Fas (APO-1/CD95) is a cell-surface receptor involved in cell death signaling. Germline mutations in the Fas gene have been associated with autoimmune lymphoproliferative syndrome, and somaticFas mutations have been found in multiple myeloma. We have examined the entire coding region and all splice sites of theFas gene in 150 cases of non-Hodgkin’s lymphoma. Overall, mutations were identified in 16 of the tumors (11%). Missense mutations within the death domain of the receptor were associated with retention of the wild-type allele, indicating a dominant-negative mechanism, whereas missense mutations outside the death domain were associated with allelic loss. Fas mutations were identified in 3 (60%) MALT-type lymphomas, 9 (21%) diffuse large B-cell lymphomas, 2 (6%) follicle center cell lymphomas, 1 (50%) anaplastic large cell lymphoma, and 1 unusual case of B-cell chronic lymphocytic leukemia with a marked tropism for skin. Among the 16 patients with somaticFas mutations, 15 showed extranodal disease at presentation, and 6 relapsed in extranodal areas. Ten of 13 evaluable patients showed features suggestive of autoreactive disease. Our data indicate that somatic disruption of Fas may play a role in the pathogenesis of some lymphomas, and suggest a link between Fas mutation, cancer and autoimmunity.

© 1998 by The American Society of Hematology.

FAS (ALSO KNOWN AS APO-1 or CD95) is a transmembrane protein of the tumor necrosis factor (TNF) receptor family, which mediates programmed cell death (apoptosis) upon trimerization induced by cross-linking to Fas ligand (FasL).1,2 Fas is expressed on the surface of activated T and B lymphocytes, and Fas/FasL induced apoptosis is important for eliminating autoreactive immature T cells during ontogenesis and for maintaining peripheral lymphocyte homeostasis.2,3 

Disruption of the Fas/FasL apoptotic pathway has been associated with benign lymphoproliferation, severe multisystem autoimmune disease, and hypergammaglobulinemia. Lpr mice that harbor deleterious mutations in the Fas gene accumulate CD4CD8 (double negative) T cells in their lymph nodes and spleen, exhibit B-cell lymphocytosis, and produce large amounts of IgG and IgM autoantibodies, including anti-DNA antibodies, and rheumatoid (Rh) factor.4 Children who carry inherited defects in the Fas gene exhibit a similar, albeit variable, pattern of phenotypes that have been collectively termed autoimmune lymphoproliferative syndrome (ALPS).5-9 

Non-Hodgkin’s lymphomas (NHL) are malignant neoplasms whose normal counterparts are the cells of the immune system.10Different lines of evidence suggest that an association exists between NHL and autoimmune disease. Patients with autoimmune diseases, including systemic lupus erythematosis (SLE), rheumatoid arthritis (RA), Sjögren’s syndrome, and autoimmune thyroid disease, have an increased risk for hematopoietic cancers, in particular lymphoma,11-13 and T-cell–rich B-cell (TRB) lymphoma and Hodgkin’s disease have been reported in patients with ALPS.9 Conversely, approximately 8% of patients with NHL exhibit autoimmune phenomena.14 Here we show that 11% of sporadic NHL harbor Fas mutations, and that the majority of patients with Fas-mutated lymphomas present with extranodal disease and clinical features suggestive of autoreactive disease.

Patients.

A total of 150 NHLs were included in this study. The patient samples had been frozen immediately after excision in either liquid N2 or a mixture of 2-methyl butane and dry ice and stored at −80°C until use. Routinely processed histological samples were available in all cases. These samples were stained with hematoxylin-eosin, examined by immunohistology, and then classified according to the Revised European-American Lymphoma (REAL) classification.10 Our series included the following histological subtypes: B-cell chronic lymphocytic leukemia (B-CLL) (n = 17); immunocytoma (n = 1); follicle center cell lymphoma (n = 33); mucosa-associated lymphoid tissue (MALT)-type lymphoma (n = 5); mantle cell lymphoma (n = 9); diffuse large B-cell lymphoma (DLC-B) (n = 43); Burkitt lymphoma (n = 5); peripheral T-cell lymphoma, unspecified (n = 35); and anaplastic large cell lymphoma of null cell type (n = 2). Uninvolved normal tissue was available as paraffin-embedded sections. Ethical committee approval for the study was obtained.

DNA isolation, denaturing gradient gel electrophoresis (DGGE), and direct sequencing.

Genomic DNA was isolated by proteinase K digestion and phenol-chloroform extraction, or by using the Puregene DNA Isolation Kit (Gentra Systems, Minneapolis, MN). Paraffin-embedded tissue was treated with xylene before DNA extraction. Mutations in the Fasgene were detected by polymerase chain reaction (PCR) amplification of genomic DNA using the 10 sets of primers listed in Table 1, followed by DGGE.15The melting characteristics of each of the nine exons with adjacent intronic sequences were attained by means of the MELT87 computer algorithm.16 To modulate the melting properties into the two-domain profile that is considered optimal for resolution of mutations, each sequence was tailored by PCR-mediated attachment of a “GC-clamp.”17 PCR was performed in 15-μL reaction mixtures containing 10 mmol/L Tris-HCl (pH 8.3), 50 mmol/L KCl, 1.5 mmol/L MgCl2, 0.002% gelatin, 0.2 mmol/L cresol red, 12% sucrose, 10 pmol of each primer, 100 μmol/L each dNTP, 100 ng of DNA, and 0.8 U of AmpliTaq polymerase (Perkin-Elmer Cetus, Emeryville, CA). The cycling parameters were: 38 cycles at 94°C for 20 seconds, 55°C for 20 seconds, and 72°C for 30 seconds. PCR products were analyzed in a 10% denaturant/6% polyacrylamide-70% denaturant/12% polyacrylamide double-gradient gel18; (100% denaturant = 7 mol/L urea and 40% formamide). The gel was run at 90 V for 16 hours in 1 × TAE buffer kept at a constant temperature of 54°C (58°C for exon 1), stained with ethidium bromide, and photographed under UV transillumination. The biallelic polymorphism at position −670 in the Fas promoter19 was detected by using primers FAS-734GC and FAS-623 (Table 1) and the PCR and DGGE conditions described above. Direct sequence analysis of PCR products was performed with a nonclamped, 33P-end-labeled primer using the ThermoPrime Cycle Sequencing Kit (Amersham Life Science, Cleveland, OH), according to the manufacturer’s instructions.

