Entry - *191041 - TROPONIN T1, SKELETAL, SLOW; TNNT1 - OMIM
* 191041

TROPONIN T1, SKELETAL, SLOW; TNNT1


Alternative titles; symbols

TROPONIN T


HGNC Approved Gene Symbol: TNNT1

Cytogenetic location: 19q13.42     Genomic coordinates (GRCh38): 19:55,132,698-55,149,206 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
19q13.42 Nemaline myopathy 5A, autosomal recessive, severe infantile 605355 AR 3
Nemaline myopathy 5B, autosomal recessive, childhood-onset 620386 AR 3
Nemaline myopathy 5C, autosomal dominant 620389 AD 3

TEXT

Description

The TNNT1 gene encodes the slow skeletal muscle troponin. The sarcomere, the contractile element in muscle, including the myocardium, is constituted by 7 major proteins and several minor ones organized into thin and thick filaments. Each tropomyosin dimer (TPM1, 191010; TPM2, 190990) interacts with 7 actins and is associated with a troponin complex. Each complex is composed of 1 molecule of each of the 3 troponins: T, C, and I. This tropomyosin-troponin complex is responsible for the calcium sensitivity of the contractile apparatus (Nadal-Ginard and Mahdavi, 1989), and thus plays an important role in linking excitation to contraction in skeletal muscle.


Cloning and Expression

Samson et al. (1994) concluded that there are 4 isoforms of human slow skeletal troponin-T mRNA and that these probably result from combinatorial alternative splicing of a single gene.

By PCR of adult human skeletal muscle total RNA, Barton et al. (1999) cloned full-length TNNT1 and identified 3 shorter splice variants. The full-length protein contains 278 amino acids. Northern blot analysis detected TNNT1 expression in adult skeletal muscle, but not in adult heart and liver or in fetal heart.

Alternative splicing of exon 5 of the TNNT1 gene generates a high molecular weight (HMW) isoform (with exon 5) and a low molecular weight isoform (LMW) isoform (without exon 5). LMW TNNT1 has a higher binding affinity for tropomyosin and generates more calcium-activated contractile force than the HMW isoform, which enables regulation and modulation of muscle contractility. Normal adult slow muscles express mainly the HMW isoform (summary by Holling et al., 2022).


Gene Structure

Barton et al. (1999) determined that the TNNT1 gene contains 14 exons and spans more than 16 kb. The first exon is noncoding.


Mapping

Samson et al. (1990) assigned the slow skeletal isoform of troponin T (TNNT1) to chromosome 19q13.3-q13.4 using a panel of somatic cell hybrids and a cDNA clone as probe.

Novelli et al. (1991, 1992) confirmed the assignment of the TNNT1 gene to human chromosome 19 by showing the presence of a specific PCR product in hybrids retaining chromosome 19. The cardiac isoform of troponin-I (TNNI3; 191044) maps to the same region.

Samson et al. (1991) achieved regional assignment to the 19q13.2-qter region by analysis of 7 somatic cell hybrids containing different portions of chromosome 19. They excluded TNNT1 as a candidate gene for myotonic dystrophy (DM1; 160900) by the finding of obligate recombination events in family linkage studies.

Samson et al. (1992) mapped the TNNT1 gene to chromosome 19q13.4 by study of somatic cell hybrids and fluorescence in situ hybridization.

Barton et al. (1999) determined that the TNNI3 gene and the TNNT1 gene are oriented head to tail, with the TNNI3 gene 2.6 kb upstream of exon 1 of the TNNT1 gene.


Molecular Genetics

Autosomal Recessive Severe Infantile Nemaline Myopathy 5A

In Amish patients with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355), Johnston et al. (2000) identified homozygosity for a nonsense mutation in the TNNT1 gene (E180X; 191041.0001).

In a Dutch boy with NEM5A, van der Pol et al. (2014) identified compound heterozygous mutations in the TNNT1 gene: a splice site mutation (191041.0002) and an intragenic deletion (ex14del; 191041.0003). The mutations were inherited from the unaffected parents. Homozygosity for the splice site mutation was identified in 2 paternal cousins with a similar disorder who died in childhood of respiratory insufficiency.

In 3 unrelated patients with NEM5A, Geraud et al. (2021) identified homozygous or compound heterozygous mutations in the TNNT1 gene (see, e.g., 191041.0004-191041.0006). Western blot analysis of skeletal muscle from the patients showed absence of the TNNT1 protein. All died by 29 months of age.

Autosomal Recessive Childhood-Onset Nemaline Myopathy 5B

In 4 patients from 3 unrelated French Canadian families with autosomal recessive childhood-onset nemaline myopathy-5B (NEM5B; 620386), Pellerin et al. (2020) identified a homozygous missense mutation in the TNNT1 gene (L96P; 191041.0007). The mutation, which was found by next-generation sequencing and confirmed by Sanger sequencing, segregated with the disorder in 3 of the families who underwent testing. The mutation was unable to rescue the abnormal muscle phenotype in tnnt1-null zebrafish, suggesting that it causes a loss of function. The authors postulated that there was some residual activity or compensation by the low molecular weight TNNT1 isoform, resulting in a milder phenotype compared to patients with complete loss of TNNT1.

In 2 brothers, born of unrelated Korean parents, with NEM5B, Lee et al. (2022) identified compound heterozygosity for a missense mutation (A242P; 191041.0008) and a splice site mutation (191041.0009) in the TNNT1 gene. The mutations, which were found by whole-exome sequencing and confirmed by Sanger sequencing, were inherited from the unaffected parents. The splice site mutation was demonstrated to result in altered splicing and frameshifts, and the A242P mutation was unable to rescue the myopathic phenotype in tnnt1-null zebrafish, suggesting that both mutations caused a loss of function.

In a 51-year-old man with NEM5B, Martin-Jimenez et al. (2022) detected homozygosity for a variant in the TNNT1 gene (c.551_552delinsCA) that resulted in an arg184-to-pro (R184P) substitution. The variant was absent from gnomAD. Segregation studies and functional studies were not performed. The patient presented at age 50 years with a respiratory infection followed by respiratory arrest requiring intubation. He subsequently developed muscle weakness in all limbs which was slowly progressive and resulted in difficulty climbing stairs.

Autosomal Dominant Nemaline Myopathy 5C

In 8 affected members of a large consanguineous Ashkenazi Jewish family with autosomal dominant nemaline myopathy-5C (NEM5C; 620389) originally reported by Spiro and Kennedy (1965) and Gonatas et al. (1966), Konersman et al. (2017) identified a heterozygous missense mutation in the TNNT1 gene (E104V; 191041.0010). The mutation, which was found by Sanger sequencing of candidate genes, segregated with the disorder in the family. Holling et al. (2022) stated that the E104V mutation affects a residue just outside of the tropomyosin-binding site 1. In vitro studies in HEK293 cells transfected with the mutation showed normal expression of TNNT1 and effective binding to tropomyosin-3 (TPM3; 191030), although binding of the mutant protein to TPM3 was reduced by 35% in the presence of calcium.

In a mother and son with NEM5C, Holling et al. (2022) identified a heterozygous missense mutation in the TNNT1 gene (D65A; 191041.0011) that occurred within the tropomyosin-binding site 1 region. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the gnomAD database. In vitro studies in HEK293 cells transfected with the mutation showed normal expression of TNNT1, whereas coimmunoprecipitation studies showed significantly increased binding of the D65A variant to TPM3 compared to controls, both in the presence and absence of calcium.


Animal Model

Fox et al. (2018) described muscle features in 2 mouse models of Tnnt1 deficiency: a targeted gene knockout (Tnnt1 -/-) model and a site-specific knock-in (Tnnt1 c.505G-T) model. Quadratus femoris muscle tissue showed absence of Tnnt1, atrophy/hypotrophy of small type 1 myofibers, hypertrophy of fast type 2 fibers, and the presence of nemaline rods in both mouse models. Both models also showed impaired muscle force generation and exercise recovery in soleus muscles.

Pellerin et al. (2020) found that morpholino knockout of the tnnt1 gene in zebrafish caused reduced muscle mass, reduced birefringence, and severe truncal curvature compared to controls. Myofibers were disorganized and showed corelike intramyofibrillar areas of sarcomeric disorganization and Z-line thickening. These defects could be rescued by expression of wildtype human TNNT1 mRNA.


ALLELIC VARIANTS ( 11 Selected Examples):

.0001 NEMALINE MYOPATHY 5A, AUTOSOMAL RECESSIVE, SEVERE INFANTILE

TNNT1, GLU180TER
  
RCV000020554...

In affected individuals with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355), also known as Amish nemaline myopathy, Johnston et al. (2000) identified a homozygous c.579G-T transversion in exon 11 of the TNNT1 gene, resulting in a stop codon at amino acid 180 (E180X) and loss of 83 C-terminal residues. Johnston et al. (2000) had treated or obtained clinical information on 71 infants and young children from 33 nuclear Amish families with this form of nemaline myopathy. In the first months of life, affected infants had tremors with hypotonia and mild contractures of the shoulders and hips. Progressive worsening of the proximal contractures, weakness, and a pectus carinatum deformity developed before the children died of respiratory insufficiency, usually in the second year.

Variant Function

Jin et al. (2003) found complete loss of the TNNT1 protein in patients with the E180X mutation. The truncated protein results in elimination of the C-terminal T2 domain that interacts with troponin C (TNNC1; 191040), I (TNNI2; 191043), and tropomyosin (TPM1; 191010), but was predicted to retain a central tropomyosin-binding site that participates in the anchoring of troponin complex to the thin actin filament. If residual truncated protein was produced, this could potentially result in a dominant-negative effect. The significant muscle atrophy observed in the disorder was consistent with the slow Tnt isoform being involved in muscle development and growth.

In a patient with Amish nemaline myopathy due to the E180X mutation, Wang et al. (2005) detected residual mutant TNNT1 mRNA in muscle tissue but no corresponding translated mutant TNNT1 protein. In vitro functional expression studies in nonmuscle cells showed that E180X-mutant protein could be produced but was not detectable when expressed in muscle cells in vitro. The findings suggested rapid degradation of E180X-mutant protein in muscle cells, rather than a loss of nonsense mRNA to explain the absence of a dominant effect. Wang et al. (2005) postulated that inefficient incorporation of mutant TNNT1 into myofilaments results in its degradation by the muscle cell as a protective mechanism.


.0002 NEMALINE MYOPATHY 5A, AUTOSOMAL RECESSIVE, SEVERE INFANTILE

TNNT1, IVS8DS, G-A, +1
  
RCV001529807...

In a Dutch boy with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355), van der Pol et al. (2014) identified compound heterozygous mutations in the TNNT1 gene: a G-to-A transition in intron 8 (c.309+1G-A, NM_003283.4) inherited from the unaffected father, and an exon 14 deletion (ex14del; 191041.0003) inherited from the unaffected mother. Homozygosity for the c.309+1G-A mutation was identified in 2 paternal cousins with a similar disorder who died in childhood of respiratory insufficiency. The splicing defect resulted in the skipping of exon 8 with an in-frame deletion of 39 functionally important residues from the TNNT1 troponin domain (Asp65_Ile103del). The mutations were predicted to result in a loss of function, although the authors noted that the predicted transcripts could be translated into shortened proteins with some residual activity.


.0003 NEMALINE MYOPATHY 5A, AUTOSOMAL RECESSIVE, SEVERE INFANTILE

TNNT1, EX14 DEL
   RCV003230310

For discussion of the exon 14 deletion (ex14del, NM_003283.4) in the TNNT1 gene that was found in compound heterozygous state in a patient with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355) by van der Pol et al. (2014), see 191041.0002.


.0004 NEMALINE MYOPATHY 5A, AUTOSOMAL RECESSIVE, SEVERE INFANTILE

TNNT1, GLU112TER
   RCV003230311

In a female infant (patient 2) with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355) resulting in death at 16 months of age, Geraud et al. (2021) identified compound heterozygous mutations in the TNNT1 gene: a c.334G-T transversion (c.334G-T, NM_003283.5), resulting in a glu112-to-ter (E112X) substitution, and a C-to-A transversion in intron 4 (c.74-67C-A; 191041.0005), predicted to result in a splicing defect. Western blot analysis of skeletal muscle from the patient showed absence of the TNNT1 protein. She had a severe form of the disorder with tongue fasciculations, kyphosis, pectus carinatum, and hypotonia.


.0005 NEMALINE MYOPATHY 5A, AUTOSOMAL RECESSIVE, SEVERE INFANTILE

TNNT1, IVS4AS, C-A, -67
   RCV003226144

For discussion of the c.74-67C-A mutation (c.74-67C-A, NM_003283.5) in intron 4 of the TNNT1 gene, resulting in a splicing defect, that was found in compound heterozygous state in a patient with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355) by Geraud et al. (2021) see 191041.0004.


.0006 NEMALINE MYOPATHY 5A, AUTOSOMAL RECESSIVE, SEVERE INFANTILE

TNNT1, GLU6TER
   RCV003230312

In a patient (patient 3) with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355) resulting in death at 6 months of age, Geraud et al. (2021) identified a homozygous c.16G-T transversion (cc.74-67C-A, NM_003283.5) in the TNNT1 gene, resulting in a glu6-to-ter (E6X) substitution. Western blot analysis of patient skeletal muscle showed absence of the TNNT1 protein.


.0007 NEMALINE MYOPATHY 5B, AUTOSOMAL RECESSIVE, CHILDHOOD-ONSET

TNNT1, LEU96PRO
   RCV003230314

In 4 patients from 3 unrelated French Canadian families with autosomal recessive childhood-onset nemaline myopathy-5B (NEM5B; 620386), Pellerin et al. (2020) identified a homozygous c.287T-C transition (c.287T-C, NM_003283.6) in exon 8 of the TNNT1 gene, resulting in a leu96-to-pro (L96P) substitution at a conserved residue. The mutation, which was found by next-generation sequencing and confirmed by Sanger sequencing, segregated with the disorder in 3 of the families who underwent testing. The mutation was predicted to disrupt the alpha-helical secondary structure of the tropomyosin-binding site 1 and impair binding affinity. The mutant protein was likely to be misincorporated into myofilaments and undergo partial degradation, as evidenced by lower levels of the high molecular weight troponin T isoform in patient muscle; low molecular weight isoforms were retained. The mutation was unable to rescue the abnormal muscle phenotype in tnnt1-null zebrafish, suggesting that it causes a loss of function. The authors postulated that there was some residual activity or compensation by the low molecular weight isoform, resulting in a milder phenotype compared to patients with complete loss of TNNT1.

Variant Function

Holling et al. (2022) found that expression of the L96P mutation was increased compared to controls when transfected into HEK293 cells. Coimmunoprecipitation studies showed significantly reduced interaction (by 88%) of the L96P mutant with tropomyosin-3 (TPM3; 191030), and this interaction was abrogated in the presence of calcium.


.0008 NEMALINE MYOPATHY 5B, AUTOSOMAL RECESSIVE, CHILDHOOD-ONSET

TNNT1, ALA242PRO
   RCV003230315

In 2 brothers, born of unrelated Korean parents, with autosomal recessive childhood-onset nemaline myopathy-5B (NEM5B; 620386), Lee et al. (2022) identified compound heterozygous mutations in the TNNT1 gene: a c.724G-C transversion (c.724G-C, NM_003283) in exon 12, resulting in an ala242-to-pro (A242P) substitution in the conserved troponin I-binding domain, and a G-to-A transition in intron 11 (c.611+1G-A; 191041.0009), resulting in a splicing defect and frameshifts. The mutations, which were found by whole-exome sequencing and confirmed by Sanger sequencing, were inherited from the unaffected parents. The splice site mutation was found once in the heterozygous state in a Korean individual in gnomAD (1 of 249,306 alleles, frequency of 4.01 x 10(-6)). The A242P mutation was unable to rescue the myopathic phenotype in tnnt1-null zebrafish, suggesting that this mutation causes a loss of function.


.0009 NEMALINE MYOPATHY 5B, AUTOSOMAL RECESSIVE, CHILDHOOD-ONSET

TNNT1, IVS11DS, G-A, +1
   RCV003230313

For discussion of the G-to-A transition in intron 11 of the TNNT1 gene (c.611+1G-A, NM_003283), resulting in a splicing defect and frameshifts, that was found in compound heterozygous state in 2 brothers with autosomal recessive childhood-onset nemaline myopathy-5B (NEM5B; 620386) by Lee et al. (2022), see 191041.0008.


.0010 NEMALINE MYOPATHY 5C, AUTOSOMAL DOMINANT

TNNT1, GLU104VAL
  
RCV001209655...

In 8 affected members of a large consanguineous Ashkenazi Jewish family with autosomal dominant nemaline myopathy-5C (NEM5C; 620389) originally reported by Spiro and Kennedy (1965) and Gonatas et al. (1966), Konersman et al. (2017) identified a heterozygous c.311A-T transversion (c.311A-T, NM_003283.4) in exon 9 of the TNNT1 gene, resulting in a glu104-to-val (E104V) substitution at a highly conserved residue in the tropomyosin-binding site 1 region. The mutation, which was found by Sanger sequencing of candidate genes, segregated with the disorder in the family. It was not present in public databases, including the Exome Variant Server, 1000 Genomes Project, and ExAC. Muscle samples from 2 patients showed normal levels of the TNNT1 protein, although low molecular weight (LMW) TNNT1 transcripts and protein levels were increased compared to controls, suggesting a compensatory mechanism. The authors postulated that the mutation could affect the affinity of TNNT1 for tropomyosin.

Variant Function

Holling et al. (2022) stated that the E104V mutation affects a residue just outside of the tropomyosin-binding site 1. In vitro studies in HEK293 cells transfected with the mutation showed normal expression of TNNT1 and effective binding to tropomyosin-3 (TPM3; 191030), although binding of the mutant protein to TPM3 was reduced by 35% in the presence of calcium.


.0011 NEMALINE MYOPATHY 5C, AUTOSOMAL DOMINANT

TNNT1, ASP65ALA
   RCV003230316

In a mother and son with autosomal dominant nemaline myopathy-5C (NEM5C; 620389), Holling et al. (2022) identified a heterozygous c.194A-C transversion (c.194A-C, NM_003283.6) in exon 8 of the TNNT1 gene, resulting in an asp65-to-ala (D65A) substitution in the tropomyosin-binding site 1 region. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the gnomAD database. Genetic studies of the mother's parents showed that the mutation occurred de novo in the mother. The total amount of TNNT1 protein in the mother's skeletal muscle was similar to controls. Muscle biopsy from the mother showed type 1 fiber hypotrophy, type 2 fiber hypertrophy, increased amounts of fast skeletal fiber myosin-2a (MYH2; 160740), and increased actin levels, which may be compensatory mechanisms. In vitro studies in HEK293 cells transfected with the mutation showed normal expression of TNNT1, and coimmunoprecipitation studies showed significantly increased binding to tropomyosin-3 (TPM3; 191030), both in the presence and absