Entry - #308230 - IMMUNODEFICIENCY WITH HYPER-IgM, TYPE 1; HIGM1 - OMIM
# 308230

IMMUNODEFICIENCY WITH HYPER-IgM, TYPE 1; HIGM1


Alternative titles; symbols

HYPER-IgM IMMUNODEFICIENCY, X-LINKED; XHIM
HYPER-IgM SYNDROME 1
HYPER-IgM SYNDROME; HIGM; IHIS
IMMUNODEFICIENCY 3; IMD3


Phenotype-Gene Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Gene/Locus Gene/Locus
MIM number
Xq26.3 Immunodeficiency, X-linked, with hyper-IgM 308230 XLR 3 TNFSF5 300386
Clinical Synopsis
 
Phenotypic Series
 

INHERITANCE
- X-linked recessive
HEAD & NECK
Mouth
- Tonsillar hypertrophy
- Gingivitis
- Ulcerative stomatitis
ABDOMEN
Liver
- Hepatomegaly
- Chronic hepatitis
Spleen
- Splenomegaly
Gastrointestinal
- Diarrhea
- Proctitis
HEMATOLOGY
- Neutropenia, chronic or cyclic
- Amemia
- Hemolytic anemia
- Thrombocytopenia
IMMUNOLOGY
- Immunodeficiency
- Dysgammaglobulinemia
- Primary dysfunction of B-lymphocyte isotype switching and memory B-cell generation
- Lymph nodes lack germinal centers
- Normal or increased IgM
- Serum IgA, IgG, and IgE severely deficient
- B-cell count normal
- Decreased T cell activation
MISCELLANEOUS
- Recurrent bacterial infections with onset in the first or second year of life
- Pneumocytosis carinii infection (12 to 42%)
- Opportunistic infections
MOLECULAR BASIS
- Caused by mutation in the tumor necrosis factor ligand superfamily, member 5 gene (TNFSF5, 300386.0001)
Immunodeficiency with hyper-IgM - PS308230 - 5 Entries
Immunodeficiency (select examples) - PS300755 - 130 Entries
Location Phenotype Inheritance Phenotype
mapping key
Phenotype
MIM number
Gene/Locus Gene/Locus
MIM number
1p36.33 Immunodeficiency 38 AR 3 616126 ISG15 147571
1p36.33 ?Immunodeficiency 16 AR 3 615593 TNFRSF4 600315
1p36.23 Immunodeficiency 109 with lymphoproliferation AR 3 620282 TNFRSF9 602250
1p36.22 Immunodeficiency 14B, autosomal recessive AR 3 619281 PIK3CD 602839
1p36.22 Immunodeficiency 14A, autosomal dominant AD 3 615513 PIK3CD 602839
1p35.2 Immunodeficiency 22 AR 3 615758 LCK 153390
1p34.2 Immunodeficiency 24 AR 3 615897 CTPS1 123860
1p22.3 ?Immunodeficiency 37 AR 3 616098 BCL10 603517
1q21.3 Immunodeficiency 42 AR 3 616622 RORC 602943
1q23.3 Immunodeficiency 20 AR 3 615707 FCGR3A 146740
1q24.2 ?Immunodeficiency 25 AR 3 610163 CD247 186780
1q25.3 Immunodeficiency 133 with autoimmunity and autoinflammation AR 3 620565 ARPC5 604227
1q25.3 Immunodeficiency 70 AD 3 618969 IVNS1ABP 609209
1q31.3-q32.1 Immunodeficiency 105, severe combined AR 3 619924 PTPRC 151460
2p16.1 Immunodeficiency 92 AR 3 619652 REL 164910
2p11.2 Immunodeficiency 116 AR 3 608957 CD8A 186910
2q11.2 Immunodeficiency 48 AR 3 269840 ZAP70 176947
2q24.2 Immunodeficiency 95 AR 3 619773 IFIH1 606951
2q32.2 Immunodeficiency 31B, mycobacterial and viral infections, autosomal recessive AR 3 613796 STAT1 600555
2q32.2 Immunodeficiency 31C, chronic mucocutaneous candidiasis, autosomal dominant AD 3 614162 STAT1 600555
2q32.2 Immunodeficiency 31A, mycobacteriosis, autosomal dominant AD 3 614892 STAT1 600555
3p22.2 Immunodeficiency 68 AR 3 612260 MYD88 602170
3q21.3 Immunodeficiency 21 AD 3 614172 GATA2 137295
3q29 Immunodeficiency 46 AR 3 616740 TFRC 190010
4q24 Immunodeficiency 75 AR 3 619126 TET2 612839
4q35.1 {Immunodeficiency 83, susceptibility to viral infections} AD, AR 3 613002 TLR3 603029
5p15.2 {Immunodeficiency 107, susceptibility to invasive staphylococcus aureus infection} AD 3 619986 OTULIN 615712
5p13.2 Immunodeficiency 104, severe combined AR 3 608971 IL7R 146661
5q11.2 ?Immunodeficiency 94 with autoinflammation and dysmorphic facies AD 3 619750 IL6ST 600694
5q13.1 Immunodeficiency 36 AD 3 616005 PIK3R1 171833
5q31.1 Immunodeficiency 93 and hypertrophic cardiomyopathy AR 3 619705 FNIP1 610594
5q31.1 Immunodeficiency 117, mycobacteriosis, autosomal recessive AR 3 620668 IRF1 147575
5q33.3 Immunodeficiency 29, mycobacteriosis AR 3 614890 IL12B 161561
5q35.1 Immunodeficiency 40 AR 3 616433 DOCK2 603122
5q35.1 Immunodeficiency 81 AR 3 619374 LCP2 601603
6p25.2 Immunodeficiency 57 with autoinflammation AR 3 618108 RIPK1 603453
6p21.31 Immunodeficiency 87 and autoimmunity AR 3 619573 DEF6 610094
6q14.1 Immunodeficiency 23 AR 3 615816 PGM3 172100
6q15 Immunodeficiency 60 and autoimmunity AD 3 618394 BACH2 605394
6q23.3 Immunodeficiency 27A, mycobacteriosis, AR AR 3 209950 IFNGR1 107470
6q23.3 Immunodeficiency 27B, mycobacteriosis, AD AD 3 615978 IFNGR1 107470
7p22.2 Immunodeficiency 11A AR 3 615206 CARD11 607210
7p22.2 Immunodeficiency 11B with atopic dermatitis AD 3 617638 CARD11 607210
7q22.1 Immunodeficiency 71 with inflammatory disease and congenital thrombocytopenia AR 3 617718 ARPC1B 604223
7q22.3 Immunodeficiency 97 with autoinflammation AR 3 619802 PIK3CG 601232
8p11.21 Immunodeficiency 15B AR 3 615592 IKBKB 603258
8p11.21 Immunodeficiency 15A AD 3 618204 IKBKB 603258
8q11.21 Immunodeficiency 26, with or without neurologic abnormalities AR 3 615966 PRKDC 600899
8q11.21 Immunodeficiency 54 AR 3 609981 MCM4 602638
9q22.2 Immunodeficiency 82 with systemic inflammation AD 3 619381 SYK 600085
9q34.3 Immunodeficiency 103, susceptibility to fungal infection AR 3 212050 CARD9 607212
10p15.1 Immunodeficiency 41 with lymphoproliferation and autoimmunity AR 3 606367 IL2RA 147730
10p13 Immunodeficiency 80 with or without cardiomyopathy AR 3 619313 MCM10 609357
11p15.5 ?Immunodeficiency 39 AR 3 616345 IRF7 605047
11p15.4 Immunodeficiency 10 AR 3 612783 STIM1 605921
11q12.1 Immunodeficiency 77 AD 3 619223 MPEG1 610390
11q13.3 Immunodeficiency 90 with encephalopathy, functional hyposplenia, and hepatic dysfunction AR 3 613759 FADD 602457
11q23.3 Immunodeficiency 18, SCID variant AR 3 615615 CD3E 186830
11q23.3 Immunodeficiency 18 AR 3 615615 CD3E 186830
11q23.3 Immunodeficiency 19, severe combined AR 3 615617 CD3D 186790
11q23.3 Immunodeficiency 17, CD3 gamma deficient AR 3 615607 CD3G 186740
11q23.3 ?Immunodeficiency 59 and hypoglycemia AR 3 233600 HYOU1 601746
12p13.31 Immunodeficiency 79 AR 3 619238 CD4 186940
12q12 Immunodeficiency 67 AR 3 607676 IRAK4 606883
12q13.13-q13.2 Immunodeficiency 72 with autoinflammation AR 3 618982 NCKAP1L 141180
12q13.3 Immunodeficiency 44 AR 3 616636 STAT2 600556
12q15 ?Immunodeficiency 69, mycobacteriosis AR 3 618963 IFNG 147570
12q24.13 Immunodeficiency 100 with pulmonary alveolar proteinosis and hypogammaglobulinemia AD 3 618042 OAS1 164350
12q24.31 Immunodeficiency 9 AR 3 612782 ORAI1 610277
13q33.1 Immunodeficiency 78 with autoimmunity and developmental delay AR 3 619220 TPP2 190470
14q11.2 Immunodeficiency 7, TCR-alpha/beta deficient AR 3 615387 TRAC 186880
14q11.2 ?Immunodeficiency 108 with autoinflammation AR 3 260570 CEBPE 600749
14q12 Immunodeficiency 115 with autoinflammation AR 3 620632 RNF31 612487
14q12 Immunodeficiency 65, susceptibility to viral infections AR 3 618648 IRF9 147574
14q32.2 Immunodeficiency 49, severe combined AD 3 617237 BCL11B 606558
15q14 Immunodeficiency 64 AR 3 618534 RASGRP1 603962
15q21.1 Immunodeficiency 43 AR 3 241600 B2M 109700
15q21.2 Immunodeficiency 86, mycobacteriosis AR 3 619549 SPPL2A 608238
16p12.1 Immunodeficiency 56 AR 3 615207 IL21R 605383
16p11.2 Immunodeficiency 52 AR 3 617514 LAT 602354
16p11.2 Immunodeficiency 8 AR 3 615401 CORO1A 605000
16q22.1 Immunodeficiency 58 AR 3 618131 CARMIL2 610859
16q24.1 Immunodeficiency 32B, monocyte and dendritic cell deficiency, autosomal recessive AR 3 226990 IRF8 601565
16q24.1 Immunodeficiency 32A, mycobacteriosis, autosomal dominant AD 3 614893 IRF8 601565
17q11.2 ?Immunodeficiency 13 AD 3 615518 UNC119 604011
17q12-q21.1 ?Immunodeficiency 84 AD 3 619437 IKZF3 606221
17q21.31 Immunodeficiency 112 AR 3 620449 MAP3K14 604655
17q21.32 ?Immunodeficiency 88 AR 3 619630 TBX21 604895
18q21.32 Immunodeficiency 12 AR 3 615468 MALT1 604860
19p13.3 Hatipoglu immunodeficiency syndrome AR 3 620331 DPP9 608258
19p13.2 Immunodeficiency 35 AR 3 611521 TYK2 176941
19p13.11 Immunodeficiency 76 AR 3 619164 FCHO1 613437
19p13.11 Immunodeficiency 30 AR 3 614891 IL12RB1 601604
19q13.2 ?Immunodeficiency 62 AR 3 618459 ARHGEF1 601855
19q13.32 ?Immunodeficiency 53 AR 3 617585 RELB 604758
19q13.33 Immunodeficiency 96 AR 3 619774 LIG1 126391
19q13.33 Immunodeficiency 120 3 620836 POLD1 174761
20p11.23 ?Immunodeficiency 101 (varicella zoster virus-specific) AD 3 619872 POLR3F 617455
20p11.21 Immunodeficiency 55 AR 3 617827 GINS1 610608
20q11.23 ?Immunodeficiency 99 with hypogammaglobulinemia and autoimmune cytopenias AR 3 619846 CTNNBL1 611537
20q13.12 T-cell immunodeficiency, recurrent infections, autoimmunity, and cardiac malformations AR 3 614868 STK4 604965
20q13.13 Immunodeficiency 91 and hyperinflammation AR 3 619644 ZNFX1 618931
21q22.11 Immunodeficiency 45 AR 3 616669 IFNAR2 602376
21q22.11 Immunodeficiency 106, susceptibility to viral infections AR 3 619935 IFNAR1 107450
21q22.11 Immunodeficiency 28, mycobacteriosis AR 3 614889 IFNGR2 147569
21q22.3 ?Immunodeficiency 119 3 620825 ICOSLG 605717
21q22.3 Immunodeficiency 114, folate-responsive AR 3 620603 SLC19A1 600424
22q11.1 Immunodeficiency 51 AR 3 613953 IL17RA 605461
22q12.3 ?Immunodeficiency 85 and autoimmunity AD 3 619510 TOM1 604700
22q12.3 Immunodeficiency 63 with lymphoproliferation and autoimmunity AR 3 618495 IL2RB 146710
22q13.1 Immunodeficiency 73B with defective neutrophil chemotaxis and lymphopenia AD 3 618986 RAC2 602049
22q13.1 Immunodeficiency 73A with defective neutrophil chemotaxix and leukocytosis AD 3 608203 RAC2 602049
22q13.1 ?Immunodeficiency 73C with defective neutrophil chemotaxis and hypogammaglobulinemia AR 3 618987 RAC2 602049
22q13.1 ?Immunodeficiency 89 and autoimmunity AR 3 619632 CARD10 607209
22q13.1-q13.2 ?Immunodeficiency 66 AR 3 618847 MKL1 606078
Xp22.2 Immunodeficiency 74, COVID19-related, X-linked XLR 3 301051 TLR7 300365
Xp22.2 Immunodeficiency 98 with autoinflammation, X-linked SMo, XL 3 301078 TLR8 300366
Xp22.12 ?Immunodeficiency 61 XLR 3 300310 SH3KBP1 300374
Xp21.1-p11.4 Immunodeficiency 34, mycobacteriosis, X-linked XLR 3 300645 CYBB 300481
Xp11.23 Wiskott-Aldrich syndrome XLR 3 301000 WAS 300392
Xq12 Immunodeficiency 50 XLR 3 300988 MSN 309845
Xq13.1 Combined immunodeficiency, X-linked, moderate XLR 3 312863 IL2RG 308380
Xq13.1 Severe combined immunodeficiency, X-linked XLR 3 300400 IL2RG 308380
Xq22.1 Agammaglobulinemia, X-linked 1 XLR 3 300755 BTK 300300
Xq24 Immunodeficiency 118, mycobacteriosis XLR 3 301115 MCTS1 300587
Xq25 Lymphoproliferative syndrome, X-linked, 1 XLR 3 308240 SH2D1A 300490
Xq26.1 Immunodeficiency 102 XLR 3 301082 SASH3 300441
Xq26.3 Immunodeficiency, X-linked, with hyper-IgM XLR 3 308230 TNFSF5 300386
Xq28 Immunodeficiency 47 XLR 3 300972 ATP6AP1 300197
Xq28 Immunodeficiency 33 XLR 3 300636 IKBKG 300248