Table 1.

Amplification Primers for DGGE-Based Mutation Analysis of the Fas Gene

Exon/ Primer Sequence (5′ → 3′)*Product Length (bp) Tm of Domain (°C)-151
FAS-734GC  [GC]-TCCCTTTTCAGAGCCCTATGG  178 75.6  
FAS-623  GACTTGCGGGGCATTTGAC  
1-F-152 cgccg[GC]-TCAGTACGGAGTTGGGGAAGC  182  76.9  
1-R GCCTATCCCCGGGACTAAGAC  
2-F  [GC]-ATCAATAAAATTCTCTTCATGC 190  68.9  
2-R  TGACTTTCACTGTAATCTCTGG  
3-F AAACACTTGCTCCTTTTTTCC  254  71.7  
3-R [GC]-TGAAATTCCAAGATTGGCC  
4-F  gcccgTCCAAACTGATTTTCTAGGC 207  69.5  
4-R  [GC]-TCTAGTGTTTTAATCAGAGAAAGAC  
5-F [GC]-CCAGGCTTTTGAATTTCTCC  182  69.1  
5-R ccgcgccgGGGAAAGGAGGATATAACCG  
6-F  ATAATATGCCAATGTTCCAACC 175  69.4  
6-R  [GC]-CCCCAAGTTATTTCAATCTGC  
7-F CATGCATTCTACAAGGCTGAG  255  67.2  
7-R [GC]-AGGAAGTAACAAAAAGCCAAATC  
8-F  TCTCTGCTTCCATTTTTTGC 159  65.4  
8-R  [GC]-TTTACTCTGAAATTGGCCTATTAC  
9I-F [GC]-TATTTTCTATTTTTCAGATGTTGAC  275  67.9  
9I-R TCATACGCTTCTTTCTTTCC  
9II-F  GTTCAACTGCTTCGTAATTG  251 67.6  
9II-R  [GC]-AGAACTGAATTTGTTGTTTTTC 
Exon/ Primer Sequence (5′ → 3′)*Product Length (bp) Tm of Domain (°C)-151
FAS-734GC  [GC]-TCCCTTTTCAGAGCCCTATGG  178 75.6  
FAS-623  GACTTGCGGGGCATTTGAC  
1-F-152 cgccg[GC]-TCAGTACGGAGTTGGGGAAGC  182  76.9  
1-R GCCTATCCCCGGGACTAAGAC  
2-F  [GC]-ATCAATAAAATTCTCTTCATGC 190  68.9  
2-R  TGACTTTCACTGTAATCTCTGG  
3-F AAACACTTGCTCCTTTTTTCC  254  71.7  
3-R [GC]-TGAAATTCCAAGATTGGCC  
4-F  gcccgTCCAAACTGATTTTCTAGGC 207  69.5  
4-R  [GC]-TCTAGTGTTTTAATCAGAGAAAGAC  
5-F [GC]-CCAGGCTTTTGAATTTCTCC  182  69.1  
5-R ccgcgccgGGGAAAGGAGGATATAACCG  
6-F  ATAATATGCCAATGTTCCAACC 175  69.4  
6-R  [GC]-CCCCAAGTTATTTCAATCTGC  
7-F CATGCATTCTACAAGGCTGAG  255  67.2  
7-R [GC]-AGGAAGTAACAAAAAGCCAAATC  
8-F  TCTCTGCTTCCATTTTTTGC 159  65.4  
8-R  [GC]-TTTACTCTGAAATTGGCCTATTAC  
9I-F [GC]-TATTTTCTATTTTTCAGATGTTGAC  275  67.9  
9I-R TCATACGCTTCTTTCTTTCC  
9II-F  GTTCAACTGCTTCGTAATTG  251 67.6  
9II-R  [GC]-AGAACTGAATTTGTTGTTTTTC 

*Lowercase characters represent nucleotides incorporated into the primer to modulate the melting profile of the amplification product.

F0-151

Melting temperature of the lower melting domain of the GC-clamped PCR product, as determined by MELT87.16 

F0-152

F, forward primer; R, reverse primer; [GC], CGCCCGCCGCGCCCCGCGCCCGTCCCGCCGCCCCCGCCCG.

Reverse transcriptase (RT)-PCR analysis.

RNA was extracted using the Purescript Isolation Kit (Gentra Systems). cDNA synthesis was performed using M-MLV SuperScript II reverse transcriptase (GIBCO-BRL, Life Technologies, Gaithersburg, MD) in a total volume of 20 μL 1× buffer (GIBCO-BRL, Life Technologies) containing 10 mmol/L dithiothreitol (DTT). Incubations were performed at 42°C for 50 minutes, 72°C for 5 minutes. Fas cDNA was PCR amplified with primers FAS533 (5′-GCAGAAAGCACAGAAAGGAAA-3′) and FAS737 (5′-TCTGTTCTGCTGTGTCTTGGA-3′) which hybridize to regions inFas exons 7 and 9, respectively, and yield a PCR product of 235 bp. Amplifications were performed in a total volume of 25 μL containing 1× PCR buffer (50 mmol/L KCl, 20 mmol/L Tris pH 8.4, 2.0 mmol/L MgCl2, 0.2 mmol/L cresol red, 12% sucrose, 0.005% [wt/vol] bovine serum albumin [BSA] [Boehringer-Mannheim, Mannheim, Germany]), 2.5 pmol of each primer, 40 mmol/L dNTPs, and 1.25 U of AmpliTaq polymerase (Perkin Elmer Cetus). The parameters used for amplification were 94°C for 20 seconds, 60°C for 20 seconds, and 72°C for 30 seconds for 40 cycles. Taq polymerase and dNTPs were added to the reaction tube at an 80°C step between the denaturation and annealing steps of the first cycle (“Hot start”). Direct sequence analysis was performed as described above.

Genomic DNA was isolated from 150 NHLs and analyzed for mutations in all 9 exons of the Fas gene by PCR/DGGE analysis (Fig 1A). Enrichment and direct sequence analysis of aberrantly migrating bands led to the identification of mutations in 16 of the samples (11%) (Fig 1B; Table 2). Normal tissue was available from 9 of 16 mutated cases. None of the normal samples showed evidence of mutations by DGGE, indicating that the mutations detected in the lymphoma specimens had arisen somatically.

