healthy controls. Figure 2A shows that all TCRBV transcripts, with the only exception of TCRBV13S1 chain, can be amplified from the patient's lymphocytes without evidence for over-representation of single TCRBV population. On the contrary, the most notable difference is the greatly reduced percentage of TCRBV3, TCRBV13S1, and TCRBV13S2 chains in the patient as compared with controls. However, because we have previously found that the level of TCRBV diversity is not necessarily related to the relative percentage of TCRBV expression,33 we also performed a heteroduplex analysis of TCRBV2, TCRBV4, TCRBV5S1, TCRBV6, and TCRBV14 transcripts, which were expressed at highest levels in the patient's T cells. This technique is based on the different ability of amplified TCRBV segments to migrate in a polyacrylamide gel, depending on their monoclonal or polyclonal composition.29 As shown in Fig 2B, all the PCR products obtained with the 5 TCRBV family-specific primers generated homoduplex bands that are indicative of predominant homogeneous molecular species in the context of a background of heteroduplex bands or of smears, respectively, representing minor oligoclonal or polyclonal expansions. On the contrary, the amplified products of the same TCRBV segments prepared from PBMC of an age-matched healthy control and chosen among those analyzed in Fig 2A generated smears or faint heteroduplex bands. This result is consistent with our previous observations suggesting that, in the absence of antigenic stimulation, most of the TCRBV segments expressed by PBMC from normal individuals are largely polyclonal.34 

Fig. 2.
(A) TCRBV usage in patient with AR-SCID (▪) and in healthy controls (▧). The relative percentage of TCRBV segments expression was calculated by normalizing the OD value of each individual TCRBV segment with respect to the sum of the OD values of all the 26 TCRBV chains as follows: % of expression:
ODiI=126ODi×100
(B) Heteroduplex analysis of the indicated TCRBV chains prepared from patient's and control's lymphocytes after PCR amplification performed with TCRBV-specific primers. TCRBV8 amplified products, obtained from the amplification of the J77 and C1-632 cells RNA29 were used as monoclonal and polyclonal controls and loaded, respectively, in the first and second lane of the gel. MW; molecular weight marker. (C) Junctional amino acid TCRBV2 and TCRBV5S1 sequences were deduced from nucleotide sequences and displayed as standard one-letter code. Only the last 3′ amino acids of the TCRBV segments and the first 5′ amino acids of TCRBJ chains are shown.
Fig. 2.
(A) TCRBV usage in patient with AR-SCID (▪) and in healthy controls (▧). The relative percentage of TCRBV segments expression was calculated by normalizing the OD value of each individual TCRBV segment with respect to the sum of the OD values of all the 26 TCRBV chains as follows: % of expression:
ODiI=126ODi×100
(B) Heteroduplex analysis of the indicated TCRBV chains prepared from patient's and control's lymphocytes after PCR amplification performed with TCRBV-specific primers. TCRBV8 amplified products, obtained from the amplification of the J77 and C1-632 cells RNA29 were used as monoclonal and polyclonal controls and loaded, respectively, in the first and second lane of the gel. MW; molecular weight marker. (C) Junctional amino acid TCRBV2 and TCRBV5S1 sequences were deduced from nucleotide sequences and displayed as standard one-letter code. Only the last 3′ amino acids of the TCRBV segments and the first 5′ amino acids of TCRBJ chains are shown.
Close modal

The strongest evidence for oligoclonal expansion that confirmed the results obtained with the heteroduplex analysis was provided by sequencing of TCRBV2 and TCRBV5S1 transcripts. In fact, in both groups of sequences the dominance of a single clone was detected (Fig 2C). We did not observe, however, amino acid similarities in the NDN regions of the two major groups of clones.

Functional studies.

In accordance with the lack of T cells at birth, in vitro response of PBMC to T-cell activating agents (PHA, anti-CD3, and anti-CD3 plus IL-2) was abolished, whereas stimulation with PMA plus ionomycin, which can also elicit B-cell activation, resulted in a substantial proliferative response (Table 2).