TEXT

A number sign (#) is used with this entry because X-linked immunodeficiency with hyper-IgM type 1 (HIGM1) is caused by mutation in the CD40LG gene (300386) on chromosome Xq26.


Description

HIGM is a rare immunodeficiency characterized by normal or elevated serum IgM levels associated with markedly decreased IgG, IgA, and IgE, resulting in a profound susceptibility to bacterial infections and an increased susceptibility to opportunistic infections. Patients with X-linked HIGM also tend to have neutropenia, as well as a high rate of gastrointestinal and central nervous system infections, often resulting in severe liver disease and/or neurodegeneration (summary by Levy et al., 1997).

Genetic Heterogeneity of Immunodeficiency with Hyper-IgM

Other forms of HIGM include HIGM2 (605258), which results from mutation in the AICDA gene (605257), HIGM3 (606843), which results from mutation in the CD40 gene (109535), and HIGM5 (608106), which results from mutation in the UNG gene (191525). See also HIGM4 (608184).


Clinical Features

The clinical course of X-linked hyper-IgM syndrome is similar to that of X-linked Bruton-type agammaglobulinemia (300755) except for a greater frequency of 'autoimmune' hematologic disorders (neutropenia, hemolytic anemia, thrombocytopenia). Neutropenia may be accompanied by gingivitis, ulcerative stomatitis, fever, and weight loss (Levy et al., 1997).

Jamieson and Kerr (1962) reported a pedigree in which 4 boys were affected. Levitt et al. (1983) reported 4 male patients with recurrent infections. Two of them had agranulocytosis or neutropenia. One had an uncle (presumably maternal) who died in infancy after developing agranulocytosis and Candida sepsis and who showed atrophic lymphoid tissue at autopsy.

Pathologically, lymphoid tissue shows disorganization of the follicular architecture and PAS-positive plasmacytoid cells containing IgM. Lymph nodes lack germinal centers (Ramesh et al., 1999). Tonsillar hypertrophy due to infiltration with these cells may occur. (The tonsils and other lymphoid tissues are atrophic in Bruton agammaglobulinemia.)

Levy et al. (1997) estimated that only 20% of patients will reach the third decade of life and that 75% of these patients will have liver complications. Hayward et al. (1997) described various gastrointestinal cancers, including cholangiocarcinoma, hepatocellular carcinoma, and adenocarcinoma in a cohort of boys with the hyper-IgM syndrome 1 and cholangiopathy. In that study, 70% of the boys who were systematically screened for infection had Cryptosporidium parvum infection (protozoan that causes bowel infection, usually in the setting of immunosuppression or immunodeficiency) and all had clinically significant chronic liver disease.

Cunningham et al. (1999) reported 3 patients with X-linked hyper-IgM syndrome from 2 families who developed enteroviral encephalitis at ages 30 months, 21 months, and 30 months. All presented with central nervous system abnormalities and the 2 surviving patients showed developmental delay. The authors stressed the importance of CSF PCR testing in similar instances.

Aschermann et al. (2007) reported a 19-year-old male patient with X-linked hyper-IgM syndrome, confirmed by genetic analysis, who developed progressive multifocal leukoencephalopathy due to opportunistic infection with the JC virus. He had decreased serum IgA, slightly increased IgM, and normal IgG due to monthly infusions. Despite combined antiviral treatment, he died after 6 weeks. The report indicated that, in addition to immunoglobulin deficiency, patients with this disorder have impaired cellular immune responses due to decreased T cell activation.

Hasegawa et al. (2014) reported a 21-year-old Japanese man, born of unrelated parents, with HIGM1 confirmed by genetic analysis. He presented in infancy with failure to thrive and recurrent otitis media and was treated with immunoglobulin. He showed clumsiness in childhood, and by age 20 years he had developed involuntary movements of the extremities, dysarthria, and hyperactive reflexes. He also had significant cognitive impairment (IQ of 58). Laboratory studies showed low serum IgG and increased serum IgM. No pathogens were detected in the cerebrospinal fluid. Brain imaging showed atrophy of the cerebral cortex and striatum, and EEG showed abnormalities in the absence of clinical seizures. Within 6 months, he was unable to walk due to severe choreoathetosis. Whole-exome sequencing detected a truncating mutation in the CD40LG gene. He also carried an in-frame deletion in the POLG gene (174763) that was not thought not to contribute to the phenotype. The patient was part of a cohort of 9 individuals with neurodegenerative features and hypogammaglobulinemia who underwent whole-exome sequencing. Hasegawa et al. (2014) noted that patients with CD40LG deficiency are susceptible to central nervous system infections, but also suggested that CD40LG may play a role in neuronal function. The report illustrated that whole-exome sequencing can lead to unpredictable molecular diagnoses and unexpected clinical features.

Palterer et al. (2022) reported a 41-year-old man who presented with laryngeal and facial mucocutaneous leishmaniasis. He was also diagnosed with hypogammaglobulinemia. He was treated with amphotericin, miltefosine, and pentamidine and with immunoglobulin replacement. He then developed an extranodal EBV-associated lymphoma of the soft palate which was treated with chemotherapy. A sib, who did not undergo molecular testing, died at 20 years of age of a lymphoproliferative disease, suggesting to Palterer et al. (2022) that he may also have had HIGM1.


Inheritance

HIGM1 is inherited as an X-linked recessive trait. Female carriers manifest normal IgG and IgA production (Hendriks et al., 1990).

Other inheritance patterns have been suggested. Kyong et al. (1978) reported 2 cases in male and female patients and suggested autosomal recessive inheritance. They referred to a case reported by Gleich et al. (1965) in which a female infant had reduced levels of IgG and IgA, elevated IgM, recurrent otitis media, pneumonia, and cervical abscesses. Brahmi et al. (1983) reported father and 2 daughters with the hyper-IgM syndrome. They concluded that the genetics of the hyper-IgM syndrome is 'still unresolved.' Probable autosomal dominant inheritance of one form was suggested. In a review paper, Notarangelo et al. (1992) stated that hyper-IgM syndrome had been shown to be X-linked, autosomal recessive, and autosomal dominant.


Diagnosis

Lin et al. (1996) pointed to PCR-SSCP screening of genomic DNA as a reliable way to establish a diagnosis of hyper-IgM syndrome 1 unequivocally and to identify carriers. Patients with the X-linked form of the disease have the onset of infections in the first few years of life and are more likely to have opportunistic infections and/or neutropenia than are patients with autosomal recessive or multifactorial disease. However, these features are not sufficiently specific to permit a definitive diagnosis of X-linked hyper-IgM syndrome.


Clinical Management

Dunn et al. (1982) found that large doses of fresh plasma corrected the neutropenia. Notarangelo et al. (1992) stated that treatment is mainly based upon regular administration of intravenous immunoglobulins, and that, in addition, steroids may be used in the treatment of neutropenia and severe autoimmune manifestations.

Thomas et al. (1995) performed successful allogeneic bone marrow transplantation in a boy with hyper-IgM syndrome 1 using his carrier sister as the donor. Full engraftment was shown by several means, including changes in red cell antigens, the results of fluorescence in situ hybridization for X and Y chromosomes, polymorphism of the CD40LG gene, and expression of the CD40 ligand by activated T cells. Transplantation was considered indicated because the patient had had P. carinii pneumonitis and came from a family in which 2 maternal uncles had died of protracted diarrhea at the ages of 6 months and 2 years, respectively. A first cousin had the same disorder with persistent diarrhea caused by cryptosporidium and with cholangitis associated with liver cirrhosis.

Hadzic et al. (2000) performed a cadaveric orthotopic liver transplantation together with nonmyeloablative bone marrow transplantation from a matched, unrelated donor in an 18-year-old man with end-stage chronic liver disease associated with the X-linked hyper-IgM syndrome. The removed liver was severely cirrhotic with alternating areas of macronodular hypertrophy and collapse. Fourteen months after liver transplantation and 13 months after bone marrow transplantation, the patient was in excellent health, with satisfactory function of both grafts.

