Skip to main content
Erschienen in: BMC Cancer 1/2014

Open Access 01.12.2014 | Research article

Family-specific, novel, deleterious germline variants provide a rich resource to identify genetic predispositions for BRCAx familial breast cancer

verfasst von: Hongxiu Wen, Yeong C Kim, Carrie Snyder, Fengxia Xiao, Elizabeth A Fleissner, Dina Becirovic, Jiangtao Luo, Bradley Downs, Simon Sherman, Kenneth H Cowan, Henry T Lynch, San Ming Wang

Erschienen in: BMC Cancer | Ausgabe 1/2014

Abstract

Background

Genetic predisposition is the primary risk factor for familial breast cancer. For the majority of familial breast cancer, however, the genetic predispositions remain unknown. All newly identified predispositions occur rarely in disease population, and the unknown genetic predispositions are estimated to reach up to total thousands. Family unit is the basic structure of genetics. Because it is an autosomal dominant disease, individuals with a history of familial breast cancer must carry the same genetic predisposition across generations. Therefore, focusing on the cases in lineages of familial breast cancer, rather than pooled cases in disease population, is expected to provide high probability to identify the genetic predisposition for each family.

Methods

In this study, we tested genetic predispositions by analyzing the family-specific variants in familial breast cancer. Using exome sequencing, we analyzed three families and 22 probands with BRCAx (BRCA-negative) familial breast cancer.

Results

We observed the presence of family-specific, novel, deleterious germline variants in each family. Of the germline variants identified, many were shared between the disease-affected family members of the same family but not found in different families, which have their own specific variants. Certain variants are putative deleterious genetic predispositions damaging functionally important genes involved in DNA replication and damaging repair, tumor suppression, signal transduction, and phosphorylation.

Conclusions

Our study demonstrates that the predispositions for many BRCAx familial breast cancer families can lie in each disease family. The application of a family-focused approach has the potential to detect many new predispositions.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-14-470) contains supplementary material, which is available to authorized users.
Hongxiu Wen, Yeong C Kim contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

FX, HW, BD performed experiments. YK performed bioinformatics data analysis. CS, DB performed pedigree analysis, identified the study subjects, and prepared DNA samples. JL performed statistical analysis. EAF, SS, KC developed the UNMC Breast Cancer Collaborative Register used in the study [43]. HL and SMW conceived the study. SMW designed the experiment and wrote the paper. All authors read and approved the final manuscript.
Abkürzungen
BRCAx
Familial breast cancer without known mutations in BRCA1 and BRCA2
Proband
the first affected family member seeking medical attention
Exome sequencing
Sequencing the entire coding region in a genome using the next generation DNA sequencing technology
SAM
Sequence Alignment/Map format used for storing sequence data in a series of tab delimited ASCII columns
BAM
A binary format for storing sequence data in a compressed, indexed, binary form
GATK
Genome Analysis Toolkit. It is a software package to analyse next-generation resequencing data
VarScan 2
a software package to detect variants in next-generation resequencing data
PolyPhen-2
a software to predict possible impact of an amino acid substitution on the structure and function of a protein
Primer3
a software for designing PCR primers
NCBI
The National Center for Biotechnology Information
dbSNP
Single Nucleotide Polymorphism Database
ESP
Exome Sequencing Project
MAF
Minor Allele Frequency.

Background

Breast cancer is a leading cancer in women [1]. About 10-20% of breast cancer cases are family clustered, with multiple family members affected by the disease [2]. Genetic predispositions are the major risk factor for the disease. However, the genetic predispositions are currently known for only 30-40% of the familial breast cancer disease families. The remaining 60-70% of women with familial breast cancer have unknown predispositions and are diagnosed with BRCAx, for their unknown predisposition of familial breast cancer [3]. It is estimated the “missing” heredity trait for BRCAx families likely consists of thousands of rare variants, each presenting a minor disease risk [4]. Indeed, broadly screening the variants across disease populations has uncovered multiple new genetic predispositions for familial breast cancer. A consistent pattern among these newly classified predispositions is that they are always present at very-low frequencies in the given disease population [510]. Their extreme rarity implies that a greater sampling size of disease populations is required to identify the germline predispositions [10]. However, such an expansion is deemed to increase the complexity of data analysis, experimental costs, and time needed. As such, focusing only on the rare variants will not likely be able to determine the entire spectrum of genetic predispositions for BRCAx familial breast cancer families. New alternative hypotheses and approaches must be explored to improve the situation. For example, mosaic mutation has implications as potential predispositions for familial breast cancer [11].
Familial breast cancer is defined as an autosomal dominant genetic disease [12]. Although incidences of breast cancer often exhibit atypical Mendelian patterns due to the factors such as low penetrance of genetic predispositions, the predisposition in a disease-prone family is expected to transmit across generations and shared between family members. Focusing on each disease family with a history of the disease is expected to improve the chance to detect the predisposition in a family compared to screening the disease population of pooled cases without family relationships, which can dilute the predisposition highly prevalent in a disease family into insignificant level.
We hypothesize that the unknown predispositions for many BRCAx familial breast cancer are specific to each family with a history of the disease. Our previous exome study of a BRCAx familial breast cancer family shows the presence of rich genetic variants [13]. In the present study, we expand the exome sequencing study by analyzing three families with BRCAx familial breast cancer; 17 members had cancer, and five members were without cancer. Our study also includes 22 probands of BRCAx familial breast cancer. Our study reveals the presence of family-specific, novel, deleterious genetic variants as putative genetic predispositions in each family with BRCAx familial breast cancer.

Methods

Use of human subjects

The use of the patient samples for the study was approved by the Institutional Review Boards (IRB) of Creighton University School of Medicine (#00-12265 ) and University of Nebraska Medical Center (718-11-EP). All subjects signed the Consent to Participate Form for cancer genetic study.
Individuals from three families with BRCAx breast cancer were used to generate exome sequences as we have previously described [13]. Family I included six individuals with breast cancer and two individuals without breast cancer. Family II included five individuals with breast cancer, one obligate carrier and two individuals without breast cancer. Family III included five individuals with breast cancer and one individual without breast cancer. Additionally, 22 probands for BRCAx familial breast cancer were included in exome sequencing. All cases used in the study were BRCA1-negative, and BRCA2-negative, 41 were female and 3 were male, the average age is 42 years old (Figure 1, Table 1).
Table 1
BRCAx familial breast cancer cases used in the study
Family
Cancer type
Pathology
BRCA1/2
Exome
    
Reads
Bases
Bases map rate (%)
Coverage
Variant called
Family 1
        
 1
Breast
Infiltrating ductal
-
42,973,730
4,340,346,730
97.6
70
184,865
 2
Breast
Not available
-
40,158,059
4,055,963,959
98.3
65
152,692
 3
Breast
Infiltrating ductal
-
46,240,754
4,670,316,154
97.2
75
176,554
 4
Prostate
Adenocarcinoma
-
23,418,595
2,365,278,095
98.1
38
207,103
 5
No Cancer
 
-
40,313,161
4,071,629,261
98.0
66
213,347
 6
Breast, Colon
Adenocarcinoma
-
17,496,012
1,767,097,212
97.9
28
183,741
 7
Brain
Not available
-
36,166,319
3,652,798,219
99.5
59
171,425
 8
Breast
Adenocarcinoma
-
27,830,687
2,810,899,387
96.3
45
104,343
Family 2
        