Table 2.

Proliferative Response (cpm)

Patient at Birth Healthy Control (newborn)Patient at 3 Months Healthy Control (3 mo)
Medium  2,700  670  3,150  950  
CD3  6,700 19,150  4,200  33,150  
PHA  5,500  75,100  6,900 85,000  
PMA plus Ionomycin  46,950  53,100  33,700 47,500  
CD3 plus IL-2  8,550  44,900  36,200  45,550 
IL-2  5,950  3,100  17,600  2,450  
MLC (vpooled unrelated donors)  ND  ND  18,250 72,250 
Patient at Birth Healthy Control (newborn)Patient at 3 Months Healthy Control (3 mo)
Medium  2,700  670  3,150  950  
CD3  6,700 19,150  4,200  33,150  
PHA  5,500  75,100  6,900 85,000  
PMA plus Ionomycin  46,950  53,100  33,700 47,500  
CD3 plus IL-2  8,550  44,900  36,200  45,550 
IL-2  5,950  3,100  17,600  2,450  
MLC (vpooled unrelated donors)  ND  ND  18,250 72,250 

Abbreviations: MLC, mixed lymphocyte culture; ND, not done.

A defect of the proliferative response to PHA, anti-CD3, and pooled unrelated cells was also observed after appearance of circulating autologous T lymphocytes. This defect was too profound to be explained solely by the reduction of the T-cell number as compared with the controls' (Table 2). A moderate increase of proliferation was obtained after the addition of exogenous IL-2 to both unstimulated and stimulated cultures, in keeping with the strongly reduced, but detectable, levels of Jak3 and STAT5 phosphorylation observed after the stimulation with IL-2.25 These observations suggested a defective IL-2 production following in vitro culture with the activating stimuli mentioned above.

In fact, even under powerful stimulating conditions (PMA plus ionomycin for 72 hours), patient PBMC produced markedly lower levels of IL-2 and IFN-γ than in the normal controls (Fig3). On the other hand, no increase in the production of IL-4 and IL-5 was observed (Fig 3), thus ruling out the possibility that the defective proliferative response was accounted for by a shift toward Th2-type cytokine secreting cells. In view of the substantial number of T cells in the periphery, this defect was also too profound to be explained solely by the reduction of the T-cell number as compared with the controls'.

Fig. 3.

Cytokine production by total PBMC from the patient at the age of 3 months (▪) and from 10 age-matched healthy controls (▨) in response to PMA (5 ng/mL) plus ionomycin (500 ng/mL). Data of healthy controls are expressed as mean ± 1 SD.

Fig. 3.

Cytokine production by total PBMC from the patient at the age of 3 months (▪) and from 10 age-matched healthy controls (▨) in response to PMA (5 ng/mL) plus ionomycin (500 ng/mL). Data of healthy controls are expressed as mean ± 1 SD.

Close modal

We considered the possibility that defective lymphocyte proliferation and the reduced cytokine production were caused by increased cell death during the culture. Indeed, an increased proportion of apoptotic cells was detected in the patient, both in unstimulated and stimulated cultures (Fig 4).

Fig. 4.

Increased rates of apoptotic cells in unstimulated and stimulated cultures of PBMC of the patient (▪) compared with 10 age-matched healthy controls (▨). Data of healthy controls are expressed as mean ± 1 SD. All cultures were performed for 72 hours.

Fig. 4.

Increased rates of apoptotic cells in unstimulated and stimulated cultures of PBMC of the patient (▪) compared with 10 age-matched healthy controls (▨). Data of healthy controls are expressed as mean ± 1 SD. All cultures were performed for 72 hours.

Close modal

Because the products of bcl-2 gene are known to modulate the susceptibility of T cells to spontaneous apoptosis,11cytoplasmic levels of this protein were evaluated by flow cytometry. The intensity of staining for bcl-2, expressed as MESF, was significantly reduced after 48 hours of unstimulated culture of lymphocytes from the patient, as compared with that observed in three age-matched healthy controls (4,592 v 14,750 ± 555; Fig 5). Taken together, these data suggest that the raised number of spontaneously dying PBMC in the patient is accounted for by downmodulation of bcl-2 gene product expression.