Gennery et al. (2000) reported successful bone marrow transplant in a patient with X-linked hyper-IgM syndrome with a 6/6 antigen matched unrelated donor.


Pathogenesis

It was first thought that the defect in this disorder was within the B cells themselves (see HISTORY section). Levitt et al. (1983) demonstrated that this disorder has a primary dysfunction of B-lymphocyte heavy chain isotype switching from IgM to IgG and IgA. Clinically, however, the recurrence of opportunistic infections (Pneumocystis carinii, toxoplasmosis) suggested anomalies of T-cell function. Moreover, isotype switch obtained in HIGM1 B cells after cocultivation with Sezary syndrome T cells, as well as a random pattern of X-chromosome inactivation in obligatory carriers of HIGM1, argued against a primary B-cell defect (Mayer et al., 1986).

Fuleihan et al. (1993) evaluated isotype switch recombination in 3 affected males by examining interleukin 4-driven IgE synthesis. T-cell-dependent IgE synthesis was completely absent in the B lymphocytes of the patients. CD40 mAb plus interleukin-4 induced the patients' B cells to synthesize IgE and to undergo deletional switch recombination. In contrast, T cells from the patients failed to induce IgE synthesis in interleukin-4-treated B cells and were unable to express the CD40 ligand on their surface. These results suggested that defective expression of the CD40 ligand underlies the failure of isotype switching in HIGM1.

Aruffo et al. (1993) found that patients with HIGM1 express functional CD40 but their T cells do not have functional CD40 ligand (which Aruffo et al. (1993) called gp39) as measured by T-cell binding of CD40-Ig. The patients expressed normal levels of gp39 mRNA, but these RNAs encoded defective gp39 proteins owing to mutations in the extracellular domain of gp39. Soluble recombinant forms of gp39 containing these mutations were unable to bind CD40 and drive normal B-cell proliferation.

Bossaller et al. (2006) found that CD40L-deficient patients, like ICOS (604558)-deficient patients, had abrogated germinal center formation and a severe reduction of CXCR5 (BLR1; 601613)-positive T cells.

Using flow cytometric analysis, van Zelm et al. (2014) found reduced numbers of all memory B-cell subsets except CD27 (TNFRSF7; 186711)-negative/IgA-positive B cells in both CD19 (107265)-deficient patients and CD40L-deficient patients compared with controls. Analysis of transcripts after class switching demonstrated that patient transcripts had fewer somatic mutations and reduced usage of IgG2 and IgA2 subclasses. There was also a deficiency in selection strength of mutations for antigen binding in patients compared with controls, whereas selection to maintain superantigen binding was normal. Selection against the autoreactive properties of immunoglobulins was impaired in patients. Somatic hypermutation analysis revealed decreased AICDA and UNG activity in CD40L deficiency, but increased UNG activity and decreased mismatch repair in CD19 deficiency. Van Zelm et al. (2014) concluded that both the B-cell antigen receptor and CD40 signaling pathways are required for selection of immunoglobulin reactivity, but that they differentially mediate DNA repair pathways during somatic hypermutation and thereby together shape the mature B-cell repertoire.

X-Inactivation Studies

If the defect in the switch mechanism is intrinsic to the B cells, a skewed X chromosome inactivation pattern would be observed in IgG- and IgA-expressing B lymphocytes of female carriers. Hendriks et al. (1990) studied lymphoblastoid B cells from 2 female carriers (see HISTORY section). Hendriks et al. (1990) concluded that the HIGM1 gene encodes a class switch inducer that is transferred to B lymphocytes from a cell of synthesis, possibly T lymphocytes.

Contrary to the findings of Hendriks et al. (1990) and those of Conley et al. (1988), Notarangelo et al. (1991) found nonrandom X-chromosome inactivation in T cells, B cells, and neutrophils, but not in fibroblasts, of obligate carriers, suggesting that several different hematopoietic cell lineages are primarily involved in HIGM1. Preferential inactivation of the paternally derived X chromosome was demonstrated by analysis of segregation of the alleles defined by 2 DNA probes. Notarangelo et al. (1991) suggested that the HIGM1 mutation may confer an advantage in differentiation and/or proliferation to hematopoietic precursors carrying the mutant allele on the active X chromosome.

In studies of X-chromosome inactivation in carriers of HIGM1, Hollenbaugh et al. (1994) found that the CD40L gene is not selectively inactivated. Furthermore, even when there was extremely skewed inactivation, normal levels of serum immunoglobulins were found. Unlike some other X-linked defects in which extreme lyonization may lead to disease, a small population of cells expressing the wildtype protein was sufficient to maintain normal humoral immunity and prevent the clinical symptoms of the disorder. Kipps (1994) commented that the findings have encouraging implications for patients with the disorder, since it seems that only a relatively small fraction of the T cells need express a functional CD40-ligand for effective immunity. Even a partial reconstitution with precursor T cells capable of expressing a functional ligand might suffice.


Mapping

Mensink et al. (1987) concluded that the locus for immunodeficiency with increased IgM (symbolized XHM by them) is linked to the DXS42 RFLP locus, which maps to Xq24-q27. Recombination between XHM and DXS17 was observed, whereas no recombination between XLA and DXS17 has been found; thus, XHM and XLA are apparently determined by separate gene loci. Padayachee et al. (1992, 1993) narrowed the location to Xq26 by multipoint linkage studies demonstrating that it is close to HPRT (308000), a gene that forms part of an extensive YAC contig mapping to Xq26; a maximum lod score of 4.89 was obtained. The existence of an easily scorable VNTR of 5 alleles within the HPRT gene means that other families with X-linked hyper-IgM syndrome are likely to be informative for this polymorphism.

Aruffo et al. (1993) mapped the GP39 gene to Xq26 by PCR analysis of a regional mapping panel, followed up by fluorescence in situ hybridization for precise localization. By YAC analysis, Pilia et al. (1994) mapped the CD40L locus between DXS144E and DXS300 in Xq26 and determined its transcription to be from 5-prime centromeric to 3-prime telomeric. This corresponded to the site where the clinical phenotype of the hyper-IgM syndrome had been mapped.

Allen et al. (1993) mapped the CD40LG gene (300386) to the proximal region of the mouse X chromosome, linked to Hprt. Hprt maps to the Xq26-q27.2 region, which suggested that the human CD40LG gene would also map to this region. This was confirmed by fluorescence in situ hybridization studies of CD40LG by Graf et al. (1992) and Allen et al. (1993).


Molecular Genetics

Allen et al. (1993) presented conclusive evidence that the defect in X-linked hyper-IgM syndrome resides in the gene for the CD40 ligand (300386). Because CD40LG induces B-cell proliferation in the absence of any costimulus and because the hyper-IgM phenotype and the CD40LG gene map to the same location, CD40LG was suggested as the site of the mutation in HIGM1. Allen et al. (1993) demonstrated this to be case by the finding of point mutations in 3 of 4 patients with the syndrome (300386.0003-300386.0005). Similarly, Aruffo et al. (1993) identified mutations in the CD40LG gene in patients with the syndrome (300386.0001-300386.0002).

In a 41-year-old man with HIGM1, Palterer et al. (2022) identified a heterozygous missense mutation (M36K; 300386.0015) in the transmembrane domain of the CD40LG gene. The expression of CD40L was reduced on activated T CD4+ cells from the patient. The patient had an atypical clinical presentation that included leishmaniasis and hypogammaglobulinemia. Palterer et al. (2022) concluded that variants in the transmembrane domain of CD40LG act as hypomorphic variants and could lead to atypical clinical features.


Animal Model

Rosen (1975) stated that 'a similar syndrome of X-linked immunodeficiency with increased IgM has been found in mice.' Xu et al. (1994) generated CD40LG-deficient mice by targeted disruption.

Using intravital microscopy and histologic examination of arterioles in mice lacking CD40L, Andre et al. (2002) observed frequent rupture and embolization of thrombi. The thrombi showed lower platelet density compared to those of wildtype mice, which contributed to platelet fragility. Administration of recombinant soluble CD40L (rsCD40L), but not rsCD40L with a mutation changing the KGD motif sequence to KGE, restored thrombus stability even in CD40 -/- mice, indicating that CD40L is not acting through CD40 ligation. Evaluation of hemostasis suggested that the thrombus instability in CD40L -/- mice is not due a lack of fibrin formation but rather a defect in platelet-platelet interaction which could be stabilized by the wildtype, but not by the mutant, rsCD40L. Flow cytometric analysis demonstrated that rsCD40L binds to activated platelets of wildtype as well as of CD40 -/- mice, but that this binding can be inhibited by a peptide interfering with ITGA2B (607759)/ITGB3 (173470) binding. Plate-binding analysis indicated specific saturable binding of rsCD40L to ITGA2B/ITGB3. Fluorescence microscopy showed that human platelets spread on but did not adhere to an rsCD40L-coated glass surface only in the absence of an inhibitor of ITGA2B/ITGB3 binding. Andre et al. (2002) concluded that CD40L is an ITGA2B/ITGB3 ligand, a platelet agonist, and necessary for the stability of arterial thrombi. They also noted that these findings suggest the careful evaluation of clinical trials with anti-CD40L therapy.


History

Ramesh et al. (1999) reviewed the history as well as other aspects of the hyper-IgM syndrome, which they abbreviated HIM. In the WHO classification of immunodeficiencies, an entity termed X-linked immunodeficiency with increased IgM was listed (Fudenberg et al., 1970). A definition of the disorder was an outcome of an international workshop (Cooper et al., 1974). It was originally hypothesized that B lymphocytes from patients with HIGM1 have an intrinsic inability to undergo immunoglobulin isotype switch.

Levitt et al. (1983) suggested that this disorder has a primary dysfunction of B-lymphocyte isotype switching. In 4 male patients with hyper-IgM immunodeficiency, the number, proportion, and proliferation of T lymphocytes were shown to be normal. IgG and IgA B lymphocytes were completely absent. In vitro stimulation of patients' B cells with both T cell-dependent and T cell-independent activators failed to induce any IgG or IgA-producing B cells. They concluded that individuals with this disorder possess an intrinsic B cell dysfunction that is not related to abnormal T cell regulation.

The observation that patients with HIM and particularly those with the X-linked form of the disorder (XHIM) were prone to opportunistic infections suggested a T-cell defect in spite of laboratory evidence for a humoral immune deficiency. The hypothesis of a primary T-cell defect was elegantly supported by the studies of Mayer et al. (1986), who demonstrated that B cells from XHIM patients could be driven to secrete immunoglobulins of various isotypes in the presence of pokeweed mitogen when cocultivated with 'helper T lymphoblasts' from a patient with a Sezary-like syndrome, a neoplastic disorder of T cells.

Hendriks et al. (1990) studied lymphoblastoid B cells from 2 female carriers who had IgG- and IgA-expressing B cells, in order to determine if the defect in the switch mechanism is intrinsic to the B cells. In an analysis of differential methylation of the polymorphic DXS255 locus, random X chromosome inactivation patterns were found in populations of T lymphocytes, in IgM-expressing B lymphocytes, and in IgG- or IgA-expressing B lymphocytes. Similar extent of heterogeneity was found for Ig H chain rearrangements and the Ig light chain usage in the IgA- or IgG-expressing B cell that had inactivated the X chromosome which carried the intact gene and in clones with the mutant-bearing X chromosome inactivated. The results indicated that the intrinsic Ig H chain class switch mechanism in this disorder is fully intact in B lymphocytes. When B lymphocytes with the X chromosome bearing the mutant IMD3 gene on the active X chromosome are placed in an in vivo environment that contains the intact gene product, i.e., in the female heterozygotes, the switch from IgM to IgG or IgA production occurs at normal frequencies. These findings are consistent with the observation that switch of hyper-IgM B cells is induced by Sezary-like T lymphoblasts (Mayer et al., 1986).


REFERENCES

  1. Allen, R. C., Armitage, R. J., Conley, M. E., Rosenblatt, H., Jenkins, N. A., Copeland, N. G., Bedell, M. A., Edelhoff, S., Disteche, C. M., Simoneaux, D. K., Fanslow, W. C., Belmont, J., Spriggs, M. K. CD40 ligand gene defects responsible for X-linked hyper-IgM syndrome. Science 259: 990-993, 1993. [PubMed: 7679801, related citations] [Full Text]

  2. Andre, P., Prasad, K. S. S., Denis, C. V., He, M., Papalia, J. M., Hynes, R. O., Phillips, D. R., Wagner, D. D. CD40L stabilizes arterial thrombi by a beta(3) integrin-dependent mechanism. Nature Med. 8: 247-252, 2002. [PubMed: 11875495, related citations] [Full Text]

  3. Aruffo, A., Farrington, M., Hollenbaugh, D., Li, X., Milatovich, A., Nonoyama, S., Bajorath, J., Grosmaire, L. S., Stenkamp, R., Neubauer, M., Roberts, R. L., Noelle, R. J., Ledbetter, J. A., Francke, U., Ochs, H. D. The CD40 ligand, gp39, is defective in activated T cells from patients with X-linked hyper-IgM syndrome. Cell 72: 291-300, 1993. [PubMed: 7678782, related citations] [Full Text]

  4. Aschermann, Z., Gomori, E., Kovacs, G. G., Pal, E., Simon, G., Komoly, S., Marodi, L., Illes, Z. X-linked hyper-IgM syndrome associated with a rapid course of multifocal leukoencephalopathy. Arch. Neurol. 64: 273-276, 2007. [PubMed: 17296845, related citations] [Full Text]

  5. Bossaller, L., Burger, J., Draeger, R., Grimbacher, B., Knoth, R., Plebani, A., Durandy, A., Baumann, U., Schlesier, M., Welcher, A. A., Peter, H. H., Warnatz, K. ICOS deficiency is associated with a severe reduction of CXCR5+CD4 germinal center Th cells. J. Immun. 177: 4927-4932, 2006. [PubMed: 16982935, related citations] [Full Text]

  6. Brahmi, Z., Lazarus, K. H., Hodes, M. E., Baehner, R. L. Immunologic studies of three family members with the immunodeficiency with hyper-IgM syndrome. J. Clin. Immun. 3: 127-134, 1983. [PubMed: 6602145, related citations] [Full Text]

  7. Conley, M. E., Brown, P., Pahwa, S., Puck, J. M. An intrinsic B cell defect in X-linked hyper IgM syndrome. (Abstract) Pediat. Res. 23: 353A, 1988.

  8. Cooper, M. D., Faulk, W. P., Fudenberg, H. H., Good, R. A., Hitzig, W., Kunkel, H. G., Roitt, I. M., Rosen, F. S., Seligmann, M., Soothill, J. F. Meeting report of the second international workshop on primary immunodeficiency diseases in man. Clin. Immun. Immunopath. 2: 416-445, 1974. [PubMed: 4596971, related citations] [Full Text]

  9. Cunningham, C. K., Bonville, C. A., Ochs, H. D., Seyama, K., John, P. A., Rotbart, H. A., Weiner, L. B. Enteroviral meningoencephalitis as a complication of X-linked hyper IgM syndrome. J. Pediat. 134: 584-588, 1999. [PubMed: 10228294, related citations] [Full Text]

  10. Dunn, K., Lubens, R., Stiehm, E. R. Reversal of neutropenia in X-linked immunodeficiency with hyper-IgM by large doses of plasma. (Abstract) Clin. Res. 30: 125A, 1982.

  11. Fudenberg, H. H., Good, R. A., Hitzig, W., Kunkel, H. G., Roitt, I. M., Rosen, F. S., Rowe, D. S., Seligmann, M., Soothill, J. R. Classification of the primary immunodeficiencies (WHO recommendation). New Eng. J. Med. 283: 656-657, 1970. [PubMed: 5456822, related citations] [Full Text]

  12. Fuleihan, R., Ramesh, N., Loh, R., Jabara, H., Rosen, F. S., Chatila, T., Fu, S. M., Stamenkovic, I., Geha, R. S. Defective expression of the CD40 ligand in X chromosome-linked immunoglobulin deficiency with normal or elevated IgM. Proc. Nat. Acad. Sci. 90: 2170-2173, 1993. [PubMed: 7681587, related citations] [Full Text]

  13. Gennery, A. R., Clark, J. E., Flood, T. J., Abinun, M., Cant, A. J. T-cell-depleted bone marrow transplantation from allogeneic sibling for X-linked hyperimmunoglobulin M syndrome. (Letter) J. Pediat. 137: 290 only, 2000. Note: Erratum: J. Pediat. 137: 592 only, 2000. [PubMed: 10931436, related citations] [Full Text]

  14. Gleich, G. J., Condemi, J. J., Vaughan, J. H. Dysgammaglobulinemia in the presence of plasma cells. New Eng. J. Med. 272: 331-340, 1965. [PubMed: 14239114, related citations] [Full Text]

  15. Graf, D., Korthauer, U., Mages, H. W., Senger, G., Kroczek, R. A. Cloning of TRAP, a ligand for CD40 on human T cells. Europ. J. Immun. 22: 3191-3194, 1992. [PubMed: 1280226, related citations] [Full Text]

  16. Hadzic, N., Pagliuca, A., Rela, M., Portmann, B., Jones, A., Veys, P., Heaton, N. D., Mufti, G. J., Mieli-Vergani, G. Correction of the hyper-IgM syndrome after liver and bone marrow transplantation. New Eng. J. Med. 342: 320-324, 2000. [PubMed: 10655530, related citations] [Full Text]

  17. Hasegawa, S., Imai, K., Yoshida, K., Okuno, Y., Muramatsu, H., Shiraishi, Y., Chiba, K., Tanaka, H., Miyano, S., Kojima, S., Ogawa, S., Morio, T., Mizutani, S., Takagi, M. Whole-exome sequence analysis of ataxia telangiectasia-like phenotype. J. Neurol. Sci. 340: 86-90, 2014. [PubMed: 24631270, related citations] [Full Text]

  18. Hayward, A. R., Levy, J., Facchetti, F., Notarangelo, L., Ochs, H. D., Etzioni, A., Bonnefoy, J. Y., Cosyns, M., Weinberg, A. Cholangiopathy and tumors of the pancreas, liver, and biliary tree in boys with X-linked immunodeficiency with hyper-IgM. J. Immun. 158: 977-983, 1997. [PubMed: 8993019, related citations]

  19. Hendriks, R. W., Kraakman, M. E. M., Craig, I. W., Espanol, T., Schuurman, R. K. B. Evidence that in X-linked immunodeficiency with hyperimmunoglobulinemia M the intrinsic immunoglobulin heavy chain class switch mechanism is intact. Europ. J. Immun. 20: 2603-2608, 1990. [PubMed: 1980111, related citations] [Full Text]

  20. Hollenbaugh, D., Wu, L. H., Ochs, H. D., Nonoyama, S., Grosmaire, L. S., Ledbetter, J. A., Noelle, R. J., Hill, H., Aruffo, A. The random inactivation of the X chromosome carrying the defective gene responsible for X-linked hyper IgM syndrome (X-HIM) in female carriers of HIGM1. J. Clin. Invest. 94: 616-622, 1994. [PubMed: 7518839, related citations] [Full Text]

  21. Jamieson, W. M., Kerr, M. R. A family with several cases of hypogammaglobulinemia. Arch. Dis. Child. 37: 330-336, 1962. [PubMed: 14451053, related citations] [Full Text]

  22. Kipps, T. J. X inactivation and immunocompetence in female carriers of the X-linked hyper-IgM syndrome (Editorial) J. Clin. Invest. 94: 469, 1994. [PubMed: 8040297, related citations] [Full Text]

  23. Kyong, C. U., Virella, G., Fundenberg, H. H., Darby, C. P. X-linked immunodeficiency with increased IgM: clinical, ethnic, and immunologic heterogeneity. Pediat. Res. 12: 1024-1026, 1978. [PubMed: 724295, related citations] [Full Text]

  24. Levitt, D., Haber, P., Rich, K., Cooper, M. D. Hyper IgM immunodeficiency: a primary dysfunction of B lymphocyte isotype switching. J. Clin. Invest. 72: 1650-1657, 1983. [PubMed: 6605368, related citations] [Full Text]

  25. Levy, J., Espanol-Boren, T., Thomas, C., Fischer, A., Tovo, P., Bordigoni, P., Resnick, I., Fasth, A., Baer, M., Gomez, L., Sanders, E. A. M., Tabone, M.-D., and 13 others. Clinical spectrum of X-linked hyper-IgM syndrome. J. Pediat. 131: 47-54, 1997. [PubMed: 9255191, related citations] [Full Text]

  26. Lin, Q., Rohrer, J., Allen, R. C., Larche, M., Greene, J. M., Shigeoka, A. O., Gatti, R. A., Derauf, D. C., Belmont, J. W., Conley, M. E. A single strand conformation polymorphism study of CD40 ligand: efficient mutation analysis and carrier detection of X-linked hyper IgM syndrome. J. Clin. Invest. 97: 196-201, 1996. [PubMed: 8550833, related citations] [Full Text]

  27. Mayer, L., Kwan, S. P., Thompson, C., Ko, H. S., Chiorazzi, N., Waldmann, T., Rosen, F. Evidence for a defect in 'switch' T cells in patients with immunodeficiency and hyperimmunoglobulinemia M. New Eng. J. Med. 314: 409-413, 1986. [PubMed: 3080678, related citations] [Full Text]

  28. Mensink, E. J. B. M., Thompson, A., Sandkuyl, L. A., Kraakman, M. E. M., Schot, J. D. L., Espanol, T., Schuurman, R. K. B. X-linked immunodeficiency with hyperimmunoglobulinemia M appears to be linked to the DXS42 restriction fragment length polymorphism locus. Hum. Genet. 76: 96-99, 1987. [PubMed: 2883112, related citations] [Full Text]

  29. Notarangelo, L. D., Duse, M., Ugazio, A. G. Immunodeficiency with hyper-IgM (HIM). Immunodef. Rev. 3: 101-122, 1992. [PubMed: 1554497, related citations]

  30. Notarangelo, L. D., Parolini, O., Albertini, A., Duse, M., Mazzolari, E., Plebani, A., Camerino, G., Ugazio, A. G. Analysis of X-chromosome inactivation in X-linked immunodeficiency with hyper-IgM (HIGM1): evidence for involvement of different hematopoietic cell lineages. Hum. Genet. 88: 130-134, 1991. [PubMed: 1757090, related citations] [Full Text]

  31. Padayachee, M., Feighery, C., Finn, A., McKeown, C., Levinsky, R. J., Kinnon, C., Malcolm, S. Mapping of the X-linked form of hyper-IgM syndrome (HIGM1) to Xq26 by close linkage to HPRT. Genomics 14: 551-553, 1992. [PubMed: 1427881, related citations] [Full Text]