Fig. 1.

Detection of Fas mutations in NHL. (A) The 5′-end of exon 9 of the Fas gene was amplified using primers 9I-F and 9I-R, and mutations were detected by DGGE analysis in follicle center cell lymphoma (FCC) 17 (E256K), MALT 145 (N248K), DLC-B 156 (N248K), DLC-B 93 (L262F), and DLC-B 86 (D244V). PBL, DNA isolated from peripheral blood lymphocytes of a normal volunteer. (B) Direct sequence analysis of heteroduplexes recovered from the gel shown in (A), revealing the G1008A transition (E256K) in FCC 17.

Fig. 1.

Detection of Fas mutations in NHL. (A) The 5′-end of exon 9 of the Fas gene was amplified using primers 9I-F and 9I-R, and mutations were detected by DGGE analysis in follicle center cell lymphoma (FCC) 17 (E256K), MALT 145 (N248K), DLC-B 156 (N248K), DLC-B 93 (L262F), and DLC-B 86 (D244V). PBL, DNA isolated from peripheral blood lymphocytes of a normal volunteer. (B) Direct sequence analysis of heteroduplexes recovered from the gel shown in (A), revealing the G1008A transition (E256K) in FCC 17.

Close modal
Table 2.

Fas Mutations in 16 Lymphomas

No. Histology Nucleotide Change*LocalizationCodon Predicted Effect
193  B-CLL  G240A Exon 2  Signal peptide  Ala → Thr  
49 FCC + DLC-B (TRB)  598 ins T  Exon 4  119 Frameshift  
128  DLC-B  C732T  Exon 6  164 Leu → Phe  
72  ALCL null  C742T  Exon 6  167 Pro → Leu  
157  DLC-B (TRB)  C787T  Exon 7  182 Thr → Ile  
69  DLC-B  T838A  Exon 7  199 Leu → STOP  
225  DLC-B  IVS7nt − 2a → g Intron 7  —  Splice defect  
146  MALT IVS8nt + 5g → a  Intron 8  —  Splice defect  
67  MALT  IVS8nt + 5g → c  Intron 8  — Splice defect  
92  DLC-B  T865G  Exon 8 208  Leu → STOP  
86  DLC-B  A973T  Exon 9 244  Asp → Val  
156  DLC-B  T986A  Exon 9 248  Asn → Lys  
145  MALT  T986G  Exon 9 248  Asn → Lys  
17  FCC  G1008A  Exon 9  256 Glu → Lys  
93  DLC-B  C1026T  Exon 9  262 Leu → Phe  
141  DLC-B  A1091T  Exon 9  283 Lys → Asn 
No. Histology Nucleotide Change*LocalizationCodon Predicted Effect
193  B-CLL  G240A Exon 2  Signal peptide  Ala → Thr  
49 FCC + DLC-B (TRB)  598 ins T  Exon 4  119 Frameshift  
128  DLC-B  C732T  Exon 6  164 Leu → Phe  
72  ALCL null  C742T  Exon 6  167 Pro → Leu  
157  DLC-B (TRB)  C787T  Exon 7  182 Thr → Ile  
69  DLC-B  T838A  Exon 7  199 Leu → STOP  
225  DLC-B  IVS7nt − 2a → g Intron 7  —  Splice defect  
146  MALT IVS8nt + 5g → a  Intron 8  —  Splice defect  
67  MALT  IVS8nt + 5g → c  Intron 8  — Splice defect  
92  DLC-B  T865G  Exon 8 208  Leu → STOP  
86  DLC-B  A973T  Exon 9 244  Asp → Val  
156  DLC-B  T986A  Exon 9 248  Asn → Lys  
145  MALT  T986G  Exon 9 248  Asn → Lys  
17  FCC  G1008A  Exon 9  256 Glu → Lys  
93  DLC-B  C1026T  Exon 9  262 Leu → Phe  
141  DLC-B  A1091T  Exon 9  283 Lys → Asn 

*Numbering according to GenBank accession no. M67454; ALCL, anaplastic large cell lymphoma; FCC, follicle center cell lymphoma.

Missense mutations.

The majority (10 of 16) of the mutations were missense variants, all of which caused nonconservative amino acid substitutions (Table 2). Six of these mutations were detected in exon 9, which encodes the death domain region of the Fas receptor. Two different transversions (T to A and T to G) at position 986 of the Fas cDNA sequence (GenBank accession no. M67454), both causing the substitution of Asn with Lys at codon 248, were found in two different tumors, suggesting that this position may represent a mutational hotspot. The remaining mutations within the death domain affected codons 244, 256, 262, and 283. Missense mutations outside the death domain involved the last residue of the signal peptide, which directs the Fas molecule to the endoplasmatic reticulum; codons 164 and 167 in exon 6, which encodes the transmembrane region of membrane-bound Fas; and codon 182 in exon 7, which encodes the intracytoplasmic anchoring region.

Allelic status.

Because missense mutations in the death domain of Fas in patients with ALPS have been suggested to affect receptor function in a dominant-negative fashion,5 we examined the allelic status of Fas in tumors carrying missense mutations. Six of the patients were heterozygous for one or both of the known biallelic polymorphims at positions −67019 and 836,20 allowing evaluation of allelic loss in their tumors by DGGE analysis (Fig 2). All three informative lymphomas carrying missense mutations in exon 9 showed equal distribution of the two alleles, indicating retention of the wild-type allele in the tumor cells. In contrast, all three tumors with missense mutations in exons 6 or 7 showed unequal distribution of the two alleles, suggesting that the wild-type allele had been lost (Fig2). Only 4 of 76 (5%) informative lymphomas in which no Fasmutations had been detected showed evidence of allelic loss.

Fig. 2.

Detection of allelic loss of the Fas gene in NHL by DGGE. A region encompassing the biallelic polymorphism, −670A/G, in the Fas promoter was amplified with primers FAS-734GC and FAS-623, and the two alleles were subsequently resolved by electrophoresis in a denaturing gradient gel. Unequal distribution of the two alleles was observed in DLC-B 128, anaplastic large cell lymphoma (ALCL) 72, and DLC-B 157, suggesting that oneFas allele was lost in the tumor cells. These three tumors harbor missense mutations in exon 6 or exon 7 of the Fas gene. In contrast, even distribution of the two alleles was observed in MALT 145, which harbors the N248K mutation in exon 9 of the Fasgene. PBL, DNA isolated from peripheral blood lymphocytes of a normal volunteer who is heterozygous for the −670A/G polymorphism.

