Entry - *608798 - GASDERMIN E; GSDME - OMIM
 
* 608798

GASDERMIN E; GSDME


Alternative titles; symbols

DFNA5 GENE; DFNA5
INVERSELY CORRELATED WITH ESTROGEN RECEPTOR EXPRESSION 1; ICERE1


HGNC Approved Gene Symbol: GSDME

Cytogenetic location: 7p15.3     Genomic coordinates (GRCh38): 7:24,698,355-24,795,539 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
7p15.3 Deafness, autosomal dominant 5 600994 AD 3

TEXT

Description

Pyroptosis is a form of cell death that involves perforation of cell membranes via pore-forming proteins. GSDME is a gasdermin protein that is specifically cleaved and activated by caspase-3 (CASP3; 600636), resulting in pyroptosis (Rogers et al., 2017; Wang et al., 2017).


Cloning and Expression

By a positional cloning strategy in the critical region defined for autosomal dominant nonsyndromic deafness-5 (600994), Van Laer et al. (1998) isolated a gene, designated DFNA5, that is expressed in the cochlea. The DFNA5 cDNA sequence predicted a 496-amino acid protein. Van Laer et al. (1998) found significant homology between DFNA5 and the 'inversely correlated with estrogen receptor expression' gene (ICERE1). ICERE1 had been identified in a differential display study comparing estrogen receptor-positive and estrogen receptor-negative breast carcinomas (Thompson and Weigel, 1998). On the basis of various considerations, Van Laer et al. (1998) concluded that ICERE1 and DFNA5 probably represent the same gene and that the start and stop codons had been identified incorrectly in ICERE1. Van Laer et al. (1998) also cloned the mouse homolog by screening a mouse cochlea cDNA library. Northern blot analysis of 8 human tissues detected a 2.2-kb DFNA5 transcript that was highly expressed in placenta, with much lower expression in heart, brain, and kidney. RT-PCR detected Dfna5 in mouse cochlear epithelial ridge and stria vascularis.

By comparing DFNA5-gasdermin (see 611218) family members, Op de Beeck et al. (2011) determined that DFNA5 has a 3-domain structure, with globular domains A and B separated by a hinge region. N-terminal domain A was predicted to have an alpha/beta fold, whereas C-terminal domain B was predicted to have long alpha-helical structures that may form coiled-coils.

Wang et al. (2017) found that GSDME was variably expressed in human cell lines and in normal human epidermal keratinocytes, placental epithelial cells, and umbilical artery smooth muscle cells, but not umbilical vein endothelial cells or aortic smooth muscle cells. Database analysis suggested that GSDME is the most ancient gasdermin, with conservation in lancelet.


Gene Structure

Van Laer et al. (1998) determined that the DFNA5 gene contains 10 exons and spans approximately 60 kb. A 5-prime UTR of 57 bp preceded the putative translation start.


Mapping

Van Laer et al. (1998) identified the DFNA5 gene within the critical region for the deafness locus mapped to chromosome 7p15. Van Laer et al. (1998) mapped the mouse homolog by FISH to chromosome 6 in a region of homology to human chromosome 7p15. They noted that no deaf mouse mutants had been mapped to that region.


Gene Function

Thompson and Weigel (1998) found that expression of the ICERE1 gene was inversely correlated with that of the estrogen receptor (ESR1; 133430) in breast carcinomas.

By microarray analysis of p53 (TP53; 191170)-induced transcripts in a human hepatocellular carcinoma cell line, Masuda et al. (2006) identified DFNA5. The expression of DFNA5 was activated by endogenous p53 in response to various cellular stresses. Sequence analysis and ChIP assays revealed a p53-binding motif in intron 1 of the DFNA5 gene. RT-PCR of tissues obtained from irradiated wildtype and p53-knockdown mice revealed p53-dependent Dfna5 induction in colon. p53-dependent expression of Dfna5 was observed in the brain and colon in the absence of DNA damage, but the expression of Dfna5 in other tissues was independent of p53 expression. Immunostaining of DFNA5-transfected cells revealed expression predominantly in the cytoplasm, with a subset of cells in the nucleus. Masuda et al. (2006) concluded that DFNA5 has a potential role in p53-regulated response to DNA damage.

By transfection of human and monkey cell lines, Op de Beeck et al. (2011) showed that overexpression of human DFNA5 domain A, but not full-length DFNA5, induced apoptosis prior to development of necrotic characteristics. Truncations within domain A revealed that the entire domain A structure was required to induce apoptosis in HEK293 cells. Microarray analysis of wildtype and Dfna5 -/- postnatal day-0 mouse inner ear samples showed that Dfna5 knockout resulted in upregulation of genes involved in cartilage maintenance and DNA repair and downregulation of genes involved in energy metabolism and apoptosis.

Using mouse and human cells, Rogers et al. (2017) found that CASP3 (600636) cleaved GSDME after asp270 to generate a necrotic N-terminal fragment that targeted itself to the plasma membrane to induce secondary necrosis/pyroptosis. Cells expressing GSDME progressed to secondary necrosis when stimulated with apoptotic triggers, such as etoposide or vesicular stomatitis virus, but disassembled into small apoptotic bodies when GSDME was deleted. Rogers et al. (2017) concluded that GSDME is a central molecule that regulates apoptotic cell disassembly and progression to secondary necrosis.

Independently, Wang et al. (2017) found that CASP3 cleaved human GSDME following asp270 in vitro and in cell lines, and that the N-terminal fragment of GSDME changed the cellular response to TNF (191160) or chemotherapeutic agents from apoptosis to pyroptosis. Human cell lines that lacked GSDME expression did not show pyroptotic response to TNF or chemotherapeutic agents. Mutation of asp267 or asp270 in the CASP3 recognition motif of GSDME, knockout of GSDME expression, or knockout or inhibition of CASP3 abrogated the pyroptotic response to TNF or chemotherapeutic agents. Liposome experiments suggested that pyroptosis involved binding of the N-terminal domain of GSDME to phosphatidylinositol-4,5-bisphosphate, formation of pores, and loss of liposome contents.