  32. Padayachee, M., Levinsky, R. J., Kinnon, C., Finn, A., McKeown, C., Feighery, C., Notarangelo, L. D., Hendriks, R. W., Read, A. P., Malcolm, S. Mapping of the X linked form of hyper IgM syndrome (HIGM1). J. Med. Genet. 30: 202-205, 1993. Note: Erratum: J. Med. Genet. 30: 528 only, 1993. [PubMed: 8097258, related citations] [Full Text]

  33. Palterer, B., Salvati, L., Capone, M., Mecheri, V., Maggi, L., Mazzoni, A., Cosmi, L., Volpi, N., Tiberi, L., Provenzano, A., Giglio, S., Parronchi, P., Maggiore, G., Gallo, O., Bartoloni, A., Annunziato, F., Zammarchi, L., Liotta, F. Variant disrupting CD40L transmembrane domain and atypical X-linked hyper-IgM syndrome: a case report with leishmaniasis and review of the literature. Front. Immun. 13: 840767, 2022. [PubMed: 35572607, images, related citations] [Full Text]

  34. Pilia, G., Porta, B., Padayachee, M., Malcolm, S., Zucchi, I., Villa, A., Macchi, P., Vezzoni, P., Schlessinger, D. Human CD40L gene maps between DXS144E and DXS300 in Xq26. Genomics 22: 249-251, 1994. [PubMed: 7959785, related citations] [Full Text]

  35. Ramesh, N., Geha, R. S., Notarangelo, L. D. CD40 ligand and the hyper-IgM syndrome. In: Ochs, H. D.; Smith, C. I. E.; Puck, J. M. (eds.): Primary Immunodeficiency Diseases: A Molecular and Genetic Approach. New York: Oxford University Press 1999. Pp. 233-249.

  36. Rosen, F. S. Immunodeficiency. In: Benacerraf, B. (ed.): Immunogenetics and Immunodeficiency. London: William Clowes & Sons, Ltd. 1975. Pp. 229-257.

  37. Thomas, C., de Saint Basile, G., Le Deist, F., Theophile, D., Benkerrou, M., Haddad, E., Blanche, S., Fischer, A. Brief report: correction of X-linked hyper-IgM syndrome by allogenic bone marrow transplantation. New Eng. J. Med. 333: 426-429, 1995. [PubMed: 7542361, related citations] [Full Text]

  38. van Zelm, M. C., Bartol, S. J. W., Driessen, G. J., Mascart, F., Reisli, I., Franco, J. L., Wolska-Kusnierz, B., Kanegane, H., Boon, L., van Dongen, J. J. M., van der Burg, M. Human CD19 and CD40L deficiencies impair antibody selection and differentially affect somatic hypermutation. J. Allergy Clin. Immun. 134: 135-144, 2014. [PubMed: 24418477, related citations] [Full Text]

  39. Xu, J., Foy, T. M., Laman, J. D., Elliott, E. A., Dunn, J. J., Waldschmidt, T. J., Elsemore, J., Noelle, R. J., Flavell, R. A. Mice deficient for the CD40 ligand. Immunity 1: 423-431, 1994. Note: Erratum: Immunity: 1: 613 only, 1994. [PubMed: 7882172, related citations] [Full Text]


Hilary J. Vernon - updated : 07/07/2022
Paul J. Converse - updated : 11/6/2015
Cassandra L. Kniffin - updated : 9/22/2014
Cassandra L. Kniffin - updated : 9/28/2007
Paul J. Converse - updated : 3/8/2007
Cassandra L. Kniffin - updated : 8/23/2004
Victor A. McKusick - updated : 10/20/2003
Cassandra L. Kniffin - updated : 4/15/2002
Cassandra L. Kniffin - reorganized : 4/5/2002
Paul J. Converse - updated : 2/28/2002
Victor A. McKusick - updated : 2/14/2002
Ada Hamosh - updated : 2/28/2000
Ada Hamosh - reorganized : 2/11/2000
Victor A. McKusick - updated : 2/7/2000
Victor A. McKusick - updated : 9/23/1999
Victor A. McKusick - updated : 11/12/1998
Ada Hamosh - updated : 7/10/1997
Creation Date:
Victor A. McKusick : 6/4/1986
alopez : 08/18/2023
alopez : 08/18/2023
carol : 07/07/2022
carol : 06/01/2017
carol : 06/24/2016
mgross : 11/6/2015
carol : 9/22/2014
ckniffin : 9/22/2014
terry : 4/4/2013
carol : 3/26/2012
wwang : 6/16/2011
wwang : 10/4/2007
ckniffin : 9/28/2007
mgross : 3/8/2007
wwang : 10/27/2005
tkritzer : 8/26/2004
ckniffin : 8/23/2004
tkritzer : 10/21/2003
terry : 10/20/2003
mgross : 10/1/2003
mgross : 9/23/2003
mgross : 9/22/2003
ckniffin : 5/15/2003
terry : 6/27/2002
carol : 4/16/2002
carol : 4/15/2002
ckniffin : 4/15/2002
ckniffin : 4/12/2002
carol : 4/12/2002
ckniffin : 4/12/2002
ckniffin : 4/5/2002
carol : 4/5/2002
ckniffin : 4/5/2002
alopez : 2/28/2002
cwells : 2/21/2002
terry : 2/14/2002
carol : 9/10/2001
terry : 1/18/2001
mgross : 9/19/2000
mcapotos : 8/9/2000
alopez : 2/28/2000
carol : 2/14/2000
carol : 2/11/2000
carol : 2/11/2000
mcapotos : 2/11/2000
terry : 2/7/2000
alopez : 11/15/1999
carol : 10/13/1999
mgross : 10/8/1999
terry : 9/23/1999
carol : 12/14/1998
carol : 11/18/1998
terry : 11/12/1998
dkim : 9/11/1998
dkim : 9/10/1998
dkim : 7/21/1998
alopez : 5/21/1998
mark : 9/2/1997
mark : 9/1/1997
alopez : 7/29/1997
alopez : 7/10/1997
alopez : 7/10/1997
alopez : 6/11/1997
mark : 11/13/1996
terry : 11/7/1996
terry : 11/7/1996
terry : 10/31/1996
mark : 2/2/1996
terry : 2/1/1996
mark : 12/5/1995
mark : 10/5/1995
terry : 9/13/1995
carol : 10/11/1994
jason : 6/28/1994
davew : 6/6/1994
warfield : 4/20/1994

# 308230

IMMUNODEFICIENCY WITH HYPER-IgM, TYPE 1; HIGM1


Alternative titles; symbols

HYPER-IgM IMMUNODEFICIENCY, X-LINKED; XHIM
HYPER-IgM SYNDROME 1
HYPER-IgM SYNDROME; HIGM; IHIS
IMMUNODEFICIENCY 3; IMD3


SNOMEDCT: 403835002;   ORPHA: 101088, 183663;   DO: 0060022, 6620;  


Phenotype-Gene Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Gene/Locus Gene/Locus
MIM number
Xq26.3 Immunodeficiency, X-linked, with hyper-IgM 308230 X-linked recessive 3 TNFSF5 300386

TEXT

A number sign (#) is used with this entry because X-linked immunodeficiency with hyper-IgM type 1 (HIGM1) is caused by mutation in the CD40LG gene (300386) on chromosome Xq26.


Description

HIGM is a rare immunodeficiency characterized by normal or elevated serum IgM levels associated with markedly decreased IgG, IgA, and IgE, resulting in a profound susceptibility to bacterial infections and an increased susceptibility to opportunistic infections. Patients with X-linked HIGM also tend to have neutropenia, as well as a high rate of gastrointestinal and central nervous system infections, often resulting in severe liver disease and/or neurodegeneration (summary by Levy et al., 1997).

Genetic Heterogeneity of Immunodeficiency with Hyper-IgM

Other forms of HIGM include HIGM2 (605258), which results from mutation in the AICDA gene (605257), HIGM3 (606843), which results from mutation in the CD40 gene (109535), and HIGM5 (608106), which results from mutation in the UNG gene (191525). See also HIGM4 (608184).


Clinical Features

The clinical course of X-linked hyper-IgM syndrome is similar to that of X-linked Bruton-type agammaglobulinemia (300755) except for a greater frequency of 'autoimmune' hematologic disorders (neutropenia, hemolytic anemia, thrombocytopenia). Neutropenia may be accompanied by gingivitis, ulcerative stomatitis, fever, and weight loss (Levy et al., 1997).

Jamieson and Kerr (1962) reported a pedigree in which 4 boys were affected. Levitt et al. (1983) reported 4 male patients with recurrent infections. Two of them had agranulocytosis or neutropenia. One had an uncle (presumably maternal) who died in infancy after developing agranulocytosis and Candida sepsis and who showed atrophic lymphoid tissue at autopsy.

Pathologically, lymphoid tissue shows disorganization of the follicular architecture and PAS-positive plasmacytoid cells containing IgM. Lymph nodes lack germinal centers (Ramesh et al., 1999). Tonsillar hypertrophy due to infiltration with these cells may occur. (The tonsils and other lymphoid tissues are atrophic in Bruton agammaglobulinemia.)

Levy et al. (1997) estimated that only 20% of patients will reach the third decade of life and that 75% of these patients will have liver complications. Hayward et al. (1997) described various gastrointestinal cancers, including cholangiocarcinoma, hepatocellular carcinoma, and adenocarcinoma in a cohort of boys with the hyper-IgM syndrome 1 and cholangiopathy. In that study, 70% of the boys who were systematically screened for infection had Cryptosporidium parvum infection (protozoan that causes bowel infection, usually in the setting of immunosuppression or immunodeficiency) and all had clinically significant chronic liver disease.

Cunningham et al. (1999) reported 3 patients with X-linked hyper-IgM syndrome from 2 families who developed enteroviral encephalitis at ages 30 months, 21 months, and 30 months. All presented with central nervous system abnormalities and the 2 surviving patients showed developmental delay. The authors stressed the importance of CSF PCR testing in similar instances.

Aschermann et al. (2007) reported a 19-year-old male patient with X-linked hyper-IgM syndrome, confirmed by genetic analysis, who developed progressive multifocal leukoencephalopathy due to opportunistic infection with the JC virus. He had decreased serum IgA, slightly increased IgM, and normal IgG due to monthly infusions. Despite combined antiviral treatment, he died after 6 weeks. The report indicated that, in addition to immunoglobulin deficiency, patients with this disorder have impaired cellular immune responses due to decreased T cell activation.

Hasegawa et al. (2014) reported a 21-year-old Japanese man, born of unrelated parents, with HIGM1 confirmed by genetic analysis. He presented in infancy with failure to thrive and recurrent otitis media and was treated with immunoglobulin. He showed clumsiness in childhood, and by age 20 years he had developed involuntary movements of the extremities, dysarthria, and hyperactive reflexes. He also had significant cognitive impairment (IQ of 58). Laboratory studies showed low serum IgG and increased serum IgM. No pathogens were detected in the cerebrospinal fluid. Brain imaging showed atrophy of the cerebral cortex and striatum, and EEG showed abnormalities in the absence of clinical seizures. Within 6 months, he was unable to walk due to severe choreoathetosis. Whole-exome sequencing detected a truncating mutation in the CD40LG gene. He also carried an in-frame deletion in the POLG gene (174763) that was not thought not to contribute to the phenotype. The patient was part of a cohort of 9 individuals with neurodegenerative features and hypogammaglobulinemia who underwent whole-exome sequencing. Hasegawa et al. (2014) noted that patients with CD40LG deficiency are susceptible to central nervous system infections, but also suggested that CD40LG may play a role in neuronal function. The report illustrated that whole-exome sequencing can lead to unpredictable molecular diagnoses and unexpected clinical features.