 1
Breast, Breast
Medullary, infiltrating ductal
-
33,419,098
3,375,328,898
92.9
54
113,079
 2
Obligated carrier
 
-
27,261,117
2,753,372,817
92.4
44
115,328
 3
Breast
Infiltrating ductal
-
40,973,473
4,138,320,773
99.6
67
127,272
 4
Breast
Ductal carcinoma in situ
-
29,561,523
2,985,713,823
91.5
48
108,655
 5
Breast
Infiltrating ductal
-
25,790,969
2,604,887,869
93.1
42
84,687
 6
Breast
Infiltrating ductal
-
37,657,589
3,803,416,489
91.6
61
139,891
 7
No Cancer
 
-
17,433,912
1,760,825,112
91.6
28
131,786
 8
No Cancer
 
-
35,977,512
3,633,728,712
97.3
59
128,680
Family 3
        
 1
Endometrial
Adenocarcinoma
-
33,662,978
3,399,960,778
93.2
55
129,754
 2
Breast, Skin
Basal, infiltrating ductal
-
29,648,460
2,994,494,460
98.3
48
198,862
 3
No Cancer
 
-
53,411,156
5,394,526,756
98.8
87
193,017
 4
Breast
Infiltrating ductal
-
31,736,845
3,205,421,345
98.3
52
130,941
 5
Breast
Ductal carcinoma in situ
-
35,014,538
3,536,468,338
98.4
57
129,754
 6
Breast
Not available
-
38,418,769
3,880,295,669
97.5
62
161,953
Probands
        
 1
Breast
Ductal carcinoma in situ
-
17,832,681
1,801,100,781
93.1
29
109,864
 2
Breast
Invasive ductal carcinoma
-
36,166,319
3,652,798,219
99.5
59
142,155
 3
Breast
Invasive ductal carcinoma
-
50,944,516
5,145,396,116
98.4
83
152,125
 4
Breast
Invasive ductal carcinoma
-
43,889,986
4,432,888,586
99.6
71
169,633
 5
Breast
Invasive ductal carcinoma
-
40,125,408
4,052,666,208
99.5
65
153,511
 6
Breast
Invasive lobular carcinoma
-
31,798,628
3,211,661,428
97.5
52
119,875
 7
Breast
Invasive ductal carcinoma
-
49,739,415
5,023,680,915
99.6
81
113,058
 8
Breast
Invasive ductal carcinoma
-
63,352,269
6,398,579,169
99.6
103
99,732
 9
Breast
Invasive ductal carcinoma
-
43,744,840
4,418,228,840
99.5
71
149,873
 10
Breast
Invasive ductal carcinoma
-
43,573,311
4,400,904,411
99.6
71
141,236
 11
Breast
Invasive ductal carcinoma
-
40,938,838
4,134,822,638
99.3
67
143,262
 12
Breast
Ductal carcinoma in situ
-
36,258,870
3,662,145,870
99.6
59
138,018
 13
Breast
Ductal carcinoma in situ
-
34,550,745
3,489,625,245
99.4
56
146,858
 14
Breast
Invasive ductal carcinoma
-
50,295,200
5,079,815,200
99.5
82
156,666
 15
Breast
Invasive ductal carcinoma
-
60,736,566
6,134,393,166
99.7
99
115,909
 16
Breast
Invasive ductal carcinoma
-
57,383,360
5,795,719,360
99.6
93
120,945
 17
Breast
Invasive ductal carcinoma
-
44,922,611
4,537,183,711
99.6
73
110,503
 18
Breast
Invasive ductal carcinoma
-
33,883,509
3,422,234,409
99.4
55
131,955
 19
Breast
Invasive ductal carcinoma
-
49,729,619
5,022,691,519
99.5
81
146,665
 20
Breast
Invasive ductal carcinoma
-
63,184,143
6,381,598,443
99.6
103
119,680
 21
Breast
Invasive ductal carcinoma
-
28,002,381
2,828,240,481
99.6
46
86,924
 22
Breast
Invasive ductal carcinoma
-
47,794,798
4,827,274,598
99.5
78
112,030
Average
38,941,211
3,933,062,277
97.7
63
140,187
   

Exome sequencing

For each sample, exome sequencing used DNA from blood cells. Exome libraries were constructed using the TruSeq Exome Enrichment Kit (62 Mb, Illumina, San Diego, CA) as per manufacturer’s procedures. Exome sequences were collected with a HiSeq™ 2000 sequencer (Illumina, San Diego, CA) with paired-end (2 × 100). All exome data were deposited in the Sequence Read Archive (SRA) database in the National Center for Biotechnology Information (NCBI) (Accession numbers SAMN02404413- SAMN02404456).

Exome sequence mapping and variant calling

Exome sequences were mapped to the human genome reference sequence hg19 by Bowtie2 with default parameters in paired mode [14]. The subsequent SAM files were converted to BAM files. Duplicates were removed using Picard (http://​picard.​sourceforge.​net). The mapped reads were locally realigned using the genome mapping tool RealignerTargetCreator from the Genome Atlas Tool Kit (GATK) [15]. The base quality scores were recalibrated using BaseRecalibrator (GATK), with NCBI dbSNP build 137, in the GATK resource bundles for reference sequence hg19. VarScan 2 was used for variant calling, [16]. VarScan 2 was run on pileup data generated from BAM files using SAMtools utilities [17]. The mpileup command, with –B parameter to disable base alignment quality (BAQ) computation, and the default parameters were used, with the minimum read depth at 10 and the minimum base quality at 30. The called variants were annotated with ANNOVAR using the software-provided databases of the Reference Sequence (RefSeq; NCBI), dbSNP 137, the 1000 Genomes Project, and the NIH Heart, Lung and Blood Institute (NHLBI) Exome Sequencing Project (ESP) 6500 (http://​evs.​gs.​washington.​edu).
Those that matched in the databases were classified as known variants and removed. Family-specific normal variants were eliminated by removing the variants shared between the affected and the unaffected family members in each family. The remaining novel variants were classified into synonymous, non-synonymous, splicing site change, stop gain- or loss groups. The variants causing synonymous changes were then removed. For the remaining variants, PolyPhen-2 was used to identify variants causing deleterious effects in the affected genes [probably damaging score: 0.909-1; possibly damaging score: 0.447 - 0.908; Benign score: 0 - 0.446; HumVar score: [18]. The variants defined as benign were removed. These processes generated a list of novel, deleterious variants only present in the cancer-affected family members and probands, Note that the variants in probands were filtered by population databases only.

Power calculation

Using a two-sided paired t-test and assuming a genetic relative risk (GRR) equal to 5.8, disease prevalence equal to 0.03, a disease locus frequency equal to 0.01, and a sib recurrence ratio of 2, a sample size of 20 achieves 81% power to detect a mutation difference with a (standardized) effect size of 0.67 between the affected member and the unaffected member. The significance level (alpha) is, in turn, 0.05 [19, 20].

Validation

Sanger sequencing was used to validate deleterious variants. Sense and antisense PCR primers for each selected variant were designed using the Primer3 program. The original DNA samples that were used in exome sequencing were served as PCR templates. PCR amplicons were subjected to BigDye sequencing. The resulting sequences were evaluated using CLC Genomics Workbench Program (Cambridge, MA) to confirm the variants called from exome sequences.

Results

Mapping exome data and calling variants

Exome sequences were collected via a blood sample from each study participant and mapped to the human genome reference sequence hg19. Variants were called from the mapping data. We focused on single-base, non-synonymous variants that affect protein coding, splicing, and stop gain- or loss mutations, which are reliably detectable by exome analysis [21]. The average exome coverage was 63x, and the average number of variants called was 140,187 per case (Table 1).
To increase the likelihood that the variants identified in the breast cancer-affected family members are breast cancer-associated, variants in each data set were filtered by: 1) removal of common variants present in human populations. All variants matching to population-derived variant databases (i.e., dbSNP137, ESP6500, and 1000 genomes) were removed; 2) Removal of family-specific normal variants. For the three families in the study, the variants shared between the affected and the unaffected members in the same family were removed. To identify those causing deleterious effects in the affected genes, the remaining variants were analyzed using the Polyphen-2 Program [18]. A total of 337 novel, deleterious variants present only in the affected members of Families I, II, and III were identified at an average of 112 variants per family (Table 2, Additional files 1: Table S1A, B, C); 689 novel, deleterious variants were identified in the 22 probands at an average of 30 variants per proband (Table 2, Additional files 2: Table S2A, B). Sanger sequencing validated the mapped variants at a validation rate of 83% (53/64), highlighting the reliability of the variants identified by exome mapping analysis (Additional file 1: Table S1D).
Table 2
Novel, deleterious variants detected in breast cancer-affected cases*
Family
Total (%)
Individual (%)
Shared**(%)
Family 1
   
 1
37
35
2
 2
26
26
0
 3
25
15
10
 4
48
39
9
 6
29
17
12
 7
12
6
6
 8
14
6
8
Subtotal
143 (199)
123 (86)
20 (14)
Family 2
   
 1
22
13
9
 2
15
5
10
 3
21
9
12
 4
21
12
9
 5
16
8
8
 6
8
2
6
Subtotal
66 (100)
47 (71)
19 (29)
Family 3
   
 1
39
13
26
 2
48
27
21
 4
21
12
9
 5
32
12
20
 6
41
19
22
Subtotal
128 (100)
83 (65)
45 (35)
Total
337 (100)
253 (75)
84 (25)
Probands
   
 1
35
10
25
 2
58
22
36
 3
74
28
46
 4
77
49
28
 5
70
28
42
 6
41
16
25
 7
31
24
7
 8
43
27
16
 9
51
19
32
 10
61
30
31
 11
70
35
35
 12
51
12
39
 13
55
15
40
 14
60
30
30
 15
51
31
20
 16
41
31
10
 17
32
18
14
 18
57
25
32
 19
58
18
40
 20
47
23
24
 21
33
25
8
 22
34
22
12
Total
689 (100)
568 (82)
121 (18)
Per proband
30
26
6
*The counts in subtotal and total are the unique number of variants.
**Shared with family members in the families, or shared with other probands.

Novel deleterious variants are mostly family-specific

We compared the variants within each family. We observed that 25% of the variants on average (14% in Family I, 29% in Family II, 35% in Family III) were shared in multiple affected members in each family, whereas 75% on average (86% in Family I, 71% in Family II and 65% in Family III) were present only in single affected member in each family (Table 2). We then compared the shared variants between the three families, and found only 1 variant was shared between Family I and Family II, four variants were shared between Family I and Family III (Figure 2A). For the 689 variants identified in the probands, 82% were proband-specific, and only 18% were shared between probands at various frequencies (Figure 2B, Additional file 2: Table S2A, S2B). The results indicate that the majority of the novel, deleterious variants identified in the three families and probands are family-specific, i.e., present only in each family but not shared with other families.

Identification of putative genetic predispositions

We analyzed the shared mutations between the affected members of the same family, the functional class of the mutated genes, and existing evidence for their contribution to cancer. In doing so, we identified the variants as the putative predispositions in Family I, II, and III, and probands (Table 3, Additional file 1: Table S1A, S1B, S1C). For Family I, this was the PTEN-Induced Putative Kinase 1 (PINK1); for Family II, these were Lysine (K) Acetyltransferase 6B (KAT6B) and Neurogenic Locus Notch Homolog Protein 2 (NOTCH2); and for Family III, this was Phosphorylase Kinase Beta (PHKB).
Table 3
Putative predispositions in familial breast cancer families and probands
 
Gene
Description
Position
Nucleotide
Amino acid
Type
PolyPhen2*
Cancer-affected member
Frequency
       
Score prediction
        
Family 1
        
1
2
3
4
6
7
8
 
 
GPRIN1
G protein regulated inducer of neurite outgrowth 1
chr5:176026123
c.T713C
p.L238S
Exonic
0.91
D
-
+
+
+
+
+
-
5
 
PINK1
PTEN induced putative kinase 1
chr1:20972051
c.960-2A > G
 
Splicing
NA
NA
-
-
+
+
-
-
-
2
 
POLK
Polymerase (DNA directed) kappa
chr5:74892737
c.A2219G
p.H740R
Exonic
0.62
P
-
-
-
+
-
-
-
1
Family 2
        
1
2
3
4
5
6
  
 
KAT6B
K(lysine) acetyltransferase 6B
chr10:76789128
c.G4546T
p.D1516Y
Exonic
0.95
D
-
+
+
+
+
+
 
5
 
KAT6B
K(lysine) acetyltransferase 6B
chr10:76789311
c.C4729T
p.R1577C
Exonic
0.96
D
-
+
+
+
+
+
 
5
 
NOTCH2
Notch 2
chr1:120459167
c.C6178T
p.R2060C
Exonic
0.99
D
-
-
+
-
-
+
 
2
Family 3
        
1
2
4
5
6
   
 
NANP
N-acetylneuraminic acid phosphatase
chr20:25596725
c.A583G
p.I195V
Exonic
0.98
D
+
-
+
-
-
  
2
 
PHKB
phosphorylase kinase, beta
chr16:47628126
c.1204 + 1G > T
 
Splicing
NA
NA
-
+
-
+
-
  
2
Proband
                
1
JAKMIP3
Janus kinase and microtubule interacting protein 3
chr10:133955524
c.G1574C
p.G525A
Exonic
1.00
D
        
2
POLQ
Polymerase (DNA directed), theta
chr3:121207798
c.A3980C
p.Q1327P
Exonic
1.00
D
        
3
DUX2
Double homeobox 2
chr10:135494906
  
Splicing
NA
NA
        
4
UBE2L3
Ubiquitin-conjugating enzyme E2L 3
chr22:21975938
c.G349A
p.E117K
Exonic
0.96
D
.
.
      
5
RAD23B
RAD23 homolog B (S. cerevisiae)
chr9:110087260
c.C1028T
p.P343L
Exonic
0.99
D
.
.
      
7
GATA3
GATA binding protein 3
chr10:8100630
c.C604T
p.R202C
Exonic
0.92
D
        
8
KAT6B
K(lysine) acetyltransferase 6B
chr10:76744854
c.G2390A
p.S797N
Exonic
0.98
D
        
9
LIG1
Ligase I, DNA, ATP-dependent
chr19:48637322
c.G1525A
p.E509K
Exonic
0.95
D
.
.
      
10
LIG4
Ligase IV, DNA, ATP-dependent
chr13:108862463
c.G1154A
p.R385K
Exonic
1.00
D
        
14
NOTCH2
Notch 2
chr1:120529603
c.G854A
p.R285H
Exonic
1.00
D
        
15
ABL1
c-abl oncogene 1, non-receptor tyrosine kinase
chr9:133729493
c.G122A
p.G41D
Exonic
0.92
D
        
16
TNK2
Tyrosine kinase, non-receptor, 2
chr3:195596385
c.C1760T
p.P587L
Exonic
1.00
D
        
17
NFRKB
Nuclear factor related to kappaB binding protein
chr11:129755398
c.G611A
p.R204H
Exonic
1.00
D
        
18
NFKBIZ
Nuclear factor of kappa light polypeptide gene enhancer
chr3:101576029
  
Splicing
NA
NA
        
19
SMG1
SMG1 phosphatidylinositol 3-kinase-related kinase
chr16:18879624
c.C3083T
p.T1028M
Exonic
0.99
D
        
20
PRKCQ
Protein kinase C, theta
chr10:6528042
c.G855C
p.Q285H
Exonic
1.00
D
        
21
ADRA2A
Adrenoceptor alpha 2A
chr10:112838117
c.C363G
p.C121W
Exonic
1.00
D
        
22
PPFIA4
Protein tyrosine phosphatase, receptor type
chr1:203025582
c.C668T
p.T223M
Exonic
0.92
D
        
D: Probably damaging (score: 0.909-1); P: Possibly damaging (score: 0.447 - 0.908).
PINK1 is a mitochondrial serine/threonine-protein kinase. Mutation in PINK1 causes autosomal recessive Parkinson’s disease [22]. KAT6B is a histone acetyl transferase involved in DNA replication, gene expression and regulation, and epigenetic modification of chromosomal structure [23]. Mutations in KAT6B cause multiple neurological diseases [24]. NOTCH2 is a member of the Notch family involved in controlling cell fate decision. Low Notch activity leads to hyperproliferative activity in breast cancer [25] and mutation in NOTCH2 causes Hajdu-Cheney syndrome [26]. PHKB regulates the function of phosphorylase kinase [27]. Mutation in PHKB causes glycogen storage disease type 9B [28]. Interestingly, a variant in Polymerase (DNA-Directed) Kappa (POLK) was present in Family I member #4. POLK is a member of Y family DNA polymerases, and functions by repairing the replication fork passing through DNA lesions [29]. Although we are not able to validate it due to the lack of DNA from the subject’s parents, it raises a possibility that this variant could be a de novo mutation in this individual. Multiple transcriptional factors were also affected by the mutations in each family. For example, the following transcriptional factors were mutated in Family I: ZNF335, LRRC66, ZNF417, ZNF587, GTF2I, ZFAND4, EIF4G2, GZF1, CCDC86, ZSCAN18, ZNF546, TAF1L, and LRIG3 (Additional file 1: Table S1A).
The variant data from probands show similar patterns as those of the three families (Table 3). In the 22 probands, four carried variants affecting the genes involved in DNA replication and damaging repair. Those include Polymerase (DNA-directed) Theta (POLQ) in Proband #2, RAD23 Homolog B (S. cerevisiae) (RAD23B) in Proband #3, Ligase I DNA, ATP-dependent (LIG1) in Proband #9, and Ligase IV DNA, ATP-dependent (LIG4) in Proband #10. POLQ repairs the apurinic sites [30]. RAD23B plays a role in nucleotide excision repair [31]. LIG1 ligates nascent DNA of the lagging strand, and a mutation in LIG1 causes replication errors, genome instability, and cancer [32]. LIG4 catalyzes double-strand break repair by joining non-homologous ends, and mutation in LIG4 causes LIG4 syndrome [33]. Several variants are found in well-known oncogenes and tumor suppressor genes, such as GATA Binding Protein 3 (GATA3) in Proband #7 and Abelson Murine Leukemia Viral Oncogene Homolog 1 (ABL1) in Proband #18. GATA3 regulates luminal epithelial cell differentiation in the mammary gland [34, 35]. The abnormal expression of GATA3 causes luminal A-type breast cancer [3638]. ABL1 is a tyrosine kinase that controls cell differentiation and division. It is involved in (9, 22) translocation, forming BCR-ABL fusion gene in chronic myelogenous leukemia (CML) [39]. Several individual variants in different cases affect the same genes but at different positions. For example, in Proband #8, a variant in KAT6B (c.G1841A/p.S614N) affects the HAT domain at the N terminal, whereas two variants in KAT6B in Family II (c.G3997T/p.D1333Y and c.C4180T/p.R1394C) affect the Met-rich domain at the C-terminal. In Proband #14 and Family II, two different NOTCH2 variants (c.G854A/p.R285H, c.C6178T/p.R2060C) were present. Multiple variants affect the genes involved in phosphorylation. These include Tyrosine Kinase Non-Receptor 2 (TNK2) in proband #16, Phosphatidylinositol 3 Kinase-Related Kinase (SMG1) in Proband #19, Protein Kinase C Theta (PRKCQ) in Proband #20, and Protein Tyrosine Phosphatase, Receptor Type F (PPFIZ4) in Proband #22.
We also performed an analysis at the pathway level by annotating the mutation-affected genes in the three families using KEGG database (http://​www.​genome.​jp/​kegg/​pathway.​html). Certain mutations were identified to affect several functional pathways. For example, the genes mutated in Family I (ACADVL, AHCY, ALDOA, SGPL1, MAT1A, GALNT8, GGT1) are involved in metabolic pathways. The genes mutated in Family 2 (NOTCH2, DUSP16) are involved in Notch signaling pathway and MAPK signaling pathway; genes mutated in Family III (SLC9A1, ITGAX, ITGAD) are involved in regulation of actin cytoskeleton.

Discussion

The majority of families with familial breast cancer lack evidence for their genetic predispositions. Efforts in past decade have made slow progress in determining the unknown genetic predispositions. Currently, population-based approach is adapted as the major promising tool to reach the goal [40]. One weakness of this approach is that it can “dilute out the effects of a very strong association in a small subset of the study population” [41]. It requires a large-size disease population of over tens of thousands but the predispositions identified will likely remain very rare in the disease population. Due to the extreme rarity, such genetic predispositions are often difficult to confirm in different disease populations and to distinguish from normal polymorphisms [5, 10]. Our study observed the presence of family-specific, novel, deleterious variants, and putative predispositions in the families and probands analyzed. The information implies that, in addition to the population-based approach, a family-based approach provides another option to determine the genetic predisposition.
Based on the higher frequencies of well-known predispositions identified by traditional approaches, the rarity of the predispositions recently identified by population-based approach, and the presence of family-specific, novel, deleterious variants in disease families revealed in our study, we propose a model to explain the genetic predispositions in familial breast cancer (Figure 3). In this model, the predisposition in BRCA1 has the highest frequency in the familial breast cancer population, other known predispositions gradually decrease their frequencies to insignificant levels, and the predispositions for many BRCAx familial breast cancers are family-specific. The model explains the difficulty in using traditional and population-based approaches to determine the unknown predispositions, and highlights that applying family-focused approach will be able to determine the genetic predispositions for many BRCAx disease families. This model can be further tested in larger number of BRCAx familial breast cancer families.
Our study aimed to determine if there are germline mutations present, rather than reach for comprehensive coverage of germline mutations in each family. We achieved this by eliminating all variants matched in population-derived variant databases (i.e., dbSNP137, ESP6500, 1000 genomes) to maximally avoid the variants representing normal polymorphism. Inclusion of such variants as the predisposition candidates, even with the use of certain cut-off such as minor allele frequency (MAF) <0.01, can increase the sensitivity but decrease the specificity of the variants referred to as putative predispositions.
Assignment of a specific mutation as a true predisposition to a disease family requires solid phenotypic evidence from in vitro analysis, cell line tests, search of the literature, bioinformatics data analysis, and animal models. This is best evidenced by determining the BRCA1 germline mutations as genetic predispositions in breast cancer, in which the definitive conclusion for its contribution to breast cancer is based on the mouse models showing development of breast cancer with the germline mutated BRCA1[42]. Our current study aims to provide evidence that the BRCAx disease families are enriched with germline damaging mutations, such that focusing on each disease family will be required to determine the genetic predisposition in each family. Indeed, even under strict mapping conditions, large numbers of mutations have been detected in each disease family and probands. While the data provide rich resources to identify the true predisposition for the disease family, the data cannot be considered as true predisposition without further phenotypic and functional evidences.

Conclusions

Our study shows that genetic predispositions in many BRCAx familial breast cancer families can be family-specific.

Acknowledgments

The study was supported by a pilot grant from Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center (SMW), and a NIH grant 1R21CA180008 (SMW). The funding bodies play no roles in design, collection, analysis, and interpretation of data. We also wish to thank for Melody A. Montgomery at the UNMC Research Editorial Office for her professional assistance in editing this manuscript.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver ( https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​ ) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

FX, HW, BD performed experiments. YK performed bioinformatics data analysis. CS, DB performed pedigree analysis, identified the study subjects, and prepared DNA samples. JL performed statistical analysis. EAF, SS, KC developed the UNMC Breast Cancer Collaborative Register used in the study [43]. HL and SMW conceived the study. SMW designed the experiment and wrote the paper. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat American Cancer Society: Cancer Facts & Figures – 2013. 2013 American Cancer Society: Cancer Facts & Figures – 2013. 2013
2.
Zurück zum Zitat Rahman N, Stratton MR: The genetics of breast cancer susceptibility. Annu Rev Genet. 1998, 32: 95-121.CrossRefPubMed Rahman N, Stratton MR: The genetics of breast cancer susceptibility. Annu Rev Genet. 1998, 32: 95-121.CrossRefPubMed
3.
Zurück zum Zitat Stratton MR, Rahman N: The emerging landscape of breast cancer susceptibility. Nat Genet. 2008, 40: 17-22.CrossRefPubMed Stratton MR, Rahman N: The emerging landscape of breast cancer susceptibility. Nat Genet. 2008, 40: 17-22.CrossRefPubMed
4.
Zurück zum Zitat Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, Nik-Zainal S, Martin S, Varela I, Bignell GR, Yates LR, Papaemmanuil E, Beare D, Butler A, Cheverton A, Gamble J, Hinton J, Jia M, Jayakumar A, Jones D, Latimer C, Lau KW, McLaren S, McBride DJ, Menzies A, Mudie L, Raine K, Rad R, Chapman MS, Teague J, et al: The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012, 486: 400-404.PubMedPubMedCentral Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, Nik-Zainal S, Martin S, Varela I, Bignell GR, Yates LR, Papaemmanuil E, Beare D, Butler A, Cheverton A, Gamble J, Hinton J, Jia M, Jayakumar A, Jones D, Latimer C, Lau KW, McLaren S, McBride DJ, Menzies A, Mudie L, Raine K, Rad R, Chapman MS, Teague J, et al: The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012, 486: 400-404.PubMedPubMedCentral
5.
Zurück zum Zitat Park DJ, Lesueur F, Nguyen-Dumont T, Pertesi M, Odefrey F, Hammet F, Neuhausen SL, John EM, Andrulis IL, Terry MB, Daly M, Buys S, Le Calvez-Kelm F, Lonie A, Pope BJ, Tsimiklis H, Voegele C, Hilbers FM, Hoogerbrugge N, Barroso A, Osorio A, Giles GG, Devilee P, Benitez J, Hopper JL, Tavtigian SV, Goldgar DE, Southey MC, Breast Cancer Family Registry; Kathleen Cuningham Foundation Consortium for Research into Familial Breast Cancer: Rare mutations in XRCC2 increase the risk of breast cancer. Am J Hum Genet. 2012, 90: 734-739.CrossRefPubMedPubMedCentral Park DJ, Lesueur F, Nguyen-Dumont T, Pertesi M, Odefrey F, Hammet F, Neuhausen SL, John EM, Andrulis IL, Terry MB, Daly M, Buys S, Le Calvez-Kelm F, Lonie A, Pope BJ, Tsimiklis H, Voegele C, Hilbers FM, Hoogerbrugge N, Barroso A, Osorio A, Giles GG, Devilee P, Benitez J, Hopper JL, Tavtigian SV, Goldgar DE, Southey MC, Breast Cancer Family Registry; Kathleen Cuningham Foundation Consortium for Research into Familial Breast Cancer: Rare mutations in XRCC2 increase the risk of breast cancer. Am J Hum Genet. 2012, 90: 734-739.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Thompson ER, Doyle MA, Ryland GL, Rowley SM, Choong DY, Tothill RW, Thorne H, Barnes DR, Li J, Ellul J, Philip GK, Antill YC, James PA, Trainer AH, Mitchell G, Campbell IG, kConFab: Exome sequencing identifies rare deleterious mutations in DNA repair genes FANCC and BLM as potential breast cancer susceptibility alleles. PLoS Genet. 2012, 8: e1002894-CrossRefPubMedPubMedCentral Thompson ER, Doyle MA, Ryland GL, Rowley SM, Choong DY, Tothill RW, Thorne H, Barnes DR, Li J, Ellul J, Philip GK, Antill YC, James PA, Trainer AH, Mitchell G, Campbell IG, kConFab: Exome sequencing identifies rare deleterious mutations in DNA repair genes FANCC and BLM as potential breast cancer susceptibility alleles. PLoS Genet. 2012, 8: e1002894-CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Snape K, Ruark E, Tarpey P, Renwick A, Turnbull C, Seal S, Murray A, Hanks S, Douglas J, Stratton MR, Rahman N: Predisposition gene identification in common cancers by exome sequencing: insights from familial breast cancer. Breast Cancer Res Treat. 2012, 134: 429-433.CrossRefPubMedPubMedCentral Snape K, Ruark E, Tarpey P, Renwick A, Turnbull C, Seal S, Murray A, Hanks S, Douglas J, Stratton MR, Rahman N: Predisposition gene identification in common cancers by exome sequencing: insights from familial breast cancer. Breast Cancer Res Treat. 2012, 134: 429-433.