Zhang et al. (2020) showed that 20 of 22 tested cancer-associated GSDME mutations reduced GSDME function. In mice, knocking out Gsdme in GSDME-expressing tumors enhanced, whereas ectopic expression in Gsdme-repressed tumors inhibited, tumor growth. This tumor suppression was mediated by killer cytotoxic lymphocytes: it was abrogated in perforin (170280)-deficient mice or mice depleted of killer lymphocytes. GSDME expression enhanced the phagocytosis of tumor cells by tumor-associated macrophages, as well as the number and functions of tumor-infiltrating natural killer and CD8+ T cells. Killer cell granzyme B (123910) also activated caspase-independent pyroptosis in target cells by directly cleaving GSDME at the same site as CASP3. Uncleavable or pore-defective GSDME proteins were not tumor-suppressive. Thus, tumor GSDME acts as a tumor suppressor by activating pyroptosis, enhancing antitumor immunity.


Molecular Genetics

Van Laer et al. (1998) identified a mutation in intron 7 of the DFNA5 gene (608798.0001) that cosegregated with nonsyndromic deafness in the Dutch family reported by Van Camp et al. (1995) and Van Laer et al. (1997); see 600994. The insertion/deletion mutation did not affect intron-exon boundaries, but deleted 5 G-triplets at the 3-prime end of the intron, causing skipping of exon 8. The absence of exon 8 resulted in a frameshift starting at amino acid 330, which introduced an aberrant stretch of 41 amino acids followed by a stop codon that prematurely terminated the protein.

Age-related hearing impairment (ARHI), or presbycusis, is a sensorineural high frequency hearing loss that is believed to result from an interaction between environmental and genetic factors. Because the hearing loss in the DFNA5 family closely resembles that observed in ARHI, Van Laer et al. (2002) performed linkage analysis on a quantitative measure of high frequency hearing loss in 328 pedigrees from the National Heart Lung and Blood Institute's Framingham Heart Study. No significant linkage between ARHI and microsatellite markers from the DFNA5 region was detected. Sequencing of the DFNA5 coding region in 10 random individuals resulted in the detection of 6 SNPs. Two of these SNPs led to amino acid substitutions (P142H and V207M) and were selected for further analysis. A pilot experiment on 116 random Belgian and Dutch subjects was performed, but no association could be detected between either SNP and ARHI. Subsequently, 93 ARHI cases and 83 controls selected from the Framingham cohort were genotyped for P142H, but no allelic or genotypic association was detected. Van Laer et al. (2002) concluded that possibly many different genes contribute to ARHI, each with a weak effect, and ARHI might turn out to be too complex to study with moderate sample sizes.

In a Chinese family with autosomal dominant nonsyndromic sensorineural deafness, Yu et al. (2003) identified a 3-bp deletion in the DFNA5 gene (608798.0002) that segregated with the phenotype and was predicted to cause skipping of exon 8, like the DFNA5 mutation previously reported by Van Laer et al. (1998).

In a 5-generation Dutch family with autosomal dominant deafness, Bischoff et al. (2004) identified a splice-site mutation in the DFNA5 gene (608798.0003) that caused skipping of exon 8. Because of the relatively low amount of short transcript in affected members in this family, the authors suggested that the mutation might have a dominant-negative effect rather than haploinsufficiency.

In transfection studies using HEK293T cells, Van Laer et al. (2004) found that posttransfection cell death approximately doubled when cells were transfected with mutant DFNA5-GFP compared to wildtype DFNA5-GFP, with cell death being attributed to necrotic rather than apoptotic events. This information, coupled with the observation that all 3 identified mutations in DFNA5 result in skipping of exon 8 at the mRNA level, led Van Laer et al. (2004) to propose that the hearing impairment associated with DFNA5 is caused by a gain-of-function mutation.

Cheng et al. (2007) reported a 5-generation Chinese family with autosomal late-onset deafness in which they identified a heterozygous splice site mutation resulting in skipping of exon 8 as confirmed by RT-PCR analysis.

In a 5-generation Iranian family segregating nonsyndromic autosomal dominant sensorineural hearing loss that appeared to map to the DFNA5 locus, Van Laer et al. (2007) identified a heterozygous truncating mutation in exon 5 of the DFNA5 gene. However, further analysis revealed that the mutation did not segregate with hearing loss in this family, and linkage to a locus on chromosome 4 was subsequently found. The authors stated that these findings supported the hypothesis that only a specific gain-of-function mutation caused by skipping of exon 8 can lead to DFNA5-associated hearing loss.

Wang et al. (2017) found that mutant GSDME protein resulting from skipping of exon 8 was unstable and triggered extensive pyroptosis when expressed in 293T cells.

Using 2 next-generation sequencing platforms, Booth et al. (2018) identified 5 families with autosomal dominant postlingual progressive nonsyndromic hearing loss with 3 novel and 2 recurrent mutations in the DFNA5 gene. The 3 novel mutations were missense mutations within exon 8 that were predicted to reduce the efficiency of, or abolish, splicing (see, e.g., Q368E, 608798.0005). Functional impact of these 3 mutations was confirmed in vitro using minigenes. The authors noted that previously overlooked silent mutations within exon 8 could alter splicing; thus, families with high frequency progressive hearing loss that links to the DFNA5 gene should be evaluated for variants in the flanking introns and in the exon itself.


Animal Model

Wang et al. (2017) found that Gsdme -/- mice developed normally. Compared with wildtype, Gsdme -/- mice were refractory to cisplatin- or bleomycin-induced injury of gastrointestinal tissues, spleen, and lung.


ALLELIC VARIANTS ( 5 Selected Examples):

.0001 DEAFNESS, AUTOSOMAL DOMINANT 5

GSDME, INS/DEL, EX8DEL
   RCV000002171

In an extended Dutch family with autosomal dominant deafness (DFNA5; 600994), Van Laer et al. (1998) found that affected members had a deletion of 1,189 bp from intron 7 of the DFNA5 gene and insertion of 127 bp from intron 8 into intron 7 in the opposite direction, followed by a GCCCA stretch of unknown origin. Exons 7 and 8, the intron-exon boundaries, and the branchpoint sequence from intron 7 were still present. It was demonstrated that exon 8 (193 bp) was skipped in affected family members. The absence of exon 8 resulted in a frameshift starting at amino acid 330, which introduced an aberrant stretch of 41 amino acids followed by a stop codon that prematurely terminated the protein. The aberrant splicing was thought to be caused by deletion of the 5 G-triplets in intron 7. There is evidence from other sources that sequences other than the intron/exon consensus and the branchpoint are involved in splice site selection. In particular, intronic G-triplets play an important role.