Fig. 5.

The expression of bcl-2 on PBMC from the patient (continuous lines) and one representative healthy control (dashed lines) after 48 hours of unstimulated culture.

Fig. 5.

The expression of bcl-2 on PBMC from the patient (continuous lines) and one representative healthy control (dashed lines) after 48 hours of unstimulated culture.

Close modal

We report a case of a child with an AR-SCID, caused by Jak3 mutations, presenting at birth a typical TB+phenotype, who developed autologous CD3+CD4+ T cells after the first few months of life. These lymphocytes showed a primed/activated phenotype; had a severely defective proliferative response to anti-CD3, PHA, and allogeneic cells; and secreted very low levels of Th1 and Th2 cytokines, as compared with controls. Functional defects, including reduced proliferative response and defective cytokine production, were accounted for by the large number of cells dying either spontaneously (as a consequence of bcl-2 downregulation) or after activation in vitro. These data are in keeping with the notion that the response induced in T lymphocytes by antigen stimulation is influenced by their activation status, because triggering of TCR/CD3 complex of recently activated lymphocytes leads to apoptosis rather than proliferation.35-37 

Our report is similar to other atypical presentations of XL-SCID, in which the presence of autologous, poorly functioning T lymphocytes was documented.4,22-24 In particular, the patient reported by DiSanto et al22 carried a splice site mutation in the γc gene leading to two alternative transcripts, only one being functional, although present in limited amounts. Furthermore, Russell et al4 reported another XL-SCID patient with a missense mutation in the cytoplasmic region of the γcprotein that substantially diminished, but did not abrogate, its association with Jak3. In this latter case, the patient at birth had no T lymphocytes and developed CD3+CD4+ cells at the age of 19 weeks.38 These results suggest that reduced but detectable expression of γc4,22 or Jak3 (present case) proteins may be enough to generate a limited number of peripheral T-cell clones. Alternatively, other hematopoietic growth factors that do not use Jak3 (eg, IL-3, Thymic Stromal Lymphopoietin,39 or stem cell factor through its receptor c-kit40) might substitute for some of the functions of Jak3-dependent cytokines allowing the generation of T cells. However, in the absence of γc/Jak3 signals the action of these cytokines might be insufficient for the correct activation and function of these lymphocytes. In particular, IL-7 receptor signaling has been recently shown to upregulate bcl-2 expression in developing thymocytes and in mature peripheral T cells,41,42 thus maintaining their viability and function.41,42 

Interestingly, murine models of defective Jak3 function (Jak3−/− mice) also have a similar, abnormal T-cell development. T-cell differentiation in the thymus is partially conserved, despite severe cell depletion. Furthermore, an age-dependent increase of CD4+ lymphocytes is observed in the periphery. These cells have an activated/memory phenotype, fail to respond to mitogens, and show a reduced IL-2 secretion following stimulation with anti-CD3 plus anti-CD28.17-19 

The observation that, like in Jak3−/− mice, preferential accumulation of CD4+ lymphocytes was observed in our patient suggests that cytokine signaling through the γc/Jak3 pathway may be more critical for the maturation or survival of CD8 than of the CD4 lineage T cells.

In our experience, oligoclonal, in vivo-activated, poorly functioning T cells were observed in other immunodeficiencies. In Omenn's syndrome, a rare inherited immunodeficiency, peripheral blood T cells, usually present in normal number, are oligoclonal43 and show an activated phenotype and a defective proliferative response to mitogens.44 As in the case described here, we have recently shown that functional T-cell defects observed in Omenn's syndrome might be explained, at least in part, by excessive cell death resulting from two main mechanisms: spontaneous apoptosis in unstimulated cultures, associated with reduced expression of bcl-2 gene product, and susceptibility to activation-induced cell death through a Fas/Fas ligand-mediated mechanism, after restimulation in vitro with mitogens.45 Activated, anergic T cells have been also detected in combined immunodeficiency caused by functional defects (unpublished observation, February 1997), in early phases of immune reconstitution following BMT,46-48 and in trans-placental engraftment of maternal T cells occurring in some cases of SCID TB+/−.49 

In all cases, the unknown developmental pathways used by T lymphocytes seem to be antigen driven; furthermore, they lead to terminally differentiated T cells that are functionally defective because of high susceptibility to undergo cell death.

