Skip to main content
Clinical and Experimental Immunology logoLink to Clinical and Experimental Immunology
. 1998 Feb;111(2):339–344. doi: 10.1046/j.1365-2249.1998.00509.x

Expression of Pax5 gene in human haematopoietic cells and tissues: comparison with immunodeficient donors

H Kaneko *, T Ariyasu *, R Inoue *, T Fukao *, K Kasahara *, T Teramoto *, E Matsui *, S Hayakawa *, N Kondo *
PMCID: PMC1904901  PMID: 9486401

Abstract

In mice, Pax5 gene is indispensable for B cell development. Pax5-deficient mice fail to produce mature B cells owing to complete arrest of B cell development at a precursor stage. However, the lineage and stage of human Pax5 gene expression have remained elusive. In this investigation expression of the human Pax5 gene was studied. Pax5 gene expression was detected in B cell lines but not in myeloma cell lines. CD19 expression was correlated with Pax5 gene expression. Adult spleen and bone marrow and fetal spleen and liver showed strong Pax5 gene expression, as did the corresponding mouse tissues, as reported previously. In common variable immunodeficiency (CVID) peripheral blood lymphocytes (PBL) with a decreased number of B cells, no Pax5 gene expression was detected. Some CVID PBL stimulated with IL-2, IL-10 and anti-CD40 monoclonal antibody, expressed the Pax5 gene. Defect of Pax5 gene expression in CVID may be caused by regulatory T cell disorder.

Keywords: Pax5, CD19, CD40, RT-PCR, common variable immunodeficiency

INTRODUCTION

The Pax5 gene encodes a B cell-specific activator protein (BSAP), which has been identified as a transcriptional factor that is expressed at early, but not late, stages of B cell differentiation [1]. Various binding sites for Pax5 in the promotors of B cell-specific genes have been identified, including sites in the promotors of the genes encoding CD19 [2], VpreB [3] and Blk [4], as well as multiple sites within the IgH locus, including a region upstream of Sγ2a and Sε [5]. Furthermore, Pax5 was identical to Sα-bp, which binds to two sites upstream of Cα, as well as to Sμ binding proteins [68]. Over-expression of Pax5 in splenic B cells stimulates proliferation of these B cells [9]. Experiments performed using mice that were Pax5-deficient due to targeted gene disruption revealed that Pax5-deficient mice fail to produce small preB, B and plasma cells, owing to a complete arrest of B cell development at an early precursor stage [10]. The expression and function of the Pax5 gene in mouse cells and tissues have been extensively investigated [1113]. It is assumed that the Pax5 gene is specifically expressed in mouse B cell lineage haematopoietic cells. However, the lineage specificity of Pax5 gene expression in human cells remains obscure.

Common variable immunodeficiency (CVID) is characterized by recurrent infection and decreased serum immunoglobulin levels [14]. Some cases of CVID are associated with a complete absence of surface IgM+ B cells and immunoglobulins of all three major isotypes, as in Pax5-deficient mice [10]. In this study we analyse the expression of the human Pax5 gene in various haematopoietic cell lines and tissues and in CVID peripheral blood lymphocytes (PBL). In human cell lines the Pax5 gene was specifically expressed in B lineage cells, which expressed CD19. Myeloma cell lines did not express the Pax5 gene. In CVID with a decreased number of mature B cells among PBL, Pax5 gene expression was not detected. Stimulation with anti-CD40 MoAb and cytokines induced Pax5 expression in some CVID PBL. We discuss the role of Pax5 in human B cell differentiation and proliferation.

MATERIALS AND METHODS

Cell lines and human tissues

Human cell lines were cultured in RPMI medium with 10% fetal calf serum (FCS). The cell lines used in this study are listed in Table 1. PBL were prepared from heparinized blood using Ficoll–Hypaque. Human adult tissues were obtained at autopsy and fetal tissues at abortion before 24 weeks of pregnancy. Informed consent was obtained before the tissues were collected.

Table 1.