.0002 DEAFNESS, AUTOSOMAL DOMINANT 5

GSMDE, 3-BP DEL, IVS7, -17CTT
  
RCV000156800...

In affected members of a family with autosomal dominant nonsyndromic sensorineural deafness (DFNA5; 600994), Yu et al. (2003) identified a mutation in the DFNA5 gene, a CTT deletion in the polypyrimidine tract of intron 7, which was predicted to create a shift in the reading frame and introduce a stop codon at position 372. This mutation, like the previously reported mutation (600994.0001), led to skipping of exon 8 of DFNA5. Yu et al. (2003) confirmed the existence of a previously identified short isoform of DFNA5 and concluded that the 3-nucleotide deletion in their family did not affect the function of this short isoform.

Park et al. (2010) identified the 3-bp deletion in intron 7, which they referred to as 991-15_991-13del, in affected members of a Korean family with nonsyndromic sensorineural deafness. Haplotype analysis showed that this family and the Chinese family reported by Yu et al. (2003) shared a common haplotype, suggesting a founder effect.


.0003 DEAFNESS, AUTOSOMAL DOMINANT 5

GSMDE, IVS7AS, C-G, -6
  
RCV000002173

In affected members of a 5-generation Dutch family with autosomal dominant deafness (DFNA5; 600994), Bischoff et al. (2004) identified a 1200C-G transversion at position -6 of the splice acceptor site of intron 7 of the DFNA5 gene. This results in skipping of exon 8, which leads to 41 aberrant codons followed by a premature stop codon. The C-to-G transversion creates an RsaI restriction site, which was used to demonstrate cosegregation of the mutation in the family. Because of the relatively low amount of the short transcript in affected members in this family, Bischoff et al. (2004) suggested that this may represent a dominant-negative effect rather than haploinsufficiency.


.0004 DEAFNESS, AUTOSOMAL DOMINANT 5

GSMDE, IVS8DS, A-G, +4
  
RCV000002174

In affected members of a large 5-generation Chinese family with autosomal dominant late-onset deafness (DFNA5; 600994), Cheng et al. (2007) identified a heterozygous splice site mutation in intron 8 of the DFNA5 gene, resulting in skipping of exon 8 as confirmed by RT-PCR analysis. The resultant protein was predicted to be prematurely terminated. The mutation was absent in 100 control chromosomes. Cheng et al. (2007) noted that all of the pathogenic mutations described in the DFNA5 gene result in skipping of exon 8, suggesting a very specific gain-of-function effect.


.0005 DEAFNESS, AUTOSOMAL DOMINANT 5

GSMDE, GLN368GLU
  
RCV001726686

In affected members of a European family (CDS-6824) with autosomal dominant postlingual progressive nonsyndromic hearing loss (DFNA5; 600994), Booth et al. (2018) identified a c.1102C-G transversion (c.1102C-G, NM_004403.2) in exon 8 of the DFNA5 gene, resulting in a gln368-to-glu (Q368E) substitution at a conserved residue, that segregated the phenotype. The variant was predicted to alter splicing due to creation of both a new cryptic donor site and cryptic acceptor site. In vitro analysis using a minigene with exon 8 of DFNA5 showed that the variant causes skipping of exon 8. The mutation was not present in the ExAC (v.0.3) or gnomAD (v.2.0.2) databases.


REFERENCES

  1. Bischoff, A. M. L. C., Luijendijk, M. W. J., Huygen, P. L. M., van Duijnhoven, G., De Leenheer, E. M. R., Oudesluijs, G. G., Van Laer, L., Cremers, F. P. M., Cremers, C. W. R. J., Kremer, H. A novel mutation identified in the DFNA5 gene in a Dutch family: a clinical and genetic evaluation. Audiol. Neurootol. 9: 34-46, 2004. [PubMed: 14676472, related citations] [Full Text]

  2. Booth, K. T., Azaiez, H., Kahrizi, K., Wang, D., Zhang, Y., Frees, K., Nishimura, C., Najmabadi, H., Smith, R. J. Exonic mutations and exon skipping: lessons learned from DFNA5. Hum. Mutat. 39: 433-440, 2018. [PubMed: 29266521, images, related citations] [Full Text]

  3. Cheng, J., Han, D. Y., Dai, P., Sun, H. J., Tao, R., Sun, Q., Yan, D., Qin, W., Wang, H. Y., Ouyang, X. M., Yang, S. Z., Cao, J. Y., Feng, G. Y., Du, L. L., Zhang, Y. Z., Zhai, S. Q., Yang, W. Y., Liu, X. Z., He, L., Yuan, H. J. A novel DFNA5 mutation, IVS8+4A-G, in the splice donor site of intron 8 causes late-onset non-syndromic hearing loss in a Chinese family. Clin. Genet. 72: 471-477, 2007. [PubMed: 17868390, related citations] [Full Text]

  4. Masuda, Y., Futamura, M., Kamino, H., Nakamura, Y., Kitamura, N., Ohnishi, S., Miyamoto, Y., Ichikawa, H., Ohta, T., Ohki, M., Kiyono, T., Egami, H., Baba, H., Arakawa, H. The potential role of DFNA5, a hearing impairment gene, in p53-mediated cellular response to DNA damage. J. Hum. Genet. 51: 652-664, 2006. [PubMed: 16897187, related citations] [Full Text]

  5. Op de Beeck, K. O., Van Camp, G., Thys, S., Cools, N., Callebaut, I., Vrijens, K., Van Nassauw, L., Van Tendeloo, V. F., Timmermans, J. P., Van Laer, L. The DFNA5 gene, responsible for hearing loss and involved in cancer, encodes a novel apoptosis-inducing protein. Europ. J. Hum. Genet. 19: 965-973, 2011. [PubMed: 21522185, images, related citations] [Full Text]

  6. Park, H.-J., Cho, H.-J., Baek, J.-I., Ben-Yosef, T., Kwon, T.-J., Griffith, A. J., Kim, U.-K. Evidence for a founder mutation causing DFNA5 hearing loss in East Asians. J. Hum. Genet. 55: 59-62, 2010. [PubMed: 19911014, related citations] [Full Text]