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Gracia-Aznarez FJ, Fernandez V, Pita G, Peterlongo P, Dominguez O, de la Hoya M, Duran M, Osorio A, Moreno L, Gonzalez-Neira A, Rosa-Rosa JM, Sinilnikova O, Mazoyer S, Hopper J, Lazaro C, Southey M, Odefrey F, Manoukian S, Catucci I, Caldes T, Lynch HT, Hilbers FS, van Asperen CJ, Vasen HF, Goldgar D, Radice P, Devilee P, Benitez J: Whole exome sequencing suggests much of non-BRCA1/BRCA2 familial breast cancer is due to moderate and low penetrance susceptibility alleles. PLoS One. 2013, 8: e55681-CrossRefPubMedPubMedCentral Gracia-Aznarez FJ, Fernandez V, Pita G, Peterlongo P, Dominguez O, de la Hoya M, Duran M, Osorio A, Moreno L, Gonzalez-Neira A, Rosa-Rosa JM, Sinilnikova O, Mazoyer S, Hopper J, Lazaro C, Southey M, Odefrey F, Manoukian S, Catucci I, Caldes T, Lynch HT, Hilbers FS, van Asperen CJ, Vasen HF, Goldgar D, Radice P, Devilee P, Benitez J: Whole exome sequencing suggests much of non-BRCA1/BRCA2 familial breast cancer is due to moderate and low penetrance susceptibility alleles. PLoS One. 2013, 8: e55681-CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Hilbers FS, Meijers CM, Laros JF, van Galen M, Hoogerbrugge N, Vasen HF, Nederlof PM, Wijnen JT, van Asperen CJ, Devilee P: Exome sequencing of germline DNA from non-BRCA1/2 familial breast cancer cases selected on the basis of aCGH tumor profiling. PLoS One. 2013, 8: e55734-CrossRefPubMedPubMedCentral Hilbers FS, Meijers CM, Laros JF, van Galen M, Hoogerbrugge N, Vasen HF, Nederlof PM, Wijnen JT, van Asperen CJ, Devilee P: Exome sequencing of germline DNA from non-BRCA1/2 familial breast cancer cases selected on the basis of aCGH tumor profiling. PLoS One. 2013, 8: e55734-CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Hilbers FS, Meijers CM, Laros JF, van Galen M, Hoogerbrugge N, Vasen HF, Nederlof PM, Wijnen JT, van Asperen CJ, Devilee P: Rare variants in XRCC2 as breast cancer susceptibility alleles. J Med Genet. 2012, 49: 618-620.CrossRefPubMed Hilbers FS, Meijers CM, Laros JF, van Galen M, Hoogerbrugge N, Vasen HF, Nederlof PM, Wijnen JT, van Asperen CJ, Devilee P: Rare variants in XRCC2 as breast cancer susceptibility alleles. J Med Genet. 2012, 49: 618-620.CrossRefPubMed
11.
Zurück zum Zitat Ruark E, Snape K, Humburg P, Loveday C, Bajrami I, Brough R, Rodrigues DN, Renwick A, Seal S, Ramsay E, Duarte Sdel V, Rivas MA, Warren-Perry M, Zachariou A, Campion-Flora A, Hanks S, Murray A, Ansari Pour N, Douglas J, Gregory L, Rimmer A, Walker NM, Yang TP, Adlard JW, Barwell J, Berg J, Brady AF, Brewer C, Brice G, Chapman C, et al: Mosaic PPM1D mutations are associated with predisposition to breast and ovarian cancer. Nature. 2013, 493: 406-410.CrossRefPubMed Ruark E, Snape K, Humburg P, Loveday C, Bajrami I, Brough R, Rodrigues DN, Renwick A, Seal S, Ramsay E, Duarte Sdel V, Rivas MA, Warren-Perry M, Zachariou A, Campion-Flora A, Hanks S, Murray A, Ansari Pour N, Douglas J, Gregory L, Rimmer A, Walker NM, Yang TP, Adlard JW, Barwell J, Berg J, Brady AF, Brewer C, Brice G, Chapman C, et al: Mosaic PPM1D mutations are associated with predisposition to breast and ovarian cancer. Nature. 2013, 493: 406-410.CrossRefPubMed
12.
Zurück zum Zitat Lynch HT, Krush AJ, Lemon HM, Kaplan AR, Condit PT, Bottomley RH: Tumor variation in families with breast cancer. JAMA. 1972, 222: 1631-1635.CrossRefPubMed Lynch HT, Krush AJ, Lemon HM, Kaplan AR, Condit PT, Bottomley RH: Tumor variation in families with breast cancer. JAMA. 1972, 222: 1631-1635.CrossRefPubMed
13.
Zurück zum Zitat Lynch H, Wen H, Kim YC, Snyder C, Kinarsky Y, Chen PX, Xiao F, Goldgar D, Cowan KH, Wang SM: Can unknown predisposition in familial breast cancer be family-specific?. Breast J. 2013, 19: 520-528.PubMed Lynch H, Wen H, Kim YC, Snyder C, Kinarsky Y, Chen PX, Xiao F, Goldgar D, Cowan KH, Wang SM: Can unknown predisposition in familial breast cancer be family-specific?. Breast J. 2013, 19: 520-528.PubMed
15.
Zurück zum Zitat McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, Garimella K, Altshuler D, Gabriel S, Daly M, DePristo MA: The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010, 20: 1297-1303.CrossRefPubMedPubMedCentral McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, Garimella K, Altshuler D, Gabriel S, Daly M, DePristo MA: The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010, 20: 1297-1303.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD, Lin L, Miller CA, Mardis ER, Ding L, Wilson RK: VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012, 22: 568-576.CrossRefPubMedPubMedCentral Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD, Lin L, Miller CA, Mardis ER, Ding L, Wilson RK: VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012, 22: 568-576.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R: 1000 Genome Project Data Processing Subgroup, 1000 Genome Project Data. The sequence alignment/map (SAM) format and SAMtools. Bioinformatics. 2009, 25: 2078-2079.CrossRefPubMedPubMedCentral Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R: 1000 Genome Project Data Processing Subgroup, 1000 Genome Project Data. The sequence alignment/map (SAM) format and SAMtools. Bioinformatics. 2009, 25: 2078-2079.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, Kondrashov AS, Sunyaev SR: A method and server for predicting damaging missense mutations. Nat Methods. 2010, 7: 248-249.CrossRefPubMedPubMedCentral Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, Kondrashov AS, Sunyaev SR: A method and server for predicting damaging missense mutations. Nat Methods. 2010, 7: 248-249.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Lonita-Laza I, Ottman R: Study designs for identification of rare disease variants in complex diseases: the utility of family-based designs. Genetics. 2011, 189 (3): 1061-1068. PMID: 21840850CrossRef Lonita-Laza I, Ottman R: Study designs for identification of rare disease variants in complex diseases: the utility of family-based designs. Genetics. 2011, 189 (3): 1061-1068. PMID: 21840850CrossRef
20.
Zurück zum Zitat Machin D, Campbell M, Fayers P, Pinol A: Sample Size Tables for Clinical Studies. 1997, Malden, MA: Blackwell Science, 2 Machin D, Campbell M, Fayers P, Pinol A: Sample Size Tables for Clinical Studies. 1997, Malden, MA: Blackwell Science, 2
21.
Zurück zum Zitat Bamshad MJ, Ng SB, Bigham AW, Tabor HK, Emond MJ, Nickerson DA, Shendure J: Exome sequencing as a tool for Mendelian disease gene discovery. Nat Rev Genet. 2011, 12: 745-755.CrossRefPubMed Bamshad MJ, Ng SB, Bigham AW, Tabor HK, Emond MJ, Nickerson DA, Shendure J: Exome sequencing as a tool for Mendelian disease gene discovery. Nat Rev Genet. 2011, 12: 745-755.CrossRefPubMed