Expression of Pax5 gene and CD marker in various haematopoietic cell lines [1628]

graphic file with name cei0111-0339-t1.jpg

Immunophenotypic analysis

Cells were stained with a panel of diagnostic reagents in suspension using the direct and indirect immunofluorescence technique [15]. Briefly, cells were first incubated with a specific MoAb, in excess, for 30 min. In cases in which cytoplasmic immunoglobulin was stained, the cells were preincubated with ice-cold methanol. After being washed, cells were stained with FITC-conjugated goat anti-mouse F(ab′)2 isotype-specific antisera (Cappel, CA). Testing for terminal deoxy-transferase (TdT) was done on methanol-fixed mononuclear cell smears with rabbit anti-calf thymus TdT antiserum followed by staining with FITC-conjugated goat anti-rabbit IgG (Cappel).

Reverse transcriptase-polymerase chain reaction for detecting mRNA expression

RNA was extracted from cell and tissues using Isogen (Nippon Gene, Toyama, Japan). cDNA was synthesized with MMTV reverse transcriptase using 1 μg or 100 ng or 10 ng of RNA and oligo dT-primer.

The polymerase chain reaction (PCR) primers were as follows: Pax5 sense 5′- AATGACACCGTGCCTAGCGT-3′, Pax5 antisense 5′-GGTGGTGAAGATGTCTGAGT-3′; CD19 sense 5′-TAAGTCATTGCTGAGCCTAGA-3′, CD19 antisense 5′-TCGCTGCTCGGGTTTCCATAA-3′; β-actin sense 5′-TGACGGGGTCACCCACACTGTGCCCATCTA-3′, β-actin antisense 5′-CTAGAAGCATTTGCGGTGGACGATGGAGGG-3′.

PCR was performed for 15–35 cycles (Fig. 1) in a PCR thermal cycler (Takara, Ootsu, Japan) at a denaturation temperature of 94°C for 1 min and extension at 72°C for 1 min; the annealing temperature was varied accordingly to the melting temperature for each specific pair of primers. The PCR products (10 μl of a total of 50 μl) were electrophoresed in a 8% acrylamide gel for Pax5 gene or 2% agarose gel for the other genes, stained with ethidium bromide and visualized using UV light.

Fig. 1.

Fig. 1

Condition of reverse transcriptase-polymerase chain reaction (RT-PCR) for detection of Pax5 gene. cDNA was synthesized using: A, 10 ng mRNA prepared from Epstein–Barr virus (EBV)-transformed cell line; B, 100 ng mRNA; C, 1 μg mRNA. Lane 1, 15 cycles; lane 2, 20 cycles; lane 3, 25 cycles; lane 4, 30 cycles; lane 5, 35 cycles.

Stimulation with anti-CD40 MoAb, IL-2 and IL-10

PBL from case 3 or 5 were cultured in RPMI medium containing 10% FCS, 1% anti-CD40 MoAb (B-B20; Serotec, Oxford, UK), 50 U/ml of human IL-2 (Shionogi, Osaka, Japan) and 100 ng/ml human IL-10 (Genzyme, Cambridge, MA). After 3 days of culture, cells were washed with PBS three times and collected by centrifugation. Cells were then resuspended in the medium containing anti-CD40 MoAb, IL-2 and IL-10, or IL-2 and IL-10 and incubated for 4 days. After the cells were washed, RNA was extracted from them.

RESULTS

Condition of reverse transcriptase-PCR for detection of Pax5 gene

Semiquantification of Pax5 gene expression was performed by amplification of 10 ng, 100 ng and 1 μg mRNA prepared with 15–35 cycles from Epstein–Barr virus (EBV)-transformed cell line (Fig. 1). The faint band was visible when 100 ng mRNA were amplified with 25 cycles. In this study positive expression of Pax5 gene with the PCR conditions of 1 μg mRNA and 30 cycles was defined as showing the band intensity stronger than the PCR conditions with 100 ng mRNA and 30 cycles or with 1 μg mRNA and 25 cycles. Reverse transcriptase (RT)-PCR was then done using 1 μg mRNA and 30 cycles.