Fig. 2.

Detection of allelic loss of the Fas gene in NHL by DGGE. A region encompassing the biallelic polymorphism, −670A/G, in the Fas promoter was amplified with primers FAS-734GC and FAS-623, and the two alleles were subsequently resolved by electrophoresis in a denaturing gradient gel. Unequal distribution of the two alleles was observed in DLC-B 128, anaplastic large cell lymphoma (ALCL) 72, and DLC-B 157, suggesting that oneFas allele was lost in the tumor cells. These three tumors harbor missense mutations in exon 6 or exon 7 of the Fas gene. In contrast, even distribution of the two alleles was observed in MALT 145, which harbors the N248K mutation in exon 9 of the Fasgene. PBL, DNA isolated from peripheral blood lymphocytes of a normal volunteer who is heterozygous for the −670A/G polymorphism.

Close modal
Nonsense, frameshift, and splice-site mutations.

The six non-missense mutations included two point mutations introducing premature termination signals at codons 199 and 208, respectively; one 1-bp insertion causing a frame shift and the introduction of a stop codon at residue 120; and three mutations affecting normal splicing ofFas mRNA (Table 2). Two of the splice mutations affected position +5 of the consensus sequence of the donor splice site of intron 8, while the remaining mutation was a transition of the invariable A at position −2 of the acceptor splice site of intron 7. Mutations at these particular splice site positions have been shown to cause cryptic splice site utilization or exon skipping in various human disease genes.21 RT-PCR analysis of a region encompassing exons 7, 8, and 9 in RNA extracted from these three tumors showed the occurrence of a shorter band in all three cases (Fig 3). Cloning and sequence analysis showed that this band lacked the sequence corresponding to exon 8, resulting in a frame shift and the introduction of a stop codon at residue 221. Together, these data suggest that all three mutations located in the splice-site consensus regions of exon 8 result in exon skipping. However, the exact ratio of normal to aberrantly spliced mRNA in tumor cells remains unknown.

Fig. 3.

RT-PCR analysis of Fas mRNA in NHLs. A region encompassing exons 7-9 was amplified with primers FAS533 and FAS737 in three lymphoma samples in which mutations had been identified in the acceptor splice site of Fas intron 7 (IVS7nt-2a → g; DLC-B 225), or in the donor splice site of Fas intron 8 (IVS8nt + 5g → a, MALT 146; and IVS8nt + 5g → c, MALT 67). In all three samples, skipping of exon 8 was demonstrated by the occurrence of a shorter band that was not present in RNA isolated from peripheral blood lymphocytes of a normal volunteer (PBL). M, 100-bp ladder.

Fig. 3.

RT-PCR analysis of Fas mRNA in NHLs. A region encompassing exons 7-9 was amplified with primers FAS533 and FAS737 in three lymphoma samples in which mutations had been identified in the acceptor splice site of Fas intron 7 (IVS7nt-2a → g; DLC-B 225), or in the donor splice site of Fas intron 8 (IVS8nt + 5g → a, MALT 146; and IVS8nt + 5g → c, MALT 67). In all three samples, skipping of exon 8 was demonstrated by the occurrence of a shorter band that was not present in RNA isolated from peripheral blood lymphocytes of a normal volunteer (PBL). M, 100-bp ladder.

Close modal
Polymorphisms.

In addition to the known polymorphisms in the enhancer region and in exons 3 and 7,19,20 we identified two previously undescribed single base changes, G377A in exon 2 and G563A in exon 4, which are predicted to leave the amino acid sequence unchanged. The A-allele of the G377A polymorphism and the A-allele of the G563A polymorphism were each found at a frequency of 1.7% (5 of 300 independent chromosomes).

Clinical and histological features.

The histological features of the Fas-mutated lymphomas are outlined in Table 3. Mutations were identified most frequently in low-grade MALT-type lymphomas (3 of 5; 60%) and DLC-B lymphomas (9 of 43; 21%). All but one of the mutated cases were B-cell lymphomas. The remaining case was an anaplastic large cell lymphoma of null cell type.

Table 3.

Distribution of Fas Mutations According to the Histological Subtypes

Histological Subtype Mutated Cases %
B-CLL  1/17 6  
Immunocytoma  0/1  0  
Follicle center cell lymphoma 2/33  6  
B-cell lymphoma of low-grade MALT type  3/5 60  
Mantle cell lymphoma  0/9  0  
Diffuse large B-cell lymphoma  9/43  21  
Burkitt lymphoma  0/5  
Peripheral T-cell lymphoma, unspecified  0/35  
Anaplastic large cell lymphoma, null cell type  1/2 50 
Histological Subtype Mutated Cases %
B-CLL  1/17 6  
Immunocytoma  0/1  0  
Follicle center cell lymphoma 2/33  6  
B-cell lymphoma of low-grade MALT type  3/5 60  
Mantle cell lymphoma  0/9  0  
Diffuse large B-cell lymphoma  9/43  21  
Burkitt lymphoma  0/5  
Peripheral T-cell lymphoma, unspecified  0/35  
Anaplastic large cell lymphoma, null cell type  1/2 50 

The clinical features of the patients with Fas-mutated lymphomas are summarized in Table 4. Fifteen of 16 patients (94%) showed extranodal disease at presentation, and 6 developed extranodal recurrences. Of 117 lymphomas with no detectable Fas mutations, 57 (49%) presented at extranodal sites, and 32 (27%) showed involvement other than bone marrow. CLL was excluded from this statement because of its implicit involvement of extranodal areas.

Table 4.