  7. Rogers, C., Fernandes-Alnemri, T., Mayes, L., Alnemri, D., Cingolani, G., Alnemri, E. S. Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death. Nature Commun. 8: 14128, 2017. Note: Electronic Article. [PubMed: 28045099, images, related citations] [Full Text]

  8. Thompson, D. A., Weigel, R. J. Characterization of a gene that is inversely correlated with estrogen receptor expression (ICERE-1) in breast carcinomas. Europ. J. Biochem. 252: 169-177, 1998. [PubMed: 9523727, related citations] [Full Text]

  9. Van Camp, G., Coucke, P., Balemans, W., Van Velzen, D., Van de Bilt, C., Van Laer, L., Smith, R. J. H., Fukushima, K., Padberg, G. W., Frants, R. R., Van de Heyning, P., Smith, S. D., Huizing, E. H., Willems, P. J. Localization of a gene for non-syndromic hearing loss (DFNA5) to chromosome 7p15. Hum. Molec. Genet. 4: 2159-2163, 1995. [PubMed: 8589696, related citations] [Full Text]

  10. Van Laer, L., DeStefano, A. L., Myers, R. H., Flothmann, K., Thys, S., Fransen, E., Gates, G. A., Van Camp, G., Baldwin, C. T. Is DFNA5 a susceptibility gene for age-related hearing impairment? Europ. J. Hum. Genet. 10: 883-886, 2002. [PubMed: 12461698, related citations] [Full Text]

  11. Van Laer, L., Huizing, E. H., Verstreken, M., van Zuijlen, D., Wauters, J. G., Bossuyt, P. J., Van de Heyning, P., McGuirt, W. T., Smith, R. J. H., Willems, P. J., Legan, P. K., Richardson, G. P., Van Camp, G. Nonsyndromic hearing impairment is associated with a mutation in DFNA5. Nature Genet. 20: 194-197, 1998. [PubMed: 9771715, related citations] [Full Text]

  12. Van Laer, L., Meyer, N. C., Malekpour, M., Riazalhosseini, Y., Moghannibashi, M., Kahrizi, K., Vandevelde, A., Alasti, F., Najmabadi, H., Van Camp, G., Smith, R. J. H. A novel DFNA5 mutation does not cause hearing loss in an Iranian family. J. Hum. Genet. 52: 549-552, 2007. [PubMed: 17427029, related citations] [Full Text]

  13. Van Laer, L., Van Camp, G., van Zuijlen, D., Green, E. D., Verstreken, M., Schatteman, I., Van de Heyning, P., Balemans, W., Coucke, P., Greinwald, J. H., Smith, R. J. H., Huizing, E., Willems, P. Refined mapping of a gene for autosomal dominant progressive sensorineural hearing loss (DFNA5) to a 2-cM region, and exclusion of a candidate gene that is expressed in the cochlea. Europ. J. Hum. Genet. 5: 397-405, 1997. [PubMed: 9450185, related citations]

  14. Van Laer, L., Vrijens, K., Thys, S., Van Tendeloo, V. F. I., Smith, R. J. H., Van Bockstaele, D. R., Timmermans, J.-P., Van Camp, G. DFNA5: hearing impairment exon instead of hearing impairment gene? J. Med. Genet. 41: 401-406, 2004. [PubMed: 15173223, related citations] [Full Text]

  15. Wang, Y., Gao, W., Shi, X., Ding, J., Liu, W., He, H., Wang, K., Shao, F. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 547: 99-103, 2017. [PubMed: 28459430, related citations] [Full Text]

  16. Yu, C., Meng, X., Zhang, S., Zhao, G., Hu, L., Kong, X. A 3-nucleotide deletion in the polypyrimidine tract of the intron 7 of the DFNA5 gene causes nonsyndromic hearing impairment in a Chinese family. Genomics 82: 575-579, 2003. [PubMed: 14559215, related citations] [Full Text]

  17. Zhang, Z., Zhang, Y., Xia, S., Kong, Q., Li, S., Liu, X., Junqueira, C., Meza-Sosa, K. F., Mok, T. M. Y., Ansara, J., Sengupta, S., Yao, Y., Wu, H., Lieberman, J. Gasdermin E suppresses tumour growth by activating anti-tumour immunity. Nature 579: 415-420, 2020. [PubMed: 32188940, images, related citations] [Full Text]


Sonja A. Rasmussen - updated : 10/01/2021
Ada Hamosh - updated : 06/17/2020
Matthew B. Gross - updated : 10/03/2017
Matthew B. Gross - updated : 08/22/2017
Patricia A. Hartz - updated : 08/22/2017
Patricia A. Hartz - updated : 9/13/2011
Cassandra L. Kniffin - updated : 6/14/2010
Marla J. F. O'Neill - updated : 6/1/2009
Cassandra L. Kniffin - updated : 8/21/2008
Patricia A. Hartz - updated : 10/23/2006
Creation Date:
Victor A. McKusick : 7/13/2004
alopez : 06/23/2023
alopez : 03/13/2023
carol : 10/01/2021
alopez : 06/17/2020
carol : 10/04/2017
mgross : 10/03/2017
mgross : 08/22/2017
mgross : 08/22/2017
alopez : 10/04/2016
carol : 08/14/2015
mgross : 9/13/2011
terry : 9/13/2011
wwang : 6/18/2010
ckniffin : 6/14/2010
wwang : 6/2/2009
terry : 6/1/2009
terry : 12/2/2008
alopez : 12/1/2008
wwang : 9/2/2008
ckniffin : 8/21/2008
carol : 10/25/2006
carol : 10/23/2006
carol : 10/23/2006
carol : 7/13/2004

* 608798

GASDERMIN E; GSDME


Alternative titles; symbols

DFNA5 GENE; DFNA5
INVERSELY CORRELATED WITH ESTROGEN RECEPTOR EXPRESSION 1; ICERE1


HGNC Approved Gene Symbol: GSDME

Cytogenetic location: 7p15.3     Genomic coordinates (GRCh38): 7:24,698,355-24,795,539 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
7p15.3 Deafness, autosomal dominant 5 600994 Autosomal dominant 3

TEXT

Description

Pyroptosis is a form of cell death that involves perforation of cell membranes via pore-forming proteins. GSDME is a gasdermin protein that is specifically cleaved and activated by caspase-3 (CASP3; 600636), resulting in pyroptosis (Rogers et al., 2017; Wang et al., 2017).


Cloning and Expression

By a positional cloning strategy in the critical region defined for autosomal dominant nonsyndromic deafness-5 (600994), Van Laer et al. (1998) isolated a gene, designated DFNA5, that is expressed in the cochlea. The DFNA5 cDNA sequence predicted a 496-amino acid protein. Van Laer et al. (1998) found significant homology between DFNA5 and the 'inversely correlated with estrogen receptor expression' gene (ICERE1). ICERE1 had been identified in a differential display study comparing estrogen receptor-positive and estrogen receptor-negative breast carcinomas (Thompson and Weigel, 1998). On the basis of various considerations, Van Laer et al. (1998) concluded that ICERE1 and DFNA5 probably represent the same gene and that the start and stop codons had been identified incorrectly in ICERE1. Van Laer et al. (1998) also cloned the mouse homolog by screening a mouse cochlea cDNA library. Northern blot analysis of 8 human tissues detected a 2.2-kb DFNA5 transcript that was highly expressed in placenta, with much lower expression in heart, brain, and kidney. RT-PCR detected Dfna5 in mouse cochlear epithelial ridge and stria vascularis.

By comparing DFNA5-gasdermin (see 611218) family members, Op de Beeck et al. (2011) determined that DFNA5 has a 3-domain structure, with globular domains A and B separated by a hinge region. N-terminal domain A was predicted to have an alpha/beta fold, whereas C-terminal domain B was predicted to have long alpha-helical structures that may form coiled-coils.

Wang et al. (2017) found that GSDME was variably expressed in human cell lines and in normal human epidermal keratinocytes, placental epithelial cells, and umbilical artery smooth muscle cells, but not umbilical vein endothelial cells or aortic smooth muscle cells. Database analysis suggested that GSDME is the most ancient gasdermin, with conservation in lancelet.


Gene Structure

Van Laer et al. (1998) determined that the DFNA5 gene contains 10 exons and spans approximately 60 kb. A 5-prime UTR of 57 bp preceded the putative translation start.


Mapping

Van Laer et al. (1998) identified the DFNA5 gene within the critical region for the deafness locus mapped to chromosome 7p15. Van Laer et al. (1998) mapped the mouse homolog by FISH to chromosome 6 in a region of homology to human chromosome 7p15. They noted that no deaf mouse mutants had been mapped to that region.


Gene Function

Thompson and Weigel (1998) found that expression of the ICERE1 gene was inversely correlated with that of the estrogen receptor (ESR1; 133430) in breast carcinomas.

By microarray analysis of p53 (TP53; 191170)-induced transcripts in a human hepatocellular carcinoma cell line, Masuda et al. (2006) identified DFNA5. The expression of DFNA5 was activated by endogenous p53 in response to various cellular stresses. Sequence analysis and ChIP assays revealed a p53-binding motif in intron 1 of the DFNA5 gene. RT-PCR of tissues obtained from irradiated wildtype and p53-knockdown mice revealed p53-dependent Dfna5 induction in colon. p53-dependent expression of Dfna5 was observed in the brain and colon in the absence of DNA damage, but the expression of Dfna5 in other tissues was independent of p53 expression. Immunostaining of DFNA5-transfected cells revealed expression predominantly in the cytoplasm, with a subset of cells in the nucleus. Masuda et al. (2006) concluded that DFNA5 has a potential role in p53-regulated response to DNA damage.

By transfection of human and monkey cell lines, Op de Beeck et al. (2011) showed that overexpression of human DFNA5 domain A, but not full-length DFNA5, induced apoptosis prior to development of necrotic characteristics. Truncations within domain A revealed that the entire domain A structure was required to induce apoptosis in HEK293 cells. Microarray analysis of wildtype and Dfna5 -/- postnatal day-0 mouse inner ear samples showed that Dfna5 knockout resulted in upregulation of genes involved in cartilage maintenance and DNA repair and downregulation of genes involved in energy metabolism and apoptosis.

Using mouse and human cells, Rogers et al. (2017) found that CASP3 (600636) cleaved GSDME after asp270 to generate a necrotic N-terminal fragment that targeted itself to the plasma membrane to induce secondary necrosis/pyroptosis. Cells expressing GSDME progressed to secondary necrosis when stimulated with apoptotic triggers, such as etoposide or vesicular stomatitis virus, but disassembled into small apoptotic bodies when GSDME was deleted. Rogers et al. (2017) concluded that GSDME is a central molecule that regulates apoptotic cell disassembly and progression to secondary necrosis.

Independently, Wang et al. (2017) found that CASP3 cleaved human GSDME following asp270 in vitro and in cell lines, and that the N-terminal fragment of GSDME changed the cellular response to TNF (191160) or chemotherapeutic agents from apoptosis to pyroptosis. Human cell lines that lacked GSDME expression did not show pyroptotic response to TNF or chemotherapeutic agents. Mutation of asp267 or asp270 in the CASP3 recognition motif of GSDME, knockout of GSDME expression, or knockout or inhibition of CASP3 abrogated the pyroptotic response to TNF or chemotherapeutic agents. Liposome experiments suggested that pyroptosis involved binding of the N-terminal domain of GSDME to phosphatidylinositol-4,5-bisphosphate, formation of pores, and loss of liposome contents.

Zhang et al. (2020) showed that 20 of 22 tested cancer-associated GSDME mutations reduced GSDME function. In mice, knocking out Gsdme in GSDME-expressing tumors enhanced, whereas ectopic expression in Gsdme-repressed tumors inhibited, tumor growth. This tumor suppression was mediated by killer cytotoxic lymphocytes: it was abrogated in perforin (170280)-deficient mice or mice depleted of killer lymphocytes. GSDME expression enhanced the phagocytosis of tumor cells by tumor-associated macrophages, as well as the number and functions of tumor-infiltrating natural killer and CD8+ T cells. Killer cell granzyme B (123910) also activated caspase-independent pyroptosis in target cells by directly cleaving GSDME at the same site as CASP3. Uncleavable or pore-defective GSDME proteins were not tumor-suppressive. Thus, tumor GSDME acts as a tumor suppressor by activating pyroptosis, enhancing antitumor immunity.