Pax5 gene expression in B cell lines and non-B cell lines

Most B cell lines we examined showed Pax5 gene expression (Fig. 2). In two myeloma cell lines, RPMI8226 and U-266, Pax5 gene expression was not detected (Table 1). In non-B cell lines, including myelomonocytic and megakaryocytic cell lines such as HL60 and MOLM-1, Pax5 gene expression was detected. In connection with CD19 surface expression, all cell lines showing CD19 surface expression we examined showed Pax5 gene expression. Expression of Tdt, immunoglobulin, CD13 and CD34 did not correlate with Pax5 gene expression.

Fig. 2.

Fig. 2

Pax5 and CD19 gene expression in various human cell lines. Lane 1, LAZ221; lane 2, NALM-1; lane 3, NALM-6; lane 4, KOPN-8; lane 5, DND-41; lane 6, NALM-19; lane 7, NALM-20; lane 8, KG-1; lane 9, MOLM-1; lane 10, HL-60.

Pax5 gene expression in adult and fetal cells and tissues

In adult PBL, bone marrow and spleen Pax5 gene expression was detected (Fig. 3). In some cord blood samples, Pax5 gene expression was scarcely detected, although cord blood samples showed variation in Pax5 PCR products (data not shown). In fetal spleen and liver, Pax5 gene expression was detected. In fetal and adult brain Pax5 gene was scarcely detected.

Fig. 3.

Fig. 3

Pax5 gene expression in human tissues. Lane 1, adult PBL; lane 2, cord blood; lane 3, adult bone marrow; lane 4, adult liver; lane 5, adult kidney; lane 6, adult spleen; lane 7, adult brain; lane 8, fetal liver; lane 9, fetal kidney; lane 10, fetal spleen; lane 11, fetal brain.

Pax5 gene expression in CVID PBL

In all CVID cases except case 7, decreased serum IgM, IgG and IgA levels were detected (Table 2). Some patients showed immunological phenotype-like X-linked aggamaglobulinaemia such as male and absence of surface immunoglobulin-positive cells, but all patients had no family history for immunodeficiency. Btk protein was detected in cases 1 and 2 (data not shown) [29]. Therefore we tentatively classified the patients as CVID. In case 7 the serum IgM level was almost normal but the serum IgG and IgA levels were decreased. No Pax5 gene expression was detected in PBL from any CVID cases except case 7 (Fig. 4a). Deduced from the comparison of the number of control surface IgM+ and CD19+ cells with CVID, CVID B cell number of PBL was not less than 10-fold dilution of control B cell number. Pax5 gene mRNA was detected in a sample of 10-fold diluted control PBL mRNA (Fig. 4b). This suggests that the decreased Pax5 gene expression in CVID PBL was due not only to the decreased number of B cells among the CVID PBL, but also to decreased Pax5 gene expression in a single CVID B cell compared with that in a single control B cell.

Table 2.

Immunological feature in immunodeficient patients

graphic file with name cei0111-0339-t2.jpg

Fig. 4.

Fig. 4

(a) Pax5 gene expression in common variable immunodeficiency (CVID) PBL. Lanes 1 and 2, control PBL; lanes 3–9, CVID cases 1–7. (b) cDNA was synthesized using: lane 1, 1000 ng mRNA prepared from control PBL; lane 2, 100 ng mRNA; lane 3, 10 n g mRNA; lane 4, 1 ng mRNA; lanes 5–9, CVID cases 1–5 PBL 1000 ng mRNA.

Induction of Pax5 gene expression in some CVID PBL stimulated with anti-CD40 MoAb, IL-2 and IL-10

CVID PBL were stimulated with anti-CD40 MoAb, IL-2 and IL-10. Arpin et al. reported that such stimulation induced generation of memory B cells and plasma cells in vitro [30]. In case 5, the stimulation induced Pax5 gene expression in CVID PBL (Fig. 5). Pax5 gene expression was stronger in CVID PBL incubated with anti-CD40 MoAb, IL-2 and IL-10 for 7 days than in CVID PBL incubated with anti-CD40 MoAb, IL-2 and IL-10 for 3 days and then IL-2 and IL-10 for 4 days. In case 3, Pax5 gene expression was slightly induced when incubated with anti-CD40 MoAb, IL-2 and IL-10 for 7 days.

Fig. 5.