22.
Zurück zum Zitat Valente EM, Abou-Sleiman PM, Caputo V, Muqit MM, Harvey K, Gispert S, Ali Z, Del Turco D, Bentivoglio AR, Healy DG, Albanese A, Nussbaum R, González-Maldonado R, Deller T, Salvi S, Cortelli P, Gilks WP, Latchman DS, Harvey RJ, Dallapiccola B, Auburger G, Wood NW: Hereditary early-onset Parkinson's disease caused by mutations in PINK1. Science. 2004, 304: 1158-1160.CrossRefPubMed Valente EM, Abou-Sleiman PM, Caputo V, Muqit MM, Harvey K, Gispert S, Ali Z, Del Turco D, Bentivoglio AR, Healy DG, Albanese A, Nussbaum R, González-Maldonado R, Deller T, Salvi S, Cortelli P, Gilks WP, Latchman DS, Harvey RJ, Dallapiccola B, Auburger G, Wood NW: Hereditary early-onset Parkinson's disease caused by mutations in PINK1. Science. 2004, 304: 1158-1160.CrossRefPubMed
23.
Zurück zum Zitat Champagne N, Bertos NR, Pelletier N, Wang AH, Vezmar M, Yang Y, Heng HH, Yang XJ: Identification of a human histone acetyltransferase related to monocytic leukemia zinc finger protein. J Biol Chem. 1999, 274: 28528-28536.CrossRefPubMed Champagne N, Bertos NR, Pelletier N, Wang AH, Vezmar M, Yang Y, Heng HH, Yang XJ: Identification of a human histone acetyltransferase related to monocytic leukemia zinc finger protein. J Biol Chem. 1999, 274: 28528-28536.CrossRefPubMed
24.
Zurück zum Zitat Kraft M, Cirstea IC, Voss AK, Thomas T, Goehring I, Sheikh BN, Gordon L, Scott H, Smyth GK, Ahmadian MR, Trautmann U, Zenker M, Tartaglia M, Ekici A, Reis A, Dörr HG, Rauch A, Thiel CT: Disruption of the histone acetyltransferase MYST4 leads to a Noonan syndrome-like phenotype and hyperactivated MAPK signaling in humans and mice. J Clin Invest. 2011, 121: 3479-3491.CrossRefPubMedPubMedCentral Kraft M, Cirstea IC, Voss AK, Thomas T, Goehring I, Sheikh BN, Gordon L, Scott H, Smyth GK, Ahmadian MR, Trautmann U, Zenker M, Tartaglia M, Ekici A, Reis A, Dörr HG, Rauch A, Thiel CT: Disruption of the histone acetyltransferase MYST4 leads to a Noonan syndrome-like phenotype and hyperactivated MAPK signaling in humans and mice. J Clin Invest. 2011, 121: 3479-3491.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Mazzone M, Selfors LM, Albeck J, Overholtzer M, Sale S, Carroll DL, Pandya D, Lu Y, Mills GB, Aster JC, Artavanis-Tsakonas S, Brugge JS: Dose-dependent induction of distinct phenotypic responses to Notch pathway activation in mammary epithelial cells. Proc Natl Acad Sci U S A. 2010, 107: 5012-5017.CrossRefPubMedPubMedCentral Mazzone M, Selfors LM, Albeck J, Overholtzer M, Sale S, Carroll DL, Pandya D, Lu Y, Mills GB, Aster JC, Artavanis-Tsakonas S, Brugge JS: Dose-dependent induction of distinct phenotypic responses to Notch pathway activation in mammary epithelial cells. Proc Natl Acad Sci U S A. 2010, 107: 5012-5017.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Simpson MA, Irving MD, Asilmaz E, Gray MJ, Dafou D, Elmslie FV, Mansour S, Holder SE, Brain CE, Burton BK, Kim KH, Pauli RM, Aftimos S, Stewart H, Kim CA, Holder-Espinasse M, Robertson SP, Drake WM, Trembath RC: Mutations in NOTCH2 cause Hajdu-Cheney syndrome, a disorder of severe and progressive bone loss. Nat Genet. 2011, 43: 303-305.CrossRefPubMed Simpson MA, Irving MD, Asilmaz E, Gray MJ, Dafou D, Elmslie FV, Mansour S, Holder SE, Brain CE, Burton BK, Kim KH, Pauli RM, Aftimos S, Stewart H, Kim CA, Holder-Espinasse M, Robertson SP, Drake WM, Trembath RC: Mutations in NOTCH2 cause Hajdu-Cheney syndrome, a disorder of severe and progressive bone loss. Nat Genet. 2011, 43: 303-305.CrossRefPubMed
27.
Zurück zum Zitat Douglas P, Zhong J, Ye R, Moorhead GB, Xu X, Lees-Miller SP: Protein phosphatase 6 interacts with the DNA-dependent protein kinase catalytic subunit and dephosphorylates gamma-H2AX. Mol Cell Biol. 2010, 30: 1368-1381.CrossRefPubMedPubMedCentral Douglas P, Zhong J, Ye R, Moorhead GB, Xu X, Lees-Miller SP: Protein phosphatase 6 interacts with the DNA-dependent protein kinase catalytic subunit and dephosphorylates gamma-H2AX. Mol Cell Biol. 2010, 30: 1368-1381.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat van den Berg IE, van Beurden EA, de Klerk JB, van Diggelen OP, Malingré HE, Boer MM, Berger R: Autosomal recessive phosphorylase kinase deficiency in liver, caused by mutations in the gene encoding the beta subunit (PHKB). Am J Hum Genet. 1997, 61: 539-546.CrossRefPubMedPubMedCentral van den Berg IE, van Beurden EA, de Klerk JB, van Diggelen OP, Malingré HE, Boer MM, Berger R: Autosomal recessive phosphorylase kinase deficiency in liver, caused by mutations in the gene encoding the beta subunit (PHKB). Am J Hum Genet. 1997, 61: 539-546.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Lone S, Townson SA, Uljon SN, Johnson RE, Brahma A, Nair DT, Prakash S, Prakash L, Aggarwal AK: Human DNA polymerase kappa encircles DNA: implications for mismatch extension and lesion bypass. Mol Cell. 2007, 25: 601-614.CrossRefPubMed Lone S, Townson SA, Uljon SN, Johnson RE, Brahma A, Nair DT, Prakash S, Prakash L, Aggarwal AK: Human DNA polymerase kappa encircles DNA: implications for mismatch extension and lesion bypass. Mol Cell. 2007, 25: 601-614.CrossRefPubMed
30.
Zurück zum Zitat Seki M, Masutani C, Yang LW, Schuffert A, Iwai S, Bahar I, Wood RD: High-efficiency bypass of DNA damage by human DNA polymerase Q. EMBO J. 2005, 23: 4484-4494.CrossRef Seki M, Masutani C, Yang LW, Schuffert A, Iwai S, Bahar I, Wood RD: High-efficiency bypass of DNA damage by human DNA polymerase Q. EMBO J. 2005, 23: 4484-4494.CrossRef
31.