Molecular Genetics

Van Laer et al. (1998) identified a mutation in intron 7 of the DFNA5 gene (608798.0001) that cosegregated with nonsyndromic deafness in the Dutch family reported by Van Camp et al. (1995) and Van Laer et al. (1997); see 600994. The insertion/deletion mutation did not affect intron-exon boundaries, but deleted 5 G-triplets at the 3-prime end of the intron, causing skipping of exon 8. The absence of exon 8 resulted in a frameshift starting at amino acid 330, which introduced an aberrant stretch of 41 amino acids followed by a stop codon that prematurely terminated the protein.

Age-related hearing impairment (ARHI), or presbycusis, is a sensorineural high frequency hearing loss that is believed to result from an interaction between environmental and genetic factors. Because the hearing loss in the DFNA5 family closely resembles that observed in ARHI, Van Laer et al. (2002) performed linkage analysis on a quantitative measure of high frequency hearing loss in 328 pedigrees from the National Heart Lung and Blood Institute's Framingham Heart Study. No significant linkage between ARHI and microsatellite markers from the DFNA5 region was detected. Sequencing of the DFNA5 coding region in 10 random individuals resulted in the detection of 6 SNPs. Two of these SNPs led to amino acid substitutions (P142H and V207M) and were selected for further analysis. A pilot experiment on 116 random Belgian and Dutch subjects was performed, but no association could be detected between either SNP and ARHI. Subsequently, 93 ARHI cases and 83 controls selected from the Framingham cohort were genotyped for P142H, but no allelic or genotypic association was detected. Van Laer et al. (2002) concluded that possibly many different genes contribute to ARHI, each with a weak effect, and ARHI might turn out to be too complex to study with moderate sample sizes.

In a Chinese family with autosomal dominant nonsyndromic sensorineural deafness, Yu et al. (2003) identified a 3-bp deletion in the DFNA5 gene (608798.0002) that segregated with the phenotype and was predicted to cause skipping of exon 8, like the DFNA5 mutation previously reported by Van Laer et al. (1998).

In a 5-generation Dutch family with autosomal dominant deafness, Bischoff et al. (2004) identified a splice-site mutation in the DFNA5 gene (608798.0003) that caused skipping of exon 8. Because of the relatively low amount of short transcript in affected members in this family, the authors suggested that the mutation might have a dominant-negative effect rather than haploinsufficiency.

In transfection studies using HEK293T cells, Van Laer et al. (2004) found that posttransfection cell death approximately doubled when cells were transfected with mutant DFNA5-GFP compared to wildtype DFNA5-GFP, with cell death being attributed to necrotic rather than apoptotic events. This information, coupled with the observation that all 3 identified mutations in DFNA5 result in skipping of exon 8 at the mRNA level, led Van Laer et al. (2004) to propose that the hearing impairment associated with DFNA5 is caused by a gain-of-function mutation.

Cheng et al. (2007) reported a 5-generation Chinese family with autosomal late-onset deafness in which they identified a heterozygous splice site mutation resulting in skipping of exon 8 as confirmed by RT-PCR analysis.

In a 5-generation Iranian family segregating nonsyndromic autosomal dominant sensorineural hearing loss that appeared to map to the DFNA5 locus, Van Laer et al. (2007) identified a heterozygous truncating mutation in exon 5 of the DFNA5 gene. However, further analysis revealed that the mutation did not segregate with hearing loss in this family, and linkage to a locus on chromosome 4 was subsequently found. The authors stated that these findings supported the hypothesis that only a specific gain-of-function mutation caused by skipping of exon 8 can lead to DFNA5-associated hearing loss.

Wang et al. (2017) found that mutant GSDME protein resulting from skipping of exon 8 was unstable and triggered extensive pyroptosis when expressed in 293T cells.

Using 2 next-generation sequencing platforms, Booth et al. (2018) identified 5 families with autosomal dominant postlingual progressive nonsyndromic hearing loss with 3 novel and 2 recurrent mutations in the DFNA5 gene. The 3 novel mutations were missense mutations within exon 8 that were predicted to reduce the efficiency of, or abolish, splicing (see, e.g., Q368E, 608798.0005). Functional impact of these 3 mutations was confirmed in vitro using minigenes. The authors noted that previously overlooked silent mutations within exon 8 could alter splicing; thus, families with high frequency progressive hearing loss that links to the DFNA5 gene should be evaluated for variants in the flanking introns and in the exon itself.


Animal Model

Wang et al. (2017) found that Gsdme -/- mice developed normally. Compared with wildtype, Gsdme -/- mice were refractory to cisplatin- or bleomycin-induced injury of gastrointestinal tissues, spleen, and lung.


ALLELIC VARIANTS 5 Selected Examples):

.0001   DEAFNESS, AUTOSOMAL DOMINANT 5

GSDME, INS/DEL, EX8DEL
ClinVar: RCV000002171

In an extended Dutch family with autosomal dominant deafness (DFNA5; 600994), Van Laer et al. (1998) found that affected members had a deletion of 1,189 bp from intron 7 of the DFNA5 gene and insertion of 127 bp from intron 8 into intron 7 in the opposite direction, followed by a GCCCA stretch of unknown origin. Exons 7 and 8, the intron-exon boundaries, and the branchpoint sequence from intron 7 were still present. It was demonstrated that exon 8 (193 bp) was skipped in affected family members. The absence of exon 8 resulted in a frameshift starting at amino acid 330, which introduced an aberrant stretch of 41 amino acids followed by a stop codon that prematurely terminated the protein. The aberrant splicing was thought to be caused by deletion of the 5 G-triplets in intron 7. There is evidence from other sources that sequences other than the intron/exon consensus and the branchpoint are involved in splice site selection. In particular, intronic G-triplets play an important role.