Fig. 5

Pax5 gene expression in common variable immunodeficiency (CVID) PBL stimulated with CD40, IL-2 and IL-10. Lane 1, CVID case3 PBL, no stimulation; lane 2, case 3 PBL, stimulation with CD40, IL-2 and IL-10; lane 3, case 3 PBL, stimulation with IL-2 and IL-10; lane 4, CVID case 5 PBL, no stimulation; lane 5, case 5 PBL, stimulation with CD40, IL-2 and IL-10; lane 6, case 5 PBL, stimulation with IL-2 and IL-10.

DISCUSSION

In this study it was revealed that the human Pax5 gene is expressed in B cell lines and some non-B cell lines. Moreover, CD19 expression is correlated with Pax5 gene expression; that is, all cell lines expressing CD19 we examined expressed the Pax5 gene. Myelomonocytic or megakaryocytic cell lines, such as HL60 and MOLM-1, showed Pax5 gene but not CD19 expression. Expression of Tdt, immunoglobulin, CD13 and CD34 did not correlate with Pax5 gene expression. These results support the hypothesis that Pax5 gene expression is coupled with CD19 expression [2]. In human adult or fetal tissues, where B lymphocytes were given a differentiation and proliferation including fetal liver, Pax5 gene was expressed. This observation is consistent with data obtained in experiments on mice which indicated that the Pax5 gene is expressed in two waves during mouse embryogenesis, with the first wave of expression resulting from transcription of the Pax5 gene in the developing central nervous system (CNS) and the second wave resulting from transcription of the Pax5 gene in fetal liver with B lymphopoiesis [1]. In our samples of fetal and adult brain, Pax5 gene expression was scarcely detected. This may suggest that in the human CNS, as in the mouse CNS, the Pax5 gene shows a spatial and temporal expression pattern in the developing CNS, and the fetal and adult brain tissues examined in this study were prepared when Pax5 gene expression was decreased, although further investigation to test this hypothesis is required.

In CVID PBL, Pax5 gene expression was not detected. This seems to reflect the decreased number of B cells among the PBL. It is of interest that cord blood from some normal infants scarcely expressed Pax5 gene. CVID patients often have immature B lymphocytes, similar to newborn infants. They have in common a markedly reduced expression of CD40 ligand and often low production of interferon-gamma (IFN-γ) and IL-4 [3133]. In both instances, circulating B cells seem to be naive rather than defective. In some CVID PBL stimulated with anti-CD40 MoAb, IL-2 and IL-10, Pax5 gene expression was detected. Although the number of B cells among the CVID PBL was very small, these B cells had the ability to respond to anti-CD40 MoAb and cytokine stimulation. Consistent with our data, Eisenstein et al. reported that anti-CD40 MoAb and IL-10 induced differentiation of CVID B cells [34]. Thus, it seems that the possible explanation for lack of Pax5 gene expression in PBL of CVID patients and the induction of Pax5 gene expression by activated B cells of CVID patients reflects the immature status of B cells of these patients, probably due to a T cell defect that induces circulating B cells to activation or maturation [35].

Vorechovsky et al. analysed the Pax5 gene of mutation in the affected individuals from each multiplex family, 10 patients with sporadic CVID and 15 patients with sporadic IgA deficiency using PCR-single-strand conformation polymorphism (SSCP) and Southern blotting analyses, and detected no mutations [36]. Meffre et al. reported a human non X-linked agammaglobulinaemia immunodeficiency disease characterized by blockage of B cell development at early proB cell stage, which showed that Pax5 cDNA was normal [37]. It is known that lesions in human Pax3 and Pax6 genes cause Waardenburg's syndrome and aniridia [38, 39]. This evidence indicates that the Pax proteins are important regulators in early development. In humans, Pax5 deficiency might be involved in the development and implicate human neoplasia of B cell [40, 41] or neurological disorder other than immunodeficiency.

Acknowledgments

We thanks Drs S. Tsukada, (Department of Medicine III, Osaka University School of Medicine) and S. Arai (Hayashibara Biochemical Laboratories Inc., Okayama) for a gift of anti-Btk monoclonal antibody.