On the other hand, not much is known about the origin of these activated and anergic T lymphocytes. Refilling of a depleted T-cell compartment may arise from two main potential ways: thymopoiesis, which creates new T cells from progenitors and maintains or increases the potential diversity of T-cell repertoire, and peripheral expansion, which probably restricts that repertoire. It is unlikely that defective thymopoiesis alone may account for the aberrant T-cell phenotype observed in our patient, as in this case accumulation of “naı̈ve” T cells would be expected. Based on recent findings in murine models, indicating that the lack of expression of Jak3 in the periphery is associated with accumulation of activated, anergic CD4+ T cells in the periphery,21 it is likely that defective immune homeostasis in the periphery accounts for the development of oligoclonal, activated, and anergic T cells in our patient.

Accordingly, the data relative to the TCR diversity further support the hypothesis of an ongoing clonal expansion. The TCRBV repertoire of this patient was not simply restricted in terms of TCRBV genes usage; each of the dominant TCRBV families expressed were found to be oligoclonal. The presence of a limited number of available TCRBV chain molecules may limit antigenic recognition and thus contribute to immunodeficiency.

Finally, the observation that our patient developed hemolytic anemia after BMT, when a persistence of autologous T cells was detected, is in agreement with the observation that deletion of self-reactive T cells in the thymus and in the periphery is defective in Jak3-deficient mice,50 and thus adds to the key role played by the γc/Jak3 signaling pathway in T-lymphocyte differentiation and function.

Partially supported by grants from Telethon Italy (grant A.42 to L.D.N.). This paper is manuscript no. 12 of the Genoma 2000/ITBA Project, funded by CARIPLO.

Address reprint requests to Luigi D. Notarangelo, MD, Clinica Pediatrica, Spedali Civili Brescia, Piazza Spedali Civili 1, 25123 Brescia, Italy.