Clinical and Histological Features in 16 Patients WithFas-Mutated Lymphomas

Case No. Histology Fas MutationPrimary Localization Localization at ProgressionDisease Status at Biopsy Autoimmune/ Paraneoplastic Phenomena Age Sex Survival From Biopsy (mo)
193  B-CLL  A-1T3-150 Blood, bone marrow Skin  Progression  Maculo-papular skin rash  78  6  
17  FCC  E256K  Waldeyers’ ring, lymph nodes Parotoid gland, lymph nodes  Progression  None  43 M  48  
49  FCC + DLC-B (TRB)  598 Ins T  Spleen Lymph nodes  Diagnosis  NA  63  M  63  
67 MALT-type  IVS8nt + 5g → c  Orbit, bone marrow, liver/spleen  Skin, lymph nodes  Progression  Arthralgy, bursitis with noduli rheumatici, conjunctivitis  43 M  132+  
145  MALT-type  N248K  Thyroid gland, regional lymph nodes  —  Diagnosis  Myxoedematous symptoms before lymphoma, elevated TSH  93  F  17  
146 MALT-type  IVS8nt + 5g → a  Paranasal sinus, nasal cavity  Salivary gland, orbit  Progression  Recurrent pleural effusions. “Sjögren-like”  70  39+  
69  DLC-B  L199X  Lymph node  Lymph nodes  Progression  NA  70  F  1  
86  DLC-B D244V  Skin, lymph node  —  Diagnosis  None  90 F  3  
92  DLC-B  L208X  Thyroid gland  —  Diagnosis  Goiter Myxoedema, elevated TSH  82  F  30  
93  DLC-B  L262F  Tonsil  Lymph node  Diagnosis  None  80  M  30  
128  DLC-B L164F  Skeletal muscle  Pancreas, stomach, liver/spleen Diagnosis  Pancreatitis  60  F  15  
141  DLC-B K283N  Mediastinum (thymus)  —  Diagnosis Monoclonal serum IgM, neuropathy  54  M  1  
156 DLC-B  N248K  Skin  —  Diagnosis  Monoclonal serum IgGκ and IgMλ, neuropathy  55  F  144+ 
225  DLC-B  IVS7nt − 2a → g  Thyroid gland Stomach  Diagnosis  Hashimoto’s thyroiditis  82  6  
157  DLC-B (TRB)  T182I  Salivary gland  Lymph nodes  Diagnosis  SLE, Sjögren syndrome  67  6  
72  ALCL, null  P167L  Skin  Lymph nodes Diagnosis  NA  62  M  
Case No. Histology Fas MutationPrimary Localization Localization at ProgressionDisease Status at Biopsy Autoimmune/ Paraneoplastic Phenomena Age Sex Survival From Biopsy (mo)
193  B-CLL  A-1T3-150 Blood, bone marrow Skin  Progression  Maculo-papular skin rash  78  6  
17  FCC  E256K  Waldeyers’ ring, lymph nodes Parotoid gland, lymph nodes  Progression  None  43 M  48  
49  FCC + DLC-B (TRB)  598 Ins T  Spleen Lymph nodes  Diagnosis  NA  63  M  63  
67 MALT-type  IVS8nt + 5g → c  Orbit, bone marrow, liver/spleen  Skin, lymph nodes  Progression  Arthralgy, bursitis with noduli rheumatici, conjunctivitis  43 M  132+  
145  MALT-type  N248K  Thyroid gland, regional lymph nodes  —  Diagnosis  Myxoedematous symptoms before lymphoma, elevated TSH  93  F  17  
146 MALT-type  IVS8nt + 5g → a  Paranasal sinus, nasal cavity  Salivary gland, orbit  Progression  Recurrent pleural effusions. “Sjögren-like”  70  39+  
69  DLC-B  L199X  Lymph node  Lymph nodes  Progression  NA  70  F  1  
86  DLC-B D244V  Skin, lymph node  —  Diagnosis  None  90 F  3  
92  DLC-B  L208X  Thyroid gland  —  Diagnosis  Goiter Myxoedema, elevated TSH  82  F  30  
93  DLC-B  L262F  Tonsil  Lymph node  Diagnosis  None  80  M  30  
128  DLC-B L164F  Skeletal muscle  Pancreas, stomach, liver/spleen Diagnosis  Pancreatitis  60  F  15  
141  DLC-B K283N  Mediastinum (thymus)  —  Diagnosis Monoclonal serum IgM, neuropathy  54  M  1  
156 DLC-B  N248K  Skin  —  Diagnosis  Monoclonal serum IgGκ and IgMλ, neuropathy  55  F  144+ 
225  DLC-B  IVS7nt − 2a → g  Thyroid gland Stomach  Diagnosis  Hashimoto’s thyroiditis  82  6  
157  DLC-B (TRB)  T182I  Salivary gland  Lymph nodes  Diagnosis  SLE, Sjögren syndrome  67  6  
72  ALCL, null  P167L  Skin  Lymph nodes Diagnosis  NA  62  M  

Abbreviations: ALCL, anaplastic large cell lymphoma; FCC, follicle center cell lymphoma; NA, not available.

F3-150

Numbering refers to the last residue of the signal peptide.

All three thyroid lymphomas in this series harbored Fasmutations. One of these (no. 225) was preceded by 4 years of well-documented Hashimoto’s thyroiditis with high titers of anti-thyroid peroxidase (anti-TPO) antibodies, hypergammaglobulinemia, elevated thyroid-stimulating hormone (TSH), and myxoedema. This patient was treated with cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP), but relapsed with a gastric lymphoma of similar histology. A second case (no. 145), presenting with thyroid low-grade MALT-type lymphoma and involvement of neck lymph nodes, had myxoedematous symptoms before lymphoma and elevated TSH. The last patient (no. 92) with thyroid lymphoma had massive goiter, elevated TSH level, and myxoedema. In these latter two cases, thyroid antibodies had not been analyzed.

Another patient (no. 157) presented in 1969 with Sjögren’s syndrome verified by sialography and salivary gland biopsy. In 1976 she developed universal arthritis, peripheral neuropathy, thrombocytopenia with positive IgM-Rh factor, antinuclear antibodies (ANA), anti-dsDNA antibodies, and hypergammaglobulinemia, and a diagnosis of SLE was made. In 1984 a DLC-B lymphoma of TRB-type was diagnosed in the salivary gland and regional lymph nodes.

Among the remaining patients in whom somatic Fas mutations had been identified, six showed a number of paraneoplastic features that may be associated with autoreactivity. One patient (no. 67) had conjunctivitis and arthralgy at diagnosis and later developed bursitis with noduli rheumatici. Another patient (no. 146) had unexplained recurrent pleural effusions 3 years before the diagnosis of lymphoma. Two patients (nos. 141 and 156) exhibited monoclonal IgM and neuropathy; one (no. 128) had persistent abdominal pain that was diagnosed as severe pancreatitis; and one (no. 193) was an unusual case of B-CLL with neoplastic skin infiltrates and a long-lasting maculo-papular skin rash.

In the remaining 6 cases, features of autoreactive/paraneoplastic disease were either not present (3 cases; nos. 86, 17, and 93), or no information was available (3 cases; nos. 49, 72, and 69).

Previous loss of heterozygosity (LOH) and karyotypic studies have suggested that a putative tumor suppressor gene at chromosome 10q23-25 may be involved in the development of NHL.22 We have recently examined PTEN/MMAC1, a gene mapping to 10q23.3 and encoding a tumor suppressor commonly altered in many types of human cancer,23-25 but found that this gene is mutated in less than 2% of NHL.26 In the present study, we have systematically examined the Fas gene on 10q24.1 and documented somatic mutations in 16 of 150 NHLs (11%). These findings, together with the recent demonstration of a similar frequency of Fasmutations in multiple myeloma,27 suggest that Fasmutations may be involved in the development of different types of lymphoid malignancies. In our series of NHL, the highest frequency of Fas mutations (60%) was seen in low-grade MALT-type lymphomas. Most of the remaining lesions were DLC-B lymphomas with a remarkable preference for extranodal sites. The possibility that some of the latter cases could be transformed low-grade MALT lymphomas is a tempting assumption which, however, could not be further elucidated in this retrospective analysis.

Although functional studies have not yet been performed, most of the mutations identified in the present study are likely to disrupt or alter the normal structure and/or function of Fas. Six of the mutations are predicted to cause premature termination of protein synthesis, aberrant RNA splicing, or frameshifts, and hence resemble typical loss-of-function mutations. However, a previous study has shown that genetic defects resulting in the production of a truncated protein may be able to confer a dominant-negative effect.5 The remaining mutations were missense variants resulting in nonconservative amino acid substitutions. Four of six missense mutations (D244V, E256K, L262F, and K283N) within the region encoding the Fas death domain affected codons that are evolutionarily highly conserved.28Furthermore, alteration of codon 244 has been shown in one case of ALPS7 and has been shown to cause reduced self association and binding of the Fas death domain to FADD/MORT1, which is necessary for transmission of the apoptotic signal.29 Likewise, alterations of residue 369 in TNFR1, which is homologous to Fas residue 256, have been shown to be associated with abrogation of TNFR1-mediated cytotoxicity.30 The functional significance of missense mutations outside of the death domain remains unknown at this stage.

The pattern of Fas inactivation demonstrated in this study is very similar to that observed in ALPS patients.5-9 In NHL, missense mutations within the death-domain-encoding region ofFas were consistently associated with retention of the wild-type allele. This finding is in line with previous observations that ALPS patients carrying a death domain mutation are heterozygous,5-9 and substantiates the notion that nonconservative amino acid substitutions in the death domain may act in a dominant-negative fashion.5 In contrast, missense mutations outside the death domain were associated with loss of the wild-type allele, suggesting that a classical two-hit mode of gene inactivation may be necessary to disrupt gene function in these cases. Missense mutations outside the death domain have been reported in two ALPS families, and in both cases were they associated with the ALPS phenotype only in the presence of concomitant mutation of the second allele.8,31 

An intriguing finding in the present study was the high incidence of autoreactive phenomena in the group of NHL patients in whose tumors we had identified Fas mutations. Two cases had well-documented autoimmune diseases (SLE/Sjögren’s syndrome and Hashimoto’s thyroiditis, respectively), and eight cases showed various paraneoplastic features suggestive of autoreactivity, including bursitis with noduli rheumatici, conjunctivitis, neuropathy, pancreatitis, and myxoedema. Ninety percent of all myxoedemas are believed to be caused by autoimmune thyroiditis, and features of autoimmune thyroiditis have been identified by histology in 5 of 5 and 8 of 8 low-grade MALT lymphomas in two independent studies.32,33 Furthermore, examination of thyroid B-cell lymphomas of large cell type has shown that low-grade MALT-type components may be found consistently if multiple sections are examined.32 These observations have suggested that most, if not all, thyroid B-cell lymphomas develop through a step of autoimmune thyroid disease, and are MALT-type lymphomas with or without features of transformation.32 

Several lines of evidence have suggested a causative role forFas alterations in the induction of autoimmune disease. First, inherited defects in Fas in mice and humans result in lymphoproliferation and systemic autoimmunity caused by the massive accumulation of autoreactive B and T cells.4-6 Second, precursor B cells for autoantibody production in Fas/FasL-intact, SLE-prone mice are resistant to Fas-mediated apoptosis due to downregulation of their Fas expression.34 Third, induction of Fas expression has been associated with increased rates of FasL-induced apoptosis in insulin-dependent diabetes mellitus and Hashimoto’s thyroiditis.35,36 

The high incidence of autoimmune phenomena observed among NHL patients with Fas-mutated tumors suggests that somatic mutation ofFas may add to the above spectrum of mechanisms causing escape from self-tolerance. An anergic and potentially self-reactive B cell which acquires a Fas mutation may no longer be susceptible to apoptosis. Instead, it may be triggered by CD4+ T cells to proliferate,3 and eventually may result in the massive production of autoantibodies. The possible association between autoimmunity, Fas mutations and lymphomas warrants further study by examination of Fas mutations in lymphomas and matched autoimmune lesions.

The identification of Fas as a mediator of apoptosis in cells of the immune system led to the speculation that disruption of the normal Fas-mediated apoptotic pathway may represent an early event in lymphomagenesis, leading to longer lymphocyte survival and thus allowing for the accumulation of additional oncogenic events.37,38 This notion was reinforced by recent work by Plumas et al,39 who showed that malignant B cells from NHL exhibit intrinsic resistance to lysis mediated by FasL expressed on cytotoxic T cells. Interestingly, this resistance was not related to the levels of Fas expression and could not be overcome by induction of Fas expression. The data from the present study provide direct evidence that the Fas-mediated apoptotic pathway is abrogated in approximately 10% of NHL cases because of alteration or loss of Fas function. Whether alterations in the expression and/or function of components downstream of Fas in the same pathway, including FADD/MORT1,40,41 caspase 8,42,43 and FLICE-inhibitory proteins (FLIPs),44 cause resistance to Fas-mediated apoptosis in the remaining cases will be subject to future studies.