Palterer et al. (2022) reported a 41-year-old man who presented with laryngeal and facial mucocutaneous leishmaniasis. He was also diagnosed with hypogammaglobulinemia. He was treated with amphotericin, miltefosine, and pentamidine and with immunoglobulin replacement. He then developed an extranodal EBV-associated lymphoma of the soft palate which was treated with chemotherapy. A sib, who did not undergo molecular testing, died at 20 years of age of a lymphoproliferative disease, suggesting to Palterer et al. (2022) that he may also have had HIGM1.


Inheritance

HIGM1 is inherited as an X-linked recessive trait. Female carriers manifest normal IgG and IgA production (Hendriks et al., 1990).

Other inheritance patterns have been suggested. Kyong et al. (1978) reported 2 cases in male and female patients and suggested autosomal recessive inheritance. They referred to a case reported by Gleich et al. (1965) in which a female infant had reduced levels of IgG and IgA, elevated IgM, recurrent otitis media, pneumonia, and cervical abscesses. Brahmi et al. (1983) reported father and 2 daughters with the hyper-IgM syndrome. They concluded that the genetics of the hyper-IgM syndrome is 'still unresolved.' Probable autosomal dominant inheritance of one form was suggested. In a review paper, Notarangelo et al. (1992) stated that hyper-IgM syndrome had been shown to be X-linked, autosomal recessive, and autosomal dominant.


Diagnosis

Lin et al. (1996) pointed to PCR-SSCP screening of genomic DNA as a reliable way to establish a diagnosis of hyper-IgM syndrome 1 unequivocally and to identify carriers. Patients with the X-linked form of the disease have the onset of infections in the first few years of life and are more likely to have opportunistic infections and/or neutropenia than are patients with autosomal recessive or multifactorial disease. However, these features are not sufficiently specific to permit a definitive diagnosis of X-linked hyper-IgM syndrome.


Clinical Management

Dunn et al. (1982) found that large doses of fresh plasma corrected the neutropenia. Notarangelo et al. (1992) stated that treatment is mainly based upon regular administration of intravenous immunoglobulins, and that, in addition, steroids may be used in the treatment of neutropenia and severe autoimmune manifestations.

Thomas et al. (1995) performed successful allogeneic bone marrow transplantation in a boy with hyper-IgM syndrome 1 using his carrier sister as the donor. Full engraftment was shown by several means, including changes in red cell antigens, the results of fluorescence in situ hybridization for X and Y chromosomes, polymorphism of the CD40LG gene, and expression of the CD40 ligand by activated T cells. Transplantation was considered indicated because the patient had had P. carinii pneumonitis and came from a family in which 2 maternal uncles had died of protracted diarrhea at the ages of 6 months and 2 years, respectively. A first cousin had the same disorder with persistent diarrhea caused by cryptosporidium and with cholangitis associated with liver cirrhosis.

Hadzic et al. (2000) performed a cadaveric orthotopic liver transplantation together with nonmyeloablative bone marrow transplantation from a matched, unrelated donor in an 18-year-old man with end-stage chronic liver disease associated with the X-linked hyper-IgM syndrome. The removed liver was severely cirrhotic with alternating areas of macronodular hypertrophy and collapse. Fourteen months after liver transplantation and 13 months after bone marrow transplantation, the patient was in excellent health, with satisfactory function of both grafts.

Gennery et al. (2000) reported successful bone marrow transplant in a patient with X-linked hyper-IgM syndrome with a 6/6 antigen matched unrelated donor.


Pathogenesis

It was first thought that the defect in this disorder was within the B cells themselves (see HISTORY section). Levitt et al. (1983) demonstrated that this disorder has a primary dysfunction of B-lymphocyte heavy chain isotype switching from IgM to IgG and IgA. Clinically, however, the recurrence of opportunistic infections (Pneumocystis carinii, toxoplasmosis) suggested anomalies of T-cell function. Moreover, isotype switch obtained in HIGM1 B cells after cocultivation with Sezary syndrome T cells, as well as a random pattern of X-chromosome inactivation in obligatory carriers of HIGM1, argued against a primary B-cell defect (Mayer et al., 1986).

Fuleihan et al. (1993) evaluated isotype switch recombination in 3 affected males by examining interleukin 4-driven IgE synthesis. T-cell-dependent IgE synthesis was completely absent in the B lymphocytes of the patients. CD40 mAb plus interleukin-4 induced the patients' B cells to synthesize IgE and to undergo deletional switch recombination. In contrast, T cells from the patients failed to induce IgE synthesis in interleukin-4-treated B cells and were unable to express the CD40 ligand on their surface. These results suggested that defective expression of the CD40 ligand underlies the failure of isotype switching in HIGM1.

Aruffo et al. (1993) found that patients with HIGM1 express functional CD40 but their T cells do not have functional CD40 ligand (which Aruffo et al. (1993) called gp39) as measured by T-cell binding of CD40-Ig. The patients expressed normal levels of gp39 mRNA, but these RNAs encoded defective gp39 proteins owing to mutations in the extracellular domain of gp39. Soluble recombinant forms of gp39 containing these mutations were unable to bind CD40 and drive normal B-cell proliferation.

Bossaller et al. (2006) found that CD40L-deficient patients, like ICOS (604558)-deficient patients, had abrogated germinal center formation and a severe reduction of CXCR5 (BLR1; 601613)-positive T cells.

Using flow cytometric analysis, van Zelm et al. (2014) found reduced numbers of all memory B-cell subsets except CD27 (TNFRSF7; 186711)-negative/IgA-positive B cells in both CD19 (107265)-deficient patients and CD40L-deficient patients compared with controls. Analysis of transcripts after class switching demonstrated that patient transcripts had fewer somatic mutations and reduced usage of IgG2 and IgA2 subclasses. There was also a deficiency in selection strength of mutations for antigen binding in patients compared with controls, whereas selection to maintain superantigen binding was normal. Selection against the autoreactive properties of immunoglobulins was impaired in patients. Somatic hypermutation analysis revealed decreased AICDA and UNG activity in CD40L deficiency, but increased UNG activity and decreased mismatch repair in CD19 deficiency. Van Zelm et al. (2014) concluded that both the B-cell antigen receptor and CD40 signaling pathways are required for selection of immunoglobulin reactivity, but that they differentially mediate DNA repair pathways during somatic hypermutation and thereby together shape the mature B-cell repertoire.

X-Inactivation Studies

If the defect in the switch mechanism is intrinsic to the B cells, a skewed X chromosome inactivation pattern would be observed in IgG- and IgA-expressing B lymphocytes of female carriers. Hendriks et al. (1990) studied lymphoblastoid B cells from 2 female carriers (see HISTORY section). Hendriks et al. (1990) concluded that the HIGM1 gene encodes a class switch inducer that is transferred to B lymphocytes from a cell of synthesis, possibly T lymphocytes.

Contrary to the findings of Hendriks et al. (1990) and those of Conley et al. (1988), Notarangelo et al. (1991) found nonrandom X-chromosome inactivation in T cells, B cells, and neutrophils, but not in fibroblasts, of obligate carriers, suggesting that several different hematopoietic cell lineages are primarily involved in HIGM1. Preferential inactivation of the paternally derived X chromosome was demonstrated by analysis of segregation of the alleles defined by 2 DNA probes. Notarangelo et al. (1991) suggested that the HIGM1 mutation may confer an advantage in differentiation and/or proliferation to hematopoietic precursors carrying the mutant allele on the active X chromosome.

In studies of X-chromosome inactivation in carriers of HIGM1, Hollenbaugh et al. (1994) found that the CD40L gene is not selectively inactivated. Furthermore, even when there was extremely skewed inactivation, normal levels of serum immunoglobulins were found. Unlike some other X-linked defects in which extreme lyonization may lead to disease, a small population of cells expressing the wildtype protein was sufficient to maintain normal humoral immunity and prevent the clinical symptoms of the disorder. Kipps (1994) commented that the findings have encouraging implications for patients with the disorder, since it seems that only a relatively small fraction of the T cells need express a functional CD40-ligand for effective immunity. Even a partial reconstitution with precursor T cells capable of expressing a functional ligand might suffice.


Mapping

Mensink et al. (1987) concluded that the locus for immunodeficiency with increased IgM (symbolized XHM by them) is linked to the DXS42 RFLP locus, which maps to Xq24-q27. Recombination between XHM and DXS17 was observed, whereas no recombination between XLA and DXS17 has been found; thus, XHM and XLA are apparently determined by separate gene loci. Padayachee et al. (1992, 1993) narrowed the location to Xq26 by multipoint linkage studies demonstrating that it is close to HPRT (308000), a gene that forms part of an extensive YAC contig mapping to Xq26; a maximum lod score of 4.89 was obtained. The existence of an easily scorable VNTR of 5 alleles within the HPRT gene means that other families with X-linked hyper-IgM syndrome are likely to be informative for this polymorphism.

Aruffo et al. (1993) mapped the GP39 gene to Xq26 by PCR analysis of a regional mapping panel, followed up by fluorescence in situ hybridization for precise localization. By YAC analysis, Pilia et al. (1994) mapped the CD40L locus between DXS144E and DXS300 in Xq26 and determined its transcription to be from 5-prime centromeric to 3-prime telomeric. This corresponded to the site where the clinical phenotype of the hyper-IgM syndrome had been mapped.

Allen et al. (1993) mapped the CD40LG gene (300386) to the proximal region of the mouse X chromosome, linked to Hprt. Hprt maps to the Xq26-q27.2 region, which suggested that the human CD40LG gene would also map to this region. This was confirmed by fluorescence in situ hybridization studies of CD40LG by Graf et al. (1992) and Allen et al. (1993).


Molecular Genetics

Allen et al. (1993) presented conclusive evidence that the defect in X-linked hyper-IgM syndrome resides in the gene for the CD40 ligand (300386). Because CD40LG induces B-cell proliferation in the absence of any costimulus and because the hyper-IgM phenotype and the CD40LG gene map to the same location, CD40LG was suggested as the site of the mutation in HIGM1. Allen et al. (1993) demonstrated this to be case by the finding of point mutations in 3 of 4 patients with the syndrome (300386.0003-300386.0005). Similarly, Aruffo et al. (1993) identified mutations in the CD40LG gene in patients with the syndrome (300386.0001-300386.0002).

In a 41-year-old man with HIGM1, Palterer et al. (2022) identified a heterozygous missense mutation (M36K; 300386.0015) in the transmembrane domain of the CD40LG gene. The expression of CD40L was reduced on activated T CD4+ cells from the patient. The patient had an atypical clinical presentation that included leishmaniasis and hypogammaglobulinemia. Palterer et al. (2022) concluded that variants in the transmembrane domain of CD40LG act as hypomorphic variants and could lead to atypical clinical features.


Animal Model

Rosen (1975) stated that 'a similar syndrome of X-linked immunodeficiency with increased IgM has been found in mice.' Xu et al. (1994) generated CD40LG-deficient mice by targeted disruption.

Using intravital microscopy and histologic examination of arterioles in mice lacking CD40L, Andre et al. (2002) observed frequent rupture and embolization of thrombi. The thrombi showed lower platelet density compared to those of wildtype mice, which contributed to platelet fragility. Administration of recombinant soluble CD40L (rsCD40L), but not rsCD40L with a mutation changing the KGD motif sequence to KGE, restored thrombus stability even in CD40 -/- mice, indicating that CD40L is not acting through CD40 ligation. Evaluation of hemostasis suggested that the thrombus instability in CD40L -/- mice is not due a lack of fibrin formation but rather a defect in platelet-platelet interaction which could be stabilized by the wildtype, but not by the mutant, rsCD40L. Flow cytometric analysis demonstrated that rsCD40L binds to activated platelets of wildtype as well as of CD40 -/- mice, but that this binding can be inhibited by a peptide interfering with ITGA2B (607759)/ITGB3 (173470) binding. Plate-binding analysis indicated specific saturable binding of rsCD40L to ITGA2B/ITGB3. Fluorescence microscopy showed that human platelets spread on but did not adhere to an rsCD40L-coated glass surface only in the absence of an inhibitor of ITGA2B/ITGB3 binding. Andre et al. (2002) concluded that CD40L is an ITGA2B/ITGB3 ligand, a platelet agonist, and necessary for the stability of arterial thrombi. They also noted that these findings suggest the careful evaluation of clinical trials with anti-CD40L therapy.


History

Ramesh et al. (1999) reviewed the history as well as other aspects of the hyper-IgM syndrome, which they abbreviated HIM. In the WHO classification of immunodeficiencies, an entity termed X-linked immunodeficiency with increased IgM was listed (Fudenberg et al., 1970). A definition of the disorder was an outcome of an international workshop (Cooper et al., 1974). It was originally hypothesized that B lymphocytes from patients with HIGM1 have an intrinsic inability to undergo immunoglobulin isotype switch.

Levitt et al. (1983) suggested that this disorder has a primary dysfunction of B-lymphocyte isotype switching. In 4 male patients with hyper-IgM immunodeficiency, the number, proportion, and proliferation of T lymphocytes were shown to be normal. IgG and IgA B lymphocytes were completely absent. In vitro stimulation of patients' B cells with both T cell-dependent and T cell-independent activators failed to induce any IgG or IgA-producing B cells. They concluded that individuals with this disorder possess an intrinsic B cell dysfunction that is not related to abnormal T cell regulation.

The observation that patients with HIM and particularly those with the X-linked form of the disorder (XHIM) were prone to opportunistic infections suggested a T-cell defect in spite of laboratory evidence for a humoral immune deficiency. The hypothesis of a primary T-cell defect was elegantly supported by the studies of Mayer et al. (1986), who demonstrated that B cells from XHIM patients could be driven to secrete immunoglobulins of various isotypes in the presence of pokeweed mitogen when cocultivated with 'helper T lymphoblasts' from a patient with a Sezary-like syndrome, a neoplastic disorder of T cells.

Hendriks et al. (1990) studied lymphoblastoid B cells from 2 female carriers who had IgG- and IgA-expressing B cells, in order to determine if the defect in the switch mechanism is intrinsic to the B cells. In an analysis of differential methylation of the polymorphic DXS255 locus, random X chromosome inactivation patterns were found in populations of T lymphocytes, in IgM-expressing B lymphocytes, and in IgG- or IgA-expressing B lymphocytes. Similar extent of heterogeneity was found for Ig H chain rearrangements and the Ig light chain usage in the IgA- or IgG-expressing B cell that had inactivated the X chromosome which carried the intact gene and in clones with the mutant-bearing X chromosome inactivated. The results indicated that the intrinsic Ig H chain class switch mechanism in this disorder is fully intact in B lymphocytes. When B lymphocytes with the X chromosome bearing the mutant IMD3 gene on the active X chromosome are placed in an in vivo environment that contains the intact gene product, i.e., in the female heterozygotes, the switch from IgM to IgG or IgA production occurs at normal frequencies. These findings are consistent with the observation that switch of hyper-IgM B cells is induced by Sezary-like T lymphoblasts (Mayer et al., 1986).


REFERENCES

  1. Allen, R. C., Armitage, R. J., Conley, M. E., Rosenblatt, H., Jenkins, N. A., Copeland, N. G., Bedell, M. A., Edelhoff, S., Disteche, C. M., Simoneaux, D. K., Fanslow, W. C., Belmont, J., Spriggs, M. K. CD40 ligand gene defects responsible for X-linked hyper-IgM syndrome. Science 259: 990-993, 1993. [PubMed: 7679801] [Full Text: https://doi.org/10.1126/science.7679801]

  2. Andre, P., Prasad, K. S. S., Denis, C. V., He, M., Papalia, J. M., Hynes, R. O., Phillips, D. R., Wagner, D. D. CD40L stabilizes arterial thrombi by a beta(3) integrin-dependent mechanism. Nature Med. 8: 247-252, 2002. [PubMed: 11875495] [Full Text: https://doi.org/10.1038/nm0302-247]

  3. Aruffo, A., Farrington, M., Hollenbaugh, D., Li, X., Milatovich, A., Nonoyama, S., Bajorath, J., Grosmaire, L. S., Stenkamp, R., Neubauer, M., Roberts, R. L., Noelle, R. J., Ledbetter, J. A., Francke, U., Ochs, H. D. The CD40 ligand, gp39, is defective in activated T cells from patients with X-linked hyper-IgM syndrome. Cell 72: 291-300, 1993. [PubMed: 7678782] [Full Text: https://doi.org/10.1016/0092-8674(93)90668-g]

  4. Aschermann, Z., Gomori, E., Kovacs, G. G., Pal, E., Simon, G., Komoly, S., Marodi, L., Illes, Z. X-linked hyper-IgM syndrome associated with a rapid course of multifocal leukoencephalopathy. Arch. Neurol. 64: 273-276, 2007. [PubMed: 17296845] [Full Text: https://doi.org/10.1001/archneur.64.2.273]

  5. Bossaller, L., Burger, J., Draeger, R., Grimbacher, B., Knoth, R., Plebani, A., Durandy, A., Baumann, U., Schlesier, M., Welcher, A. A., Peter, H. H., Warnatz, K. ICOS deficiency is associated with a severe reduction of CXCR5+CD4 germinal center Th cells. J. Immun. 177: 4927-4932, 2006. [PubMed: 16982935] [Full Text: https://doi.org/10.4049/jimmunol.177.7.4927]

  6. Brahmi, Z., Lazarus, K. H., Hodes, M. E., Baehner, R. L. Immunologic studies of three family members with the immunodeficiency with hyper-IgM syndrome. J. Clin. Immun. 3: 127-134, 1983. [PubMed: 6602145] [Full Text: https://doi.org/10.1007/BF00915483]

  7. Conley, M. E., Brown, P., Pahwa, S., Puck, J. M. An intrinsic B cell defect in X-linked hyper IgM syndrome. (Abstract) Pediat. Res. 23: 353A, 1988.

  8. Cooper, M. D., Faulk, W. P., Fudenberg, H. H., Good, R. A., Hitzig, W., Kunkel, H. G., Roitt, I. M., Rosen, F. S., Seligmann, M., Soothill, J. F. Meeting report of the second international workshop on primary immunodeficiency diseases in man. Clin. Immun. Immunopath. 2: 416-445, 1974. [PubMed: 4596971] [Full Text: https://doi.org/10.1016/0090-1229(74)90059-2]

  9. Cunningham, C. K., Bonville, C. A., Ochs, H. D., Seyama, K., John, P. A., Rotbart, H. A., Weiner, L. B. Enteroviral meningoencephalitis as a complication of X-linked hyper IgM syndrome. J. Pediat. 134: 584-588, 1999. [PubMed: 10228294] [Full Text: https://doi.org/10.1016/s0022-3476(99)70245-3]

  10. Dunn, K., Lubens, R., Stiehm, E. R. Reversal of neutropenia in X-linked immunodeficiency with hyper-IgM by large doses of plasma. (Abstract) Clin. Res. 30: 125A, 1982.

  11. Fudenberg, H. H., Good, R. A., Hitzig, W., Kunkel, H. G., Roitt, I. M., Rosen, F. S., Rowe, D. S., Seligmann, M., Soothill, J. R. Classification of the primary immunodeficiencies (WHO recommendation). New Eng. J. Med. 283: 656-657, 1970. [PubMed: 5456822] [Full Text: https://doi.org/10.1056/NEJM197009172831211]

  12. Fuleihan, R., Ramesh, N., Loh, R., Jabara, H., Rosen, F. S., Chatila, T., Fu, S. M., Stamenkovic, I., Geha, R. S. Defective expression of the CD40 ligand in X chromosome-linked immunoglobulin deficiency with normal or elevated IgM. Proc. Nat. Acad. Sci. 90: 2170-2173, 1993. [PubMed: 7681587] [Full Text: https://doi.org/10.1073/pnas.90.6.2170]

  13. Gennery, A. R., Clark, J. E., Flood, T. J., Abinun, M., Cant, A. J. T-cell-depleted bone marrow transplantation from allogeneic sibling for X-linked hyperimmunoglobulin M syndrome. (Letter) J. Pediat. 137: 290 only, 2000. Note: Erratum: J. Pediat. 137: 592 only, 2000. [PubMed: 10931436] [Full Text: https://doi.org/10.1067/mpd.2000.106445]

  14. Gleich, G. J., Condemi, J. J., Vaughan, J. H. Dysgammaglobulinemia in the presence of plasma cells. New Eng. J. Med. 272: 331-340, 1965. [PubMed: 14239114] [Full Text: https://doi.org/10.1056/NEJM196502182720702]

  15. Graf, D., Korthauer, U., Mages, H. W., Senger, G., Kroczek, R. A. Cloning of TRAP, a ligand for CD40 on human T cells. Europ. J. Immun. 22: 3191-3194, 1992. [PubMed: 1280226] [Full Text: https://doi.org/10.1002/eji.1830221226]

  16. Hadzic, N., Pagliuca, A., Rela, M., Portmann, B., Jones, A., Veys, P., Heaton, N. D., Mufti, G. J., Mieli-Vergani, G. Correction of the hyper-IgM syndrome after liver and bone marrow transplantation. New Eng. J. Med. 342: 320-324, 2000. [PubMed: 10655530] [Full Text: https://doi.org/10.1056/NEJM200002033420504]

  17. Hasegawa, S., Imai, K., Yoshida, K., Okuno, Y., Muramatsu, H., Shiraishi, Y., Chiba, K., Tanaka, H., Miyano, S., Kojima, S., Ogawa, S., Morio, T., Mizutani, S., Takagi, M. Whole-exome sequence analysis of ataxia telangiectasia-like phenotype. J. Neurol. Sci. 340: 86-90, 2014. [PubMed: 24631270] [Full Text: https://doi.org/10.1016/j.jns.2014.02.033]

  18. Hayward, A. R., Levy, J., Facchetti, F., Notarangelo, L., Ochs, H. D., Etzioni, A., Bonnefoy, J. Y., Cosyns, M., Weinberg, A. Cholangiopathy and tumors of the pancreas, liver, and biliary tree in boys with X-linked immunodeficiency with hyper-IgM. J. Immun. 158: 977-983, 1997. [PubMed: 8993019]