.0002   DEAFNESS, AUTOSOMAL DOMINANT 5

GSMDE, 3-BP DEL, IVS7, -17CTT
SNP: rs727505273, gnomAD: rs727505273, ClinVar: RCV000156800, RCV001092560, RCV001808425

In affected members of a family with autosomal dominant nonsyndromic sensorineural deafness (DFNA5; 600994), Yu et al. (2003) identified a mutation in the DFNA5 gene, a CTT deletion in the polypyrimidine tract of intron 7, which was predicted to create a shift in the reading frame and introduce a stop codon at position 372. This mutation, like the previously reported mutation (600994.0001), led to skipping of exon 8 of DFNA5. Yu et al. (2003) confirmed the existence of a previously identified short isoform of DFNA5 and concluded that the 3-nucleotide deletion in their family did not affect the function of this short isoform.

Park et al. (2010) identified the 3-bp deletion in intron 7, which they referred to as 991-15_991-13del, in affected members of a Korean family with nonsyndromic sensorineural deafness. Haplotype analysis showed that this family and the Chinese family reported by Yu et al. (2003) shared a common haplotype, suggesting a founder effect.


.0003   DEAFNESS, AUTOSOMAL DOMINANT 5

GSMDE, IVS7AS, C-G, -6
SNP: rs1562687726, ClinVar: RCV000002173

In affected members of a 5-generation Dutch family with autosomal dominant deafness (DFNA5; 600994), Bischoff et al. (2004) identified a 1200C-G transversion at position -6 of the splice acceptor site of intron 7 of the DFNA5 gene. This results in skipping of exon 8, which leads to 41 aberrant codons followed by a premature stop codon. The C-to-G transversion creates an RsaI restriction site, which was used to demonstrate cosegregation of the mutation in the family. Because of the relatively low amount of the short transcript in affected members in this family, Bischoff et al. (2004) suggested that this may represent a dominant-negative effect rather than haploinsufficiency.


.0004   DEAFNESS, AUTOSOMAL DOMINANT 5

GSMDE, IVS8DS, A-G, +4
SNP: rs1562687295, ClinVar: RCV000002174

In affected members of a large 5-generation Chinese family with autosomal dominant late-onset deafness (DFNA5; 600994), Cheng et al. (2007) identified a heterozygous splice site mutation in intron 8 of the DFNA5 gene, resulting in skipping of exon 8 as confirmed by RT-PCR analysis. The resultant protein was predicted to be prematurely terminated. The mutation was absent in 100 control chromosomes. Cheng et al. (2007) noted that all of the pathogenic mutations described in the DFNA5 gene result in skipping of exon 8, suggesting a very specific gain-of-function effect.


.0005   DEAFNESS, AUTOSOMAL DOMINANT 5

GSMDE, GLN368GLU
SNP: rs2128047712, ClinVar: RCV001726686

In affected members of a European family (CDS-6824) with autosomal dominant postlingual progressive nonsyndromic hearing loss (DFNA5; 600994), Booth et al. (2018) identified a c.1102C-G transversion (c.1102C-G, NM_004403.2) in exon 8 of the DFNA5 gene, resulting in a gln368-to-glu (Q368E) substitution at a conserved residue, that segregated the phenotype. The variant was predicted to alter splicing due to creation of both a new cryptic donor site and cryptic acceptor site. In vitro analysis using a minigene with exon 8 of DFNA5 showed that the variant causes skipping of exon 8. The mutation was not present in the ExAC (v.0.3) or gnomAD (v.2.0.2) databases.


REFERENCES

  1. Bischoff, A. M. L. C., Luijendijk, M. W. J., Huygen, P. L. M., van Duijnhoven, G., De Leenheer, E. M. R., Oudesluijs, G. G., Van Laer, L., Cremers, F. P. M., Cremers, C. W. R. J., Kremer, H. A novel mutation identified in the DFNA5 gene in a Dutch family: a clinical and genetic evaluation. Audiol. Neurootol. 9: 34-46, 2004. [PubMed: 14676472] [Full Text: https://doi.org/10.1159/000074185]

  2. Booth, K. T., Azaiez, H., Kahrizi, K., Wang, D., Zhang, Y., Frees, K., Nishimura, C., Najmabadi, H., Smith, R. J. Exonic mutations and exon skipping: lessons learned from DFNA5. Hum. Mutat. 39: 433-440, 2018. [PubMed: 29266521] [Full Text: https://doi.org/10.1002/humu.23384]

  3. Cheng, J., Han, D. Y., Dai, P., Sun, H. J., Tao, R., Sun, Q., Yan, D., Qin, W., Wang, H. Y., Ouyang, X. M., Yang, S. Z., Cao, J. Y., Feng, G. Y., Du, L. L., Zhang, Y. Z., Zhai, S. Q., Yang, W. Y., Liu, X. Z., He, L., Yuan, H. J. A novel DFNA5 mutation, IVS8+4A-G, in the splice donor site of intron 8 causes late-onset non-syndromic hearing loss in a Chinese family. Clin. Genet. 72: 471-477, 2007. [PubMed: 17868390] [Full Text: https://doi.org/10.1111/j.1399-0004.2007.00889.x]

  4. Masuda, Y., Futamura, M., Kamino, H., Nakamura, Y., Kitamura, N., Ohnishi, S., Miyamoto, Y., Ichikawa, H., Ohta, T., Ohki, M., Kiyono, T., Egami, H., Baba, H., Arakawa, H. The potential role of DFNA5, a hearing impairment gene, in p53-mediated cellular response to DNA damage. J. Hum. Genet. 51: 652-664, 2006. [PubMed: 16897187] [Full Text: https://doi.org/10.1007/s10038-006-0004-6]

  5. Op de Beeck, K. O., Van Camp, G., Thys, S., Cools, N., Callebaut, I., Vrijens, K., Van Nassauw, L., Van Tendeloo, V. F., Timmermans, J. P., Van Laer, L. The DFNA5 gene, responsible for hearing loss and involved in cancer, encodes a novel apoptosis-inducing protein. Europ. J. Hum. Genet. 19: 965-973, 2011. [PubMed: 21522185] [Full Text: https://doi.org/10.1038/ejhg.2011.63]

  6. Park, H.-J., Cho, H.-J., Baek, J.-I., Ben-Yosef, T., Kwon, T.-J., Griffith, A. J., Kim, U.-K. Evidence for a founder mutation causing DFNA5 hearing loss in East Asians. J. Hum. Genet. 55: 59-62, 2010. [PubMed: 19911014] [Full Text: https://doi.org/10.1038/jhg.2009.114]