Supported by Grants from the Danish Cancer Society, the Ellen and Aage Fausbøll Foundation, the Arthur and Poula Søndergaard Foundation, and the Kaarsen Foundation.

Address reprint requests to Per Guldberg, PhD, Department of Tumor Cell Biology, Institute of Cancer Biology, Danish Cancer Society, Strandboulevarden 49, DK-2100 Copenhagen, Denmark; e-mail:perg@bio.cancer.dk.

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

1
Suda
T
Takahashi
T
Golstein
P
Nagata
S
Molecular cloning and expression of the Fas ligand, a novel member of the tumor necrosis factor family.
Cell
75
1993
1169
2
Nagata
S
Apoptosis by death factor.
Cell
88
1997
355
3
Rathmell
JC
Townsend
SE
Xu
JC
Flavell
RA
Goodnow
CC
Expansion or elimination of B cells in vivo: Dual roles for CD40-and Fas (CD95)-ligands modulated by the B cell antigen receptor.
Cell
87
1996
319
4
Watanabe-Fukunaga
R
Brannan
CI
Copeland
NG
Jenkins
NA
Nagata
S
Lymphoproliferation disorder in mice explained by defects in Fas antigen that mediates apoptosis.
Nature
356
1992
314
5
Fisher
GH
Rosenberg
FJ
Straus
SE
Dale
JK
Middleton
LA
Lin
AY
Strober
W
Lenardo
MJ
Puck
JM
Dominant interfering Fas gene mutations impair apoptosis in a human autoimmune lymphoproliferative syndrome.
Cell
81
1995
935
6
Rieux-Laucat
F
Le Deist
F
Hivroz
C
Roberts
IA
Debatin
KM
Fischer
A
de Villartay
JP
Mutations in Fas associated with human lymphoproliferative syndrome and autoimmunity.
Science
268
1995
1347
7
Drappa
J
Vaishnaw
AK
Sullivan
KE
Chu
JL
Elkon
KB
Fas gene mutations in the Canale-Smith syndrome, an inherited lymphoproliferative disorder associated with autoimmunity.
N Engl J Med
335
1996
1643
8
Bettinardi
A
Brugnoni
D
Quiros-Roldan
E
Malagoli
A
La Grutta
S
Correra
A
Notarangelo
LD
Missense mutations in the Fas gene resulting in autoimmune lymphoproliferative syndrome: A molecular and immunological analysis.
Blood
89
1997
902
9
Sneller
MC
Wang
J
Dale
JK
Strober
W
Middelton
LA
Choi
Y
Fleisher
TA
Lim
MS
Jaffe
ES
Puck
JM
Lenardo
MJ
Straus
SE
Clincial, immunologic, and genetic features of an autoimmune lymphoproliferative syndrome associated with abnormal lymphocyte apoptosis.
Blood
89
1997
1341
10
Harris
NL
Jaffe
ES
Stein
H
Banks
PM
Chan
JKC
Cleary
ML
Delsol
G
De Wolf-Peeters
C
Falini
B
Gatter
KC
Grogan
TM
Isaacson
PG
Knowles
DM
Mason
DY
Muller-Hermelink
HK
Pileri
SA
Piris
MA
Ralfkiaer
E
Warnke
RA
A revised European-American classification of lymphoid neoplasms: A proposal from the international lymphoma study group.
Blood
84
1994
1361
11
Polliack
A
Lugassy
G
Autoimmunity and auto-immune syndromes associated with and preceding the development of lymphoproliferative disorders.
Leukemia
6
1992
152
(suppl 4)
12
Kinlen
LJ
Malignancy in autoimmune diseases.
J Autoimmun
5
1992
363
(suppl A)
13
Mellemkjaer
L
Andersen
V
Linet
MS
Gridley
G
Hoover
R
Olsen
JH
Non-Hodgkin’s lymphoma and other cancers among a cohort of patients with systemic lupus erythematosus.
Arthritis Rheum
40
1997
761
14
Grønbæk
K
D’Amore
F
Schmidt
K
Autoimmune phenomena in non-Hodgkin’s lymphoma.
Leuk Lymphoma
18
1995
311
15
Abrams
ES
Stanton
VP
Use of denaturing gradient gel electrophoresis to study conformational transitions in nucleic acids.
Methods Enzymol
212
1992
71
16
Lerman
LS
Silverstein
K
Computational simulation of DNA melting and its application to denaturing gradient gel electrophoresis.
Methods Enzymol
155
1987
482
17
Sheffield
VC
Cox
DR
Lerman
LS
Myers
RM
Attachment of a 40-base-pair G+C-rich sequence (GC-clamp) to genomic DNA fragments by the polymerase chain reaction in improved detection of single-base changes.
Proc Natl Acad Sci USA
86
1989
232
18
Cremonesi
L
Firpo
S
Ferrari
M
Righetti
PG
Gelfi
C
Double-gradient DGGE for optimized detection of DNA point mutations.
Biotechniques
22
1997
326
19
Huang
QR
Morris
D
Manolios
N
Identification and characterization of polymorphisms in the promoter region of the human Apo-1/Fas (CD95) gene.
Mol Immunol
34
1997
577
20
Fiucci
G
Ruberti
G
Detection of polymorphisms within the Fas cDNA gene sequence by GC-clamp denaturing gradient gel electrophoresis.
Immunogenetics
39
1994
437
21
Krawczak
M
Reiss
J
Cooper
DN
The mutational spectrum of single base-pair substitutions in mRNA splice junctions of human genes: Causes and consequences.
Hum Genet
90
1992
41
22
Speaks
SL
Sanger
WG
Masih
AS
Harrington
DS
Hess
M
Armitage
JO
Recurrent abnormalities of chromosome bands 10q23-25 in non-Hodgkin’s lymphoma.