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Taniguchi
T
Cytokine signaling through nonreceptor protein tyrosine kinases.
Science
268
1995
251
2
Ihle
JN
Cytokine receptor signalling.
Nature
377
1995
591
3
Johnston
JA
Kawamura
M
Kirken
RA
Chen
YQ
Blake
TB
Shibuya
K
Ortaldo
JR
McVicar
DW
O'Shea
JJ
Phosphorylation and activation of the Jak-3 Janus kinase in response to interleukin-2.
Nature
370
1994
151
4
Russell
SM
Johnston
JA
Noguchi
M
Kawamura
M
Bacon
CM
Friedmann
M
Berg
M
McVicar
DW
Witthuhn
BA
Silvennoinen
O
Goldman
AS
Schmalstieg
FC
Ihle
JN
O'Shea
JJ
Leonard
WJ
Interaction of IL-2Rβ and γc chains with Jak1 and Jak3: Implication for XSCID and XCID.
Science
266
1994
1042
5
Miyazaki
T
Kawahara
A
Fujii
H
Nakagawa
Y
Minami
Y
Liu
Z-J
Oishi
I
Silvennoinen
O
Witthuhn
BA
Ihle
JN
Taniguchi
T
Functional activation of Jak1 and Jak3 by selective association with IL-2 receptor subunits.
Science
266
1994
1045
6
Sugamura
K
Asao
H
Kondo
M
Tanaka
N
Ishii
N
Nakamura
M
Takeshita
T
The common γ-chain for multiple cytokine receptors.
Adv Immunol
59
1995
225
7
Leonard
WJ
Shores
EW
Love
PE
Role of the common cytokine receptor γc in cytokine signaling and lymphoid development.
Immunol Rev
48
1995
97
8
Cavazzana-Calvo
M
Hacein-Bey
S
De Saint-Basile
G
De Coene
C
Selz
F
Le Deist
F
Fischer
A
Role of interleukin-2 (IL-2), IL-7, and IL-15 in natural killer cell differentiation from cord blood hematopoietic progenitor cells and from γc transduced severe combined immunodeficiency X1 bone marrow cells.
Blood
88
1996
3901
9
Nakajima
H
Shores
EW
Noguchi
M
Leonard
WJ
The common cytokine receptor γ chain plays an essential role in regulating lymphoid homeostasis.
J Exp Med
185
1997
189
10
Boussiotis
VA
Barber
DL
Nakarai
T
Freeman
GJ
Gribben
JG
Bernstein
GM
D'Andrea
AD
Ritz
J
Nadler
LM
Prevention of T cell anergy by signaling through the γc chain of the IL-2 receptor.
Science
266
1994
1039
11
Akbar
AN
Borthwick
JN
Wickremasinghe
RG
Panayoitidis
P
Pilling
D
Bofill
M
Krajewski
S
Reed
JC
Salmon
M
Interleukin-2 receptor common gamma-chain signaling cytokines regulate activated T cell apoptosis in response to growth factor withdrawal: Selective induction of anti-apoptotic (bcl-2, bcl-xL) but not pro-apoptotic (bax, bcl-xS) gene expression.
Eur J Immunol
26
1996
294
12
Noguchi
M
Yi
H
Rosenblatt
HM
Filipovich
AH
Adelstein
AH
Modi
WS
McBride
DW
Leonard
WJ
Interleukin-2 receptor γ chain mutation results in X-linked severe combined immunodeficiency in humans.
Cell
73
1993
147
13
Cao
X
Shores
EW
Hu-Li
J
Anver
MR
Kelsall
BL
Russell
SM
Drago
J
Noguchi
M
Gringerg
A
Bloom
ET
Paul
WE
Katz
SI
Love
PE
Leonard
WJ
Defective lymphoid development in mice lacking expression of the common cytokine receptor γ chain.
Immunity
2
1995
223
14
DiSanto
JP
Müller
W
Guy-Grand
D
Fischer
A
Rajewsky
K
Lymphoid development in mice with a targeted deletion of the interleukin-2 receptor γ chain.
Proc Natl Acad Sci USA
92
1995
377
15
Macchi
P
Villa
A
Giliani
S
Sacco
MG
Frattini
A
Porta
F
Ugazio
AG
Johnston
JA
Candotti
F
O'Shea
JJ
Vezzoni
P
Notarangelo
LD
Mutations of Jak-3 gene in patients with autosomal severe combined immune deficiency (SCID).
Nature
377
1995
65
16
Russell
SM
Tayebi
N
Nakajima
H
Riedy
MC
Roberts
JL
Aman
MJ
Migone
T-S
Noguchi
M
Markert
ML
Buckley
RH
O'Shea
JJ
Leonard
WJ