References

  • 1.Adams B, Dorfler P, Aguzzi A, Kozmik Z, Urbanek P, Maurer-Fogy I, Busslinger M. Pax-5 encodes the transcriptional factor BSAP and is expressed in B lymphocytes, the developing CNS, and adults testis. Genes Dev. 1992;6:1589–607. doi: 10.1101/gad.6.9.1589. [DOI] [PubMed] [Google Scholar]
  • 2.Kozmik Z, Wang S, Dorfler P, Adams B, Busslinger M. The promoter of the CD19 gene is a target for the B-cell-specific transcription factor BSAP. Mol Cell Biol. 1992;12:2662–72. doi: 10.1128/mcb.12.6.2662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Okabe T, Watanabe T, Kudo A. A pre-B- and B cell-specific DNA-binding protein, EBB-1, which binds to the promoter of the VpreB1 gene. Eur J Immunol. 1992;22:37–43. doi: 10.1002/eji.1830220107. [DOI] [PubMed] [Google Scholar]
  • 4.Zwollo P, Desiderio S. Specific recognition of the blk promoter by the B-lymphoid transcription factor B-cell-specific activator protein. J Biol Chem. 1994;269:15310–7. [PubMed] [Google Scholar]
  • 5.Rothman P, Li SC, Gorham B, Glimcher L, Alt F, Boothby M. Identification of a conserved lipopolysaccharide-plus -interleukin-4-responsive element located at the promoter of germ line ε transcripts. Mol Cell Biol. 1991;11:5551–61. doi: 10.1128/mcb.11.11.5551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Liao F, Giannini SL, Birshtein BK. A nuclear DNA-binding protein expressed during early stages of B cell differentiation interacts with diverse segments within and 3′ of the IgH chain gene cluster. J Immunol. 1992;148:2909–17. [PubMed] [Google Scholar]
  • 7.Singh M, Birshtein K. NF-HB (BSAP) is a repressor of the murine immunoglobulin heavy-chain 3′α enhancer at early stage of B-cell differentiation. Mol Cell Biol. 1993;13:3611–22. doi: 10.1128/mcb.13.6.3611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Neurath MF, Strober W, Wakatsuki Y. The murine Ig3′α enhancer is a target site with repressor function for B cell lineage-specific transcription factor BSAP (NF-HB, Sα-BP) J Immunol. 1994;153:730–42. [PubMed] [Google Scholar]
  • 9.Wakatsuki YW, Neurath MF, Max EE, Strober W. The B cell-specific transcription factor BSAP regulates B cell proliferation. J Exp Med. 1994;179:1099–108. doi: 10.1084/jem.179.4.1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Urbanek P, Wang ZQ, Fetka I, Wagner EF, Busslinger M. Complete block of early B cell differentiation and altered patterning of the posterior midbrain in mice lacking Pax5/BSAP. Cell. 1994;79:901–12. doi: 10.1016/0092-8674(94)90079-5. [DOI] [PubMed] [Google Scholar]
  • 11.Neurath MF, Stuber ER, Strober W. BSAP: a key regulator of B-cell development and differentiation. Immunol Today. 1995;16:564–9. doi: 10.1016/0167-5699(95)80078-6. [DOI] [PubMed] [Google Scholar]
  • 12.Shivdasani RA, Orkin SH. The transcriptional control of hematopoiesis. Blood. 1996;87:4025–39. [PubMed] [Google Scholar]
  • 13.Michaelson JS, Singh M, Birshten BK. B cell lineage-specific activator protein (BSAP) a player at multiple stages of B cell development. J Immunol. 1996;156:2349–51. [PubMed] [Google Scholar]
  • 14.Kaneko H, Kondo N, Motoyoshi F, Mori S, Kobayashi Y, Inoue Y, Orii T. Expression of immunoglobulin genes in common variable immunodeficiency. J Clin Immunol. 1991;11:262–7. doi: 10.1007/BF00918184. [DOI] [PubMed] [Google Scholar]
  • 15.Ariyasu T, Kimura N, Kikuchi M, Kohno K, Sugamura K, Watanabe T, Minowada J. Induced natural killer -like cytotoxic function in the TCR δ-1 positive human leukemic T-cell lines. Leukemia. 1991;5:807–12. [PubMed] [Google Scholar]