Genes Chromosomes Cancer
5
1992
239
23
Steck
PA
Pershouse
MA
Jasser
SA
Yung
WK
Lin
H
Ligon
AH
Langford
LA
Baumgard
ML
Hattier
T
Davis
T
Frye
C
Hu
R
Swedlund
B
Teng
DH
Tavtigian
SV
Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers.
Nat Genet
15
1997
356
24
Li
J
Yen
C
Liaw
D
Podsypanina
K
Bose
S
Wang
SI
Puc
J
Miliaresis
C
Rodgers
L
McCombie
R
Bigner
SH
Giovanella
BC
Ittmann
M
Tycko
B
Hibshoosh
H
Wigler
MH
Parsons
R
PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer.
Science
275
1997
1943
25
Teng
DH
Hu
R
Lin
H
Davis
T
Iliev
D
Frye
C
Swedlund
B
Hansen
KL
Vinson
VL
Gumpper
KL
Ellis
L
El-Naggar
A
Frazier
M
Jasser
S
Langford
LA
Lee
J
Mills
GB
Pershouse
MA
Pollack
RE
Tornos
C
Troncoso
P
Yung
WK
Fujii
G
Berson
A
Steck
PA
MMAC1/PTEN mutations in primary tumor specimens and tumor cell lines.
Cancer Res
57
1997
5221
26
Grønbæk
K
Ralfkiær
E
Zeuthen
J
Hou-Jensen
K
Guldberg
P
Alterations of the MMAC1/PTEN gene in lymphoid malignancies.
Blood
91
1998
4388
27
Landowski
TH
Qu
N
Buyuksal
I
Painter
JS
Dalton
WS
Mutations in the Fas antigen in patients with multiple myeloma.
Blood
90
1997
4266
28
Itoh
N
Nagata
S
A novel protein domain required for apoptosis. Mutational analysis of human Fas antigen.
J Biol Chem
268
1993
10932
29
Huang
B
Eberstadt
M
Olejniczak
ET
Meadows
RP
Fesik
SW
NMR structure and mutagenesis of the Fas (APO-1/CD95) death domain.
Nature
384
1996
638
30
Loetscher
H
Pan
YC
Lahm
HW
Gentz
R
Brockhaus
M
Tabuchi
H
Lesslauer
W
Molecular cloning and expression of the human 55 kd tumor necrosis factor receptor.
Cell
61
1990
351
31
Pensati
L
Costanzo
A
Ianni
A
Accapezzato
D
Iorio
R
Natoli
G
Nisini
R
Almerighi
C
Balsano
C
Vajro
P
Vegnente
A
Levrero
M
Fas/Apo1 mutations and autoimmune lymphoproliferative syndrome in a patient with type 2 autoimmune hepatitis.
Gastroenterology
113
1997
1384
32
Hyjek
E
Isaacson
PG
Primary B cell lymphoma of the thyroid and its relationship to Hashimoto’s thyroiditis.
Hum Pathol
19
1988
1315
33
Pedersen
RK
Pedersen
NT
Primary non-Hodgkin’s lymphoma of the thyroid gland: A population based study.
Histopathology
28
1996
25
34
Hirose
S
Yan
K
Abe
M
Jiang
Y
Hamano
Y
Tsurui
H
Shirai
T
Precursor B cells for autoantibody production in genomically Fas-intact autoimmune disease are not subject to Fas-mediated immune elimination.
Proc Natl Acad Sci USA
94
1997
9291
35
Chervonsky
AV
Wang
Y
Wong
FS
Visintin
I
Flavell
RA
Janeway
CAJ
Matis
LA
The role of Fas in autoimmune diabetes.
Cell
89
1997
17
36
Giordano
C
Stassi
G
De Maria
R
Todaro
M
Richiusa
P
Papoff
G
Ruberti
G
Bagnasco
M
Testi
R
Galluzzo
A
Potential involvement of Fas and its ligand in the pathogenesis of Hashimoto’s thyroiditis.
Science
275
1997
960
37
Trauth
BC
Klas
C
Peters
AM
Matzku
S
Moller
P
Falk
W
Debatin
KM
Krammer
PH
Monoclonal antibody-mediated tumor regression by induction of apoptosis.
Science
245
1989
301
38
Falk
MH
Trauth
BC
Debatin
KM
Klas
C
Gregory
CD
Rickinson
AB
Calender
A
Lenoir
GM
Ellwart
JW
Krammer
PH
Expression of the APO-1 antigen in Burkitt lymphoma cell lines correlates with a shift towards a lymphoblastoid phenotype.
Blood
79
1992
3300
39
Plumas
J
Jacob
MC
Chaperot
L
Molens
JP
Sotto
JJ
Bensa
JC
Tumor B cells from non-Hodgkin’s lymphoma are resistant to CD95 (Fas/Apo-1)-mediated apoptosis.
Blood
91
1998
2875
40
Chinnaiyan
AM
O’Rourke
K
Tewari
M
Dixit
VM
FADD, a novel death domain-containing protein, interacts with the death domain of Fas and initiates apoptosis.
Cell
81
1995
505
41
Boldin
MP
Varfolomeev
EE
Pancer
Z
Mett
IL
Camonis
JH
Wallach
D
A novel protein that interacts with the death domain of Fas/APO1 contains a sequence motif related to the death domain.
J Biol Chem
270
1995
7795
42
Muzio
M
Chinnaiyan
AM
Kischkel
FC
O’Rourke
K
Shevchenko
A
Ni
J
Scaffidi
C
Bretz
JD
Zhang
M
Gentz
R
Mann
M
Krammer
PH
Peter
ME
Dixit
VM
FLICE, a novel FADD-homologous ICE/CED-3-like protease, is recruited to the CD95 (Fas/APO-1) death-inducing signaling complex.
Cell
85
1996
817
43
Boldin
MP
Goncharov
TM
Goltsev
YV
Wallach
D
Involvement of MACH, a novel MORT1/FADD-interacting protease, in Fas/APO-1-and TNF receptor-induced cell death.
Cell
85
1996
803
44
Thome
M
Schneider
P
Hofmann
K
Fickenscher
H
Meinl
E
Neipel
F
Mattmann
C
Burns
K
Bodmer
JL
Schroter
M
Scaffidi
C
Krammer
PH
Peter
ME
Tschopp
J
Viral FLICE-inhibitory proteins (FLIPs) prevent apoptosis induced by death receptors.
Nature
386
1997
517
Sign in via your Institution