Mutation of Jak3 in a patient with SCID: Essential role of Jak3 in lymphoid development.
Science
270
1995
797
17
Park
SY
Saijo
K
Takahashi
T
Osawa
M
Arase
H
Hirayama
N
Miyake
K
Nakauchi
H
Shirasawa
T
Saito
T
Developmental defects of lymphoid cells in Jak3 kinase-deficient mice.
Immunity
3
1995
771
18
Thomis
DC
Gurniak
CB
Tivol
E
Sharpe
AH
Berg
LJ
Defects in B lymphocyte maturation and T lymphocyte activation in mice lacking Jak3.
Science
270
1995
794
19
Nosaka
T
van Deursen
JMA
Tripp
RA
Thierfelder
WE
Witthuhn
BA
McMickle
AP
Doherty
PC
Grosveld
GC
Ihle
JN
Defective lymphoid development in mice lacking Jak3.
Science
270
1995
800
20
Leonard
WJ
The molecular basis of X-linked severe combined immunodeficiency: Defective cytokine receptor signaling.
Annu Rev Med
47
1996
229
21
Thomis
DC
Berg
LJ
Peripheral expression of Jak3 is required to maintain T lymphocyte function.
J Exp Med
185
1997
197
22
DiSanto
J
Rieux-Laucat
F
Dautry-Varsat
A
Fischer
A
De Saint-Basile
G
Defective human interleukin 2 receptor γ chain in an atypical X chromosome-linked severe combined immunodeficiency with peripheral T cells.
Proc Natl Acad Sci USA
91
1994
9466
23
Morelon
E
Dautry-Varsat
A
Le Deist
F
Hacein-Bay
S
Fischer
A
De Saint-Basile
G
T-lymphocyte differentiation and proliferation in the absence of the cytoplasmic tail of the common cytokine receptor γc chain in a severe combined immune deficiency X1 patient.
Blood
88
1996
1708
24
Stephan
V
Wahn
V
Le Deist
F
Dirksen
U
Bröker
B
Müller-Fleckenstein
I
Horneff
G
Schroten
H
Fischer
A
De Saint-Basile
G
Atypical X-linked severe combined immunodeficiency due to possible spontaneous reversion of the genetic defect in T cells.
N Engl J Med
335
1996
1563
25
Candotti
F
Oakes
SA
Johnston
JA
Giliani
S
Schumacher
RF
Mella
P
Fiorini
M
Ugazio
AG
Badolato
R
Notarangelo
LD
Bozzi
F
Macchi
P
Strina
D
Vezzoni
P
Blaese
RM
O'Shea
JJ
Villa
A
Structural and functional basis for JAK3-deficient severe combined immunodeficiency.
Blood
90
1997
3996
26
Nicoletti
I
Migliorati
G
Pagliacci
MC
Grignani
F
Riccardi
C
A rapid and simple method for measuring thymocyte apoptosis by propidium iodide staining and flow cytometry.
J Immunol Methods
139
1991
271
27
Bettinardi
A
Brugnoni
D
Quiròs-Roldan
E
Malagoli
A
La Grutta
S
Correra
A
Notarangelo
LD
Missense mutations in the Fas gene resulting in autoimmune lymphoproliferative syndrome: A molecular and immunological analysis.
Blood
89
1997
902
28
Bettinardi
A
Imberti
L
Sottini
A
Primi
D
Analysis of amplified T cell receptor Vβ transcripts by a non-isotopic immunoassay.
J Immunol Methods
146
1992
71
29
Sottini
A
Quiròs-Roldan
E
Albertini
A
Primi
D
Imberti
L
Assessment of T cell receptor variable beta chains diversity by heteroduplex analysis.
Hum Immunol
48
1996
12
30
Imberti
L
Sottini
A
Signorini
S
Gorla
R
Primi
D
Oligoclonal CD4+CD57+ T-cell expansions contribute to the imbalanced T-cell receptor repertoire of rheumatoid arthritis patients.
Blood
89
1997
2822
31
Toyonaga
B
Yoshikai
Y
Vadasz
V
Chin
B
Mak
TW
Organization and sequences of the diversity, joining, and constant region genes of the human T-cell receptor β chain.
Proc Natl Acad Sci USA
82
1985
8624
32
Arden
B
Clark
SP
Kabelitz
D
Mak
YW
Human T-cell receptor variable gene segment families.
Immunogenetics
42
1995
455
33