  • 16.Lazarus H, Barell EF, Krishan A, Livingston DM, Harris K, Schlossman SF, Chess L. Characterization of a unique cell line (LAZ 221) from human acute lymphocytic (“null” cell) leukemia. Cancer Res. 1978;38:1362–7. [PubMed] [Google Scholar]
  • 17.Minowada J, Tsubota T, Greaves MF, Walters TR. A non-T, non-B human leukemia cell line (NALM-1): establishment of the cell line and presence of leukemia-associated antigens. J Natl Cancer Inst. 1977;59:83–7. doi: 10.1093/jnci/59.1.83. [DOI] [PubMed] [Google Scholar]
  • 18.Han T, Minowada J. Use of stimulating capacity of mixed lymphocyte reaction (MLR-S) as a possible marker for the cell-origin of null-cell acute lymphoblastic leukaemia. Immunology. 1978;35:333–9. [PMC free article] [PubMed] [Google Scholar]
  • 19.Vallera DA, Quinones RR, Azemove SM, Soderling CC. Monoclonal antibody-toxin conjugates reactive against human T lymphocytes. A comparison of antibody linked to intact ricin toxin with antibody linked to ricin A chain. Transplantation. 1984;37:387–92. doi: 10.1097/00007890-198404000-00015. [DOI] [PubMed] [Google Scholar]
  • 20.Matsuo Y, Ariyasu T, Adachi T, Tsubota T, Imanishi J, Minowada J. Establishment and characterization of a bi-phenotypic leukemic cell line, NALM-19, from a patient with acute leukemia. Hum Cell. 1991;4:257–60. [PubMed] [Google Scholar]
  • 21.Matsuo Y, Ariyasu T, Ohmoto E, Kimura I, Minowada J. Bi-phenotypic t(9;22)-positive leukemia cell lines from a patient with acute leukemia: NALM-20, established at the onset; and NALM-21, NALM-22 and NALM-23, established after relapse. Hum Cell. 1991;4:335–8. [PubMed] [Google Scholar]
  • 22.Krueger RG, Staneck LD, Boehlecke JM. Tumor-associated antigens in human myeloma. J Natl Cancer Inst. 1976;56:711–5. doi: 10.1093/jnci/56.4.711. [DOI] [PubMed] [Google Scholar]
  • 23.Han T, Dadey B, Minowada J, Golde DW. Human myeloma cells and their strong stimulating capacity in ‘one-way’ mixed lymphocyte reaction: a comparative study with leukaemic B lymphoid cells. Immunology. 1979;38:63–8. [PMC free article] [PubMed] [Google Scholar]
  • 24.Seon BK, Negoro S, Barcos MP, Tebbi CK, Chervinsky D, Fukukawa T. Monoclonal antibody SN2 defining a human T cell leukemia-associated cell surface glycoprotein. J Immunol. 1984;132:2089–95. [PubMed] [Google Scholar]
  • 25.Morikawa S, Tatsumi E, Baba M, Harada T, Yasuhira K. Two E-rosette-forming lymphoid cell lines. Int J Cancer. 1978;21:166–70. doi: 10.1002/ijc.2910210207. [DOI] [PubMed] [Google Scholar]
  • 26.Koeffler HP, Lowe L, Golde DW. Amygdalin (Laetrile): effect on clonogenic cells from human myeloid leukemia cell lines and normal human marrow. Cancer Treat Rep. 1980;64:105–9. [PubMed] [Google Scholar]
  • 27.Breitman TR, Selonick SE, Collins SJ. Induction of differentiation of the human promyelocytic leukemia cell line (HL-60) by retinoic acid. Proc Natl Acad Sci USA. 1980;77:2936–40. doi: 10.1073/pnas.77.5.2936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Matsuo Y, Adachi T, Tsubota T, Imanishi J, Minowada J. Establishment and characterization of a novel megakaryoblastic cell line, MOLM-1, from a patient with chronic myelogenous leukemia. Hum Cell. 1991;4:261–4. [PubMed] [Google Scholar]
  • 29.Hashimoto S, Tsukada S, Matsushita M, et al. Identification of Bruton's tyrosine kinase (Btk) gene mutations and characterization of derived proteins in 35 X-linked agammaglobulinemia families: a nationwide study of Btk deficiency in Japan. Blood. 1996;88:561–73. [PubMed] [Google Scholar]
  • 30.Arpin C, Dechanet J, Kooten CV, Merville P, Grouard G, Briere F, Banchereau J, Liu YF. Generation of memory B cells and plasma cells in vitro. Science. 1995;268:720–2. doi: 10.1126/science.7537388. [DOI] [PubMed] [Google Scholar]
  • 31.Fuleihan R, Ahern D, Geha RS. Decreased expression of the ligand for CD40 in newborn lymphocytes. Eur J Immunol. 1994;24:1925–8. doi: 10.1002/eji.1830240832. [DOI] [PubMed] [Google Scholar]
  • 32.North ME, Ivory K, Funauchi M, Webster AD, Lane AC, Farrant J. Intracellular cytokine production by human CD4+ and CD8+ T cells from normal and immunodeficient donors using directly conjugated anti-cytokine antibodies and three-colour flow cytometry. Clin Exp Immunol. 1996;105:517–22. doi: 10.1046/j.1365-2249.1996.d01-795.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ferrer JM, Iglesias J, Hernandez M, Matamoros N. Alternations in interleukin secretion (IL-2 and IL-4) by CD4 and CD4 CD45RO cells from common variable immunodeficiency (CVI) patients. Clin Exp Immunol. 1995;102:286–9. doi: 10.1111/j.1365-2249.1995.tb03779.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Eisenstein EM, Chua K, Strober W. B cell differentiation defects in common variable immunodeficiency are ameliorated after stimulation with anti-CD40 antibody and IL10. J Immunol. 1994;152:5957–68. [PubMed] [Google Scholar]
  • 35.Funauchi M, Farrant J, Moreno C, Webster AD. Defects in antigen-driven lymphocyte responses in common variable immunodeficiency (CVID) are due to a reduction in the number of antigen-specific CD4+ T cells. Clin Exp Immunol. 1995;101:82–8. doi: 10.1111/j.1365-2249.1995.tb02281.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Vorechovsky I, Koskinen S, Paganelli R, Smith CIE, Busslinger M, Hammarstrom L. The Pax5 gene: a linkage and mutation analysis in candidate human primary immunodeficiency. Immunogenetics. 1995;42:149–52. doi: 10.1007/BF00178590. [DOI] [PubMed] [Google Scholar]
  • 37.Meffre E, LeDeist F, de Saint-Basile G, Deville A, Fougereau M, Fischer A, Schiff C. A human non-XLA immunodeficiency disease characterized by blockage of B cell development at an early proB cell stage. J Clin Invest. 1996;98:1519–26. doi: 10.1172/JCI118943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Baldwin CT, Hoth CF, Amos JA, da-Silva EO, Milunsky A. An exonic mutation in the HuP2 paired domain gene causes Waardenburg's syndrome. Nature. 1992;355:637–8. doi: 10.1038/355637a0. [DOI] [PubMed] [Google Scholar]
  • 39.Tassabehji M, Read AP, Newton VE, Harris R, Balling R, Gruss P, Strachan T. Waardenburg's syndrome patients have mutations in the human homologue of Pax-3 paired box gene. Nature. 1992;355:635–6. doi: 10.1038/355635a0. [DOI] [PubMed] [Google Scholar]
  • 40.Mahmoud MS, Huang N, Nobuyoshi M, Lisukov IA, Tanaka H, Kawano MM. Altered expression of Pax-5 gene in human myeloma cells. Blood. 1996;87:4311–5. [PubMed] [Google Scholar]
  • 41.Iida S, Rao PH, Nallasivam P, et al. The t(9; 14)(p13; q32) chromosomal translocation associated with lymphoplasmacytoid lymphoma involves the Pax-5 gene. Blood. 1996;88:4110–7. [PubMed] [Google Scholar]

Articles from Clinical and Experimental Immunology are provided here courtesy of British Society for Immunology

RESOURCES