Zurück zum Zitat van der Spek PJ, Smit EM, Beverloo HB, Sugasawa K, Masutani C, Hanaoka F, Hoeijmakers JH, Hagemeijer A: Chromosomal localization of three repair genes: the xeroderma pigmentosum group C gene and two human homologs of yeast RAD23. Genomics. 1995, 23: 651-658.CrossRef van der Spek PJ, Smit EM, Beverloo HB, Sugasawa K, Masutani C, Hanaoka F, Hoeijmakers JH, Hagemeijer A: Chromosomal localization of three repair genes: the xeroderma pigmentosum group C gene and two human homologs of yeast RAD23. Genomics. 1995, 23: 651-658.CrossRef
33.
Zurück zum Zitat O'Driscoll M, Cerosaletti KM, Girard PM, Dai Y, Stumm M, Kysela B, Hirsch B, Gennery A, Palmer SE, Seidel J, Gatti RA, Varon R, Oettinger MA, Neitzel H, Jeggo PA, Concannon P: DNA ligase IV mutations identified in patients exhibiting developmental delay and immunodeficiency. Mol Cell. 2001, 8: 1175-1185.CrossRefPubMed O'Driscoll M, Cerosaletti KM, Girard PM, Dai Y, Stumm M, Kysela B, Hirsch B, Gennery A, Palmer SE, Seidel J, Gatti RA, Varon R, Oettinger MA, Neitzel H, Jeggo PA, Concannon P: DNA ligase IV mutations identified in patients exhibiting developmental delay and immunodeficiency. Mol Cell. 2001, 8: 1175-1185.CrossRefPubMed
34.
Zurück zum Zitat Kouros-Mehr H, Slorach EM, Sternlicht MD, Werb Z: GATA-3 maintains the differentiation of the luminal cell fate in the mammary gland. Cell. 2006, 127: 1041-1055.CrossRefPubMedPubMedCentral Kouros-Mehr H, Slorach EM, Sternlicht MD, Werb Z: GATA-3 maintains the differentiation of the luminal cell fate in the mammary gland. Cell. 2006, 127: 1041-1055.CrossRefPubMedPubMedCentral
35.
36.
Zurück zum Zitat Network CGA: Comprehensive molecular portraits of human breast tumours. Nature. 2012, 490: 61-70.CrossRef Network CGA: Comprehensive molecular portraits of human breast tumours. Nature. 2012, 490: 61-70.CrossRef
37.
Zurück zum Zitat Wilson BJ, Giguere V: Meta-analysis of human cancer microarrays reveals that GATA3 is integral to the estrogen receptor alpha pathway. Mol Cancer. 2008, 7: 49-CrossRefPubMedPubMedCentral Wilson BJ, Giguere V: Meta-analysis of human cancer microarrays reveals that GATA3 is integral to the estrogen receptor alpha pathway. Mol Cancer. 2008, 7: 49-CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Dydensborg AB, Rose AA, Wilson BJ, Grote D, Paquet M, Giguère V, Siegel PM, Bouchard M: GATA3 inhibits breast cancer growth and pulmonary breast cancer metastasis. Oncogene. 2009, 28: 2634-2642.CrossRefPubMed Dydensborg AB, Rose AA, Wilson BJ, Grote D, Paquet M, Giguère V, Siegel PM, Bouchard M: GATA3 inhibits breast cancer growth and pulmonary breast cancer metastasis. Oncogene. 2009, 28: 2634-2642.CrossRefPubMed
39.
Zurück zum Zitat Rowley JD: A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature. 1973, 243: 290-293.CrossRefPubMed Rowley JD: A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature. 1973, 243: 290-293.CrossRefPubMed
40.
Zurück zum Zitat Southey MC, Park DJ, Nguyen-Dumont T, Campbell I, Thompson E, Trainer AH, Chenevix-Trench G, Simard J, Dumont M, Soucy P, Thomassen M, Jønson L, Pedersen IS, Hansen TV, Nevanlinna H, Khan S, Sinilnikova O, Mazoyer S, Lesueur F, Damiola F, Schmutzler R, Meindl A, Hahnen E, Dufault MR, Chris Chan T, Kwong A, Barkardóttir R, Radice P, Peterlongo P, COMPLEXO, et al: COMPLEXO: identifying the missing heritability of breast cancer via next generation collaboration. Breast Cancer Res. 2013, 15: 402-PubMedPubMedCentral Southey MC, Park DJ, Nguyen-Dumont T, Campbell I, Thompson E, Trainer AH, Chenevix-Trench G, Simard J, Dumont M, Soucy P, Thomassen M, Jønson L, Pedersen IS, Hansen TV, Nevanlinna H, Khan S, Sinilnikova O, Mazoyer S, Lesueur F, Damiola F, Schmutzler R, Meindl A, Hahnen E, Dufault MR, Chris Chan T, Kwong A, Barkardóttir R, Radice P, Peterlongo P, COMPLEXO, et al: COMPLEXO: identifying the missing heritability of breast cancer via next generation collaboration. Breast Cancer Res. 2013, 15: 402-PubMedPubMedCentral
41.
Zurück zum Zitat Hill SM, Klotz DM, Cohn CS: Genetics of Breast cancer. Hormone and Cancer. Edited by: Vedeckis, Wayne V. 1996, Boston: Birkhauser, 199- Hill SM, Klotz DM, Cohn CS: Genetics of Breast cancer. Hormone and Cancer. Edited by: Vedeckis, Wayne V. 1996, Boston: Birkhauser, 199-
42.
Zurück zum Zitat Xu X, Wagner KU, Larson D, Weaver Z, Li C, Ried T, Hennighausen L, Wynshaw-Boris A, Deng CX: Conditional mutation of Brca1 in mammary epithelial cells results in blunted ductal morphogenesis and tumour formation. Nat Genet. 1999, 22: 37-43.CrossRefPubMed Xu X, Wagner KU, Larson D, Weaver Z, Li C, Ried T, Hennighausen L, Wynshaw-Boris A, Deng CX: Conditional mutation of Brca1 in mammary epithelial cells results in blunted ductal morphogenesis and tumour formation. Nat Genet. 1999, 22: 37-43.CrossRefPubMed
43.
Zurück zum Zitat Sherman S, Shats O, Fleissner E, Bascom G, Yiee K, Copur M, Crow K, Rooney J, Mateen Z, Ketcham MA, Feng J, Sherman A, Gleason M, Kinarsky L, Silva-Lopez E, Edney J, Reed E, Berger A, Cowan K: Multicenter breast cancer collaborative registry. Cancer Inform. 2011, 10: 217-226. PMID: 21918596CrossRefPubMedPubMedCentral Sherman S, Shats O, Fleissner E, Bascom G, Yiee K, Copur M, Crow K, Rooney J, Mateen Z, Ketcham MA, Feng J, Sherman A, Gleason M, Kinarsky L, Silva-Lopez E, Edney J, Reed E, Berger A, Cowan K: Multicenter breast cancer collaborative registry. Cancer Inform. 2011, 10: 217-226. PMID: 21918596CrossRefPubMedPubMedCentral
Metadaten
Titel
Family-specific, novel, deleterious germline variants provide a rich resource to identify genetic predispositions for BRCAx familial breast cancer
verfasst von
Hongxiu Wen
Yeong C Kim
Carrie Snyder
Fengxia Xiao
Elizabeth A Fleissner
Dina Becirovic
Jiangtao Luo
Bradley Downs
Simon Sherman
Kenneth H Cowan
Henry T Lynch
San Ming Wang
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2014
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-14-470

Weitere Artikel der Ausgabe 1/2014

BMC Cancer 1/2014 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.