Sottini A, Primi D, Signorini S, Quiròs-Roldan, Castelli F, Imberti L: Generalized restriction of the T cell receptor diversity of CD8+ cells in HIV-infected patients. (submitted)
34
Quiròs-Roldan
E
Sottini
A
Bettinardi
A
Albertini
A
Imberti
L
Primi
D
Different TCRBV genes generate biased patterns of V-D-J diversity in human T cells.
Immunogenetics
41
1995
91
35
Russel
JH
Activation-induced death of mature T cells in the regulation of immune responses.
Curr Opin Immunol
7
1995
382
36
Van Parijs
L
Abbas
AK
Role of Fas-mediated cell death in the regulation of immune responses.
Curr Opin Immunol
8
1996
355
37
Akbar
AN
Salmon
M
Cellular environments and apoptosis: Tissue microenvironments control activated T-cell death.
Immunol Today
18
1997
72
38
Schmalstieg
FC
Wirt
DP
Adkins
LT
Brooks
EG
Stansberry
SD
Swischuk
LE
Goldman
AS
Postnatal development of T lymphocytes in a novel X-linked immunodeficiency disease.
Clin Immunol Immunopathol
64
1992
71
39
Lee Friend
S
Hosier
S
Nelson
A
Foxworthe
D
Williams
DE
Farr
A
A thymic stromal cell line supports in vitro development of surface IgM+ B cells and produces a novel growth factor affecting B and T lineage cells.
Exp Hematol
22
1994
321
40
Rodewald HR, Ogawa M, Haller C, Waskow C, DiSanto JP: Pro-thymocyte expansion by c-kit and the common cytokine receptor γ chain is essential for repertoire formation. Immunity 6:265,1997
41
Maraskovsky
E
O'Reilly
LA
Teepe
M
Corcoran
LM
Peschon
JJ
Strasser
A
Bcl-2 can rescue T lymphocyte development in interleukin-7 receptor-deficient mice but not in mutant rag-1−/− mice.
Cell
89
1997
1011
42
Akashi
K
Kondo
M
von Freeden-Jeffry
U
Murray
R
Weissman
IL
Bcl-2 rescues T lymphopoiesis in interleukin-7 receptor-deficient mice.
Cell
89
1997
1033
43
De Saint-Basile
G
Le Deist
F
deVillartay
JP
Cerf-Bensussan
N
Journet
O
Brousse
N
Griscelli
C
Fischer
A
Restricted heterogeneity of T lymphocytes in combined immunodeficiency with hypereosinophilia (in Omenn's Syndrome).
J Clin Invest
87
1991
1352
44
Wirt
DP
Brooks
EG
Vaidya
AS
Klimpel
GR
Waldmann
TA
Goldblum
RL
Novel T-lymphocyte population in combined immunodeficiency with features of graft versus host disease.
N Engl J Med
32
1989
370
45
Brugnoni D, Airò P, Facchetti F, Blanzuoli L, Ugazio AG, Cattaneo R, Notarangelo LD: In vitro cell death of activated lymphocytes in Omenn's syndrome. Eur J Immunol 1997 (in press)
46
(letter)
Gorla
R
Airò
P
Ferremi-Leali
P
Rossi
G
Prati
E
Brugnoni
D
Cattaneo
R
Predominance of memory phenotype within CD4+ and CD8+ lymphocyte subsets after allogeneic BMT.
Bone Marrow Transplant
11
1993
346
47
Storek
J
Witherspoon
RP
Storb
R
T cell reconstitution after bone marrow transplantation into adult patients does not resemble T cell development in early life.
Bone Marrow Transplant
16
1995
413
48
Donnenberg
AD
Margolick
JB
Beltz
LA
Donnenberg
VS
Rinaldo
CR
Jr
Apoptosis parallels lymphopoiesis in bone marrow transplantation and HIV disease.
Res Immunol
146
1995
11
49
Sottini
A
Quiròs-Roldan
E
Notarangelo
LD
Malagoli
A
Primi
D
Imberti
L
Engrafted maternal T cells in a severe combined immunodeficiency patient express T-cell receptor variable beta segments characterized by a restricted V-D-J junctional diversity.
Blood
85
1995
2105
50
Saijo
K
Park
SY
Ishida
Y
Arase
H
Saito
T
Crucial role of Jak3 in negative selection of self-reactive T cells.
J Exp Med
185
1997
351
Sign in via your Institution