  7. Rogers, C., Fernandes-Alnemri, T., Mayes, L., Alnemri, D., Cingolani, G., Alnemri, E. S. Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death. Nature Commun. 8: 14128, 2017. Note: Electronic Article. [PubMed: 28045099] [Full Text: https://doi.org/10.1038/ncomms14128]

  8. Thompson, D. A., Weigel, R. J. Characterization of a gene that is inversely correlated with estrogen receptor expression (ICERE-1) in breast carcinomas. Europ. J. Biochem. 252: 169-177, 1998. [PubMed: 9523727] [Full Text: https://doi.org/10.1046/j.1432-1327.1998.2520169.x]

  9. Van Camp, G., Coucke, P., Balemans, W., Van Velzen, D., Van de Bilt, C., Van Laer, L., Smith, R. J. H., Fukushima, K., Padberg, G. W., Frants, R. R., Van de Heyning, P., Smith, S. D., Huizing, E. H., Willems, P. J. Localization of a gene for non-syndromic hearing loss (DFNA5) to chromosome 7p15. Hum. Molec. Genet. 4: 2159-2163, 1995. [PubMed: 8589696] [Full Text: https://doi.org/10.1093/hmg/4.11.2159]

  10. Van Laer, L., DeStefano, A. L., Myers, R. H., Flothmann, K., Thys, S., Fransen, E., Gates, G. A., Van Camp, G., Baldwin, C. T. Is DFNA5 a susceptibility gene for age-related hearing impairment? Europ. J. Hum. Genet. 10: 883-886, 2002. [PubMed: 12461698] [Full Text: https://doi.org/10.1038/sj.ejhg.5200878]

  11. Van Laer, L., Huizing, E. H., Verstreken, M., van Zuijlen, D., Wauters, J. G., Bossuyt, P. J., Van de Heyning, P., McGuirt, W. T., Smith, R. J. H., Willems, P. J., Legan, P. K., Richardson, G. P., Van Camp, G. Nonsyndromic hearing impairment is associated with a mutation in DFNA5. Nature Genet. 20: 194-197, 1998. [PubMed: 9771715] [Full Text: https://doi.org/10.1038/2503]

  12. Van Laer, L., Meyer, N. C., Malekpour, M., Riazalhosseini, Y., Moghannibashi, M., Kahrizi, K., Vandevelde, A., Alasti, F., Najmabadi, H., Van Camp, G., Smith, R. J. H. A novel DFNA5 mutation does not cause hearing loss in an Iranian family. J. Hum. Genet. 52: 549-552, 2007. [PubMed: 17427029] [Full Text: https://doi.org/10.1007/s10038-007-0137-2]

  13. Van Laer, L., Van Camp, G., van Zuijlen, D., Green, E. D., Verstreken, M., Schatteman, I., Van de Heyning, P., Balemans, W., Coucke, P., Greinwald, J. H., Smith, R. J. H., Huizing, E., Willems, P. Refined mapping of a gene for autosomal dominant progressive sensorineural hearing loss (DFNA5) to a 2-cM region, and exclusion of a candidate gene that is expressed in the cochlea. Europ. J. Hum. Genet. 5: 397-405, 1997. [PubMed: 9450185]

  14. Van Laer, L., Vrijens, K., Thys, S., Van Tendeloo, V. F. I., Smith, R. J. H., Van Bockstaele, D. R., Timmermans, J.-P., Van Camp, G. DFNA5: hearing impairment exon instead of hearing impairment gene? J. Med. Genet. 41: 401-406, 2004. [PubMed: 15173223] [Full Text: https://doi.org/10.1136/jmg.2003.015073]

  15. Wang, Y., Gao, W., Shi, X., Ding, J., Liu, W., He, H., Wang, K., Shao, F. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 547: 99-103, 2017. [PubMed: 28459430] [Full Text: https://doi.org/10.1038/nature22393]

  16. Yu, C., Meng, X., Zhang, S., Zhao, G., Hu, L., Kong, X. A 3-nucleotide deletion in the polypyrimidine tract of the intron 7 of the DFNA5 gene causes nonsyndromic hearing impairment in a Chinese family. Genomics 82: 575-579, 2003. [PubMed: 14559215] [Full Text: https://doi.org/10.1016/s0888-7543(03)00175-7]

  17. Zhang, Z., Zhang, Y., Xia, S., Kong, Q., Li, S., Liu, X., Junqueira, C., Meza-Sosa, K. F., Mok, T. M. Y., Ansara, J., Sengupta, S., Yao, Y., Wu, H., Lieberman, J. Gasdermin E suppresses tumour growth by activating anti-tumour immunity. Nature 579: 415-420, 2020. [PubMed: 32188940] [Full Text: https://doi.org/10.1038/s41586-020-2071-9]


Contributors:
Sonja A. Rasmussen - updated : 10/01/2021
Ada Hamosh - updated : 06/17/2020
Matthew B. Gross - updated : 10/03/2017
Matthew B. Gross - updated : 08/22/2017
Patricia A. Hartz - updated : 08/22/2017
Patricia A. Hartz - updated : 9/13/2011
Cassandra L. Kniffin - updated : 6/14/2010
Marla J. F. O'Neill - updated : 6/1/2009
Cassandra L. Kniffin - updated : 8/21/2008
Patricia A. Hartz - updated : 10/23/2006

Creation Date:
Victor A. McKusick : 7/13/2004

Edit History:
alopez : 06/23/2023
alopez : 03/13/2023
carol : 10/01/2021
alopez : 06/17/2020
carol : 10/04/2017
mgross : 10/03/2017
mgross : 08/22/2017
mgross : 08/22/2017
alopez : 10/04/2016
carol : 08/14/2015
mgross : 9/13/2011
terry : 9/13/2011
wwang : 6/18/2010
ckniffin : 6/14/2010
wwang : 6/2/2009
terry : 6/1/2009
terry : 12/2/2008
alopez : 12/1/2008
wwang : 9/2/2008
ckniffin : 8/21/2008
carol : 10/25/2006
carol : 10/23/2006
carol : 10/23/2006
carol : 7/13/2004



-