  19. Hendriks, R. W., Kraakman, M. E. M., Craig, I. W., Espanol, T., Schuurman, R. K. B. Evidence that in X-linked immunodeficiency with hyperimmunoglobulinemia M the intrinsic immunoglobulin heavy chain class switch mechanism is intact. Europ. J. Immun. 20: 2603-2608, 1990. [PubMed: 1980111] [Full Text: https://doi.org/10.1002/eji.1830201212]

  20. Hollenbaugh, D., Wu, L. H., Ochs, H. D., Nonoyama, S., Grosmaire, L. S., Ledbetter, J. A., Noelle, R. J., Hill, H., Aruffo, A. The random inactivation of the X chromosome carrying the defective gene responsible for X-linked hyper IgM syndrome (X-HIM) in female carriers of HIGM1. J. Clin. Invest. 94: 616-622, 1994. [PubMed: 7518839] [Full Text: https://doi.org/10.1172/JCI117377]

  21. Jamieson, W. M., Kerr, M. R. A family with several cases of hypogammaglobulinemia. Arch. Dis. Child. 37: 330-336, 1962. [PubMed: 14451053] [Full Text: https://doi.org/10.1136/adc.37.193.330]

  22. Kipps, T. J. X inactivation and immunocompetence in female carriers of the X-linked hyper-IgM syndrome (Editorial) J. Clin. Invest. 94: 469, 1994. [PubMed: 8040297] [Full Text: https://doi.org/10.1172/JCI117355]

  23. Kyong, C. U., Virella, G., Fundenberg, H. H., Darby, C. P. X-linked immunodeficiency with increased IgM: clinical, ethnic, and immunologic heterogeneity. Pediat. Res. 12: 1024-1026, 1978. [PubMed: 724295] [Full Text: https://doi.org/10.1203/00006450-197810000-00015]

  24. Levitt, D., Haber, P., Rich, K., Cooper, M. D. Hyper IgM immunodeficiency: a primary dysfunction of B lymphocyte isotype switching. J. Clin. Invest. 72: 1650-1657, 1983. [PubMed: 6605368] [Full Text: https://doi.org/10.1172/JCI111124]

  25. Levy, J., Espanol-Boren, T., Thomas, C., Fischer, A., Tovo, P., Bordigoni, P., Resnick, I., Fasth, A., Baer, M., Gomez, L., Sanders, E. A. M., Tabone, M.-D., and 13 others. Clinical spectrum of X-linked hyper-IgM syndrome. J. Pediat. 131: 47-54, 1997. [PubMed: 9255191] [Full Text: https://doi.org/10.1016/s0022-3476(97)70123-9]

  26. Lin, Q., Rohrer, J., Allen, R. C., Larche, M., Greene, J. M., Shigeoka, A. O., Gatti, R. A., Derauf, D. C., Belmont, J. W., Conley, M. E. A single strand conformation polymorphism study of CD40 ligand: efficient mutation analysis and carrier detection of X-linked hyper IgM syndrome. J. Clin. Invest. 97: 196-201, 1996. [PubMed: 8550833] [Full Text: https://doi.org/10.1172/JCI118389]

  27. Mayer, L., Kwan, S. P., Thompson, C., Ko, H. S., Chiorazzi, N., Waldmann, T., Rosen, F. Evidence for a defect in 'switch' T cells in patients with immunodeficiency and hyperimmunoglobulinemia M. New Eng. J. Med. 314: 409-413, 1986. [PubMed: 3080678] [Full Text: https://doi.org/10.1056/NEJM198602133140703]

  28. Mensink, E. J. B. M., Thompson, A., Sandkuyl, L. A., Kraakman, M. E. M., Schot, J. D. L., Espanol, T., Schuurman, R. K. B. X-linked immunodeficiency with hyperimmunoglobulinemia M appears to be linked to the DXS42 restriction fragment length polymorphism locus. Hum. Genet. 76: 96-99, 1987. [PubMed: 2883112] [Full Text: https://doi.org/10.1007/BF00283057]

  29. Notarangelo, L. D., Duse, M., Ugazio, A. G. Immunodeficiency with hyper-IgM (HIM). Immunodef. Rev. 3: 101-122, 1992. [PubMed: 1554497]

  30. Notarangelo, L. D., Parolini, O., Albertini, A., Duse, M., Mazzolari, E., Plebani, A., Camerino, G., Ugazio, A. G. Analysis of X-chromosome inactivation in X-linked immunodeficiency with hyper-IgM (HIGM1): evidence for involvement of different hematopoietic cell lineages. Hum. Genet. 88: 130-134, 1991. [PubMed: 1757090] [Full Text: https://doi.org/10.1007/BF00206059]

  31. Padayachee, M., Feighery, C., Finn, A., McKeown, C., Levinsky, R. J., Kinnon, C., Malcolm, S. Mapping of the X-linked form of hyper-IgM syndrome (HIGM1) to Xq26 by close linkage to HPRT. Genomics 14: 551-553, 1992. [PubMed: 1427881] [Full Text: https://doi.org/10.1016/s0888-7543(05)80270-8]

  32. Padayachee, M., Levinsky, R. J., Kinnon, C., Finn, A., McKeown, C., Feighery, C., Notarangelo, L. D., Hendriks, R. W., Read, A. P., Malcolm, S. Mapping of the X linked form of hyper IgM syndrome (HIGM1). J. Med. Genet. 30: 202-205, 1993. Note: Erratum: J. Med. Genet. 30: 528 only, 1993. [PubMed: 8097258] [Full Text: https://doi.org/10.1136/jmg.30.3.202]

  33. Palterer, B., Salvati, L., Capone, M., Mecheri, V., Maggi, L., Mazzoni, A., Cosmi, L., Volpi, N., Tiberi, L., Provenzano, A., Giglio, S., Parronchi, P., Maggiore, G., Gallo, O., Bartoloni, A., Annunziato, F., Zammarchi, L., Liotta, F. Variant disrupting CD40L transmembrane domain and atypical X-linked hyper-IgM syndrome: a case report with leishmaniasis and review of the literature. Front. Immun. 13: 840767, 2022. [PubMed: 35572607] [Full Text: https://doi.org/10.3389/fimmu.2022.840767]

  34. Pilia, G., Porta, B., Padayachee, M., Malcolm, S., Zucchi, I., Villa, A., Macchi, P., Vezzoni, P., Schlessinger, D. Human CD40L gene maps between DXS144E and DXS300 in Xq26. Genomics 22: 249-251, 1994. [PubMed: 7959785] [Full Text: https://doi.org/10.1006/geno.1994.1378]

  35. Ramesh, N., Geha, R. S., Notarangelo, L. D. CD40 ligand and the hyper-IgM syndrome. In: Ochs, H. D.; Smith, C. I. E.; Puck, J. M. (eds.): Primary Immunodeficiency Diseases: A Molecular and Genetic Approach. New York: Oxford University Press 1999. Pp. 233-249.

  36. Rosen, F. S. Immunodeficiency. In: Benacerraf, B. (ed.): Immunogenetics and Immunodeficiency. London: William Clowes & Sons, Ltd. 1975. Pp. 229-257.

  37. Thomas, C., de Saint Basile, G., Le Deist, F., Theophile, D., Benkerrou, M., Haddad, E., Blanche, S., Fischer, A. Brief report: correction of X-linked hyper-IgM syndrome by allogenic bone marrow transplantation. New Eng. J. Med. 333: 426-429, 1995. [PubMed: 7542361] [Full Text: https://doi.org/10.1056/NEJM199508173330705]

  38. van Zelm, M. C., Bartol, S. J. W., Driessen, G. J., Mascart, F., Reisli, I., Franco, J. L., Wolska-Kusnierz, B., Kanegane, H., Boon, L., van Dongen, J. J. M., van der Burg, M. Human CD19 and CD40L deficiencies impair antibody selection and differentially affect somatic hypermutation. J. Allergy Clin. Immun. 134: 135-144, 2014. [PubMed: 24418477] [Full Text: https://doi.org/10.1016/j.jaci.2013.11.015]

  39. Xu, J., Foy, T. M., Laman, J. D., Elliott, E. A., Dunn, J. J., Waldschmidt, T. J., Elsemore, J., Noelle, R. J., Flavell, R. A. Mice deficient for the CD40 ligand. Immunity 1: 423-431, 1994. Note: Erratum: Immunity: 1: 613 only, 1994. [PubMed: 7882172] [Full Text: https://doi.org/10.1016/1074-7613(94)90073-6]


Contributors:
Hilary J. Vernon - updated : 07/07/2022
Paul J. Converse - updated : 11/6/2015
Cassandra L. Kniffin - updated : 9/22/2014
Cassandra L. Kniffin - updated : 9/28/2007
Paul J. Converse - updated : 3/8/2007
Cassandra L. Kniffin - updated : 8/23/2004
Victor A. McKusick - updated : 10/20/2003
Cassandra L. Kniffin - updated : 4/15/2002
Cassandra L. Kniffin - reorganized : 4/5/2002
Paul J. Converse - updated : 2/28/2002
Victor A. McKusick - updated : 2/14/2002
Ada Hamosh - updated : 2/28/2000
Ada Hamosh - reorganized : 2/11/2000
Victor A. McKusick - updated : 2/7/2000
Victor A. McKusick - updated : 9/23/1999
Victor A. McKusick - updated : 11/12/1998
Ada Hamosh - updated : 7/10/1997

Creation Date:
Victor A. McKusick : 6/4/1986

Edit History:
alopez : 08/18/2023
alopez : 08/18/2023
carol : 07/07/2022
carol : 06/01/2017
carol : 06/24/2016
mgross : 11/6/2015
carol : 9/22/2014
ckniffin : 9/22/2014
terry : 4/4/2013
carol : 3/26/2012
wwang : 6/16/2011
wwang : 10/4/2007
ckniffin : 9/28/2007
mgross : 3/8/2007
wwang : 10/27/2005
tkritzer : 8/26/2004
ckniffin : 8/23/2004
tkritzer : 10/21/2003
terry : 10/20/2003
mgross : 10/1/2003
mgross : 9/23/2003
mgross : 9/22/2003
ckniffin : 5/15/2003
terry : 6/27/2002
carol : 4/16/2002
carol : 4/15/2002
ckniffin : 4/15/2002
ckniffin : 4/12/2002
carol : 4/12/2002
ckniffin : 4/12/2002
ckniffin : 4/5/2002
carol : 4/5/2002
ckniffin : 4/5/2002
alopez : 2/28/2002
cwells : 2/21/2002
terry : 2/14/2002
carol : 9/10/2001
terry : 1/18/2001
mgross : 9/19/2000
mcapotos : 8/9/2000
alopez : 2/28/2000
carol : 2/14/2000
carol : 2/11/2000
carol : 2/11/2000
mcapotos : 2/11/2000
terry : 2/7/2000
alopez : 11/15/1999
carol : 10/13/1999
mgross : 10/8/1999
terry : 9/23/1999
carol : 12/14/1998
carol : 11/18/1998
terry : 11/12/1998
dkim : 9/11/1998
dkim : 9/10/1998
dkim : 7/21/1998
alopez : 5/21/1998
mark : 9/2/1997
mark : 9/1/1997
alopez : 7/29/1997
alopez : 7/10/1997
alopez : 7/10/1997
alopez : 6/11/1997
mark : 11/13/1996
terry : 11/7/1996
terry : 11/7/1996
terry : 10/31/1996
mark : 2/2/1996
terry : 2/1/1996
mark : 12/5/1995
mark : 10/5/1995
terry : 9/13/1995
carol : 10/11/1994
jason : 6/28/1994
davew : 6/6/1994
warfield : 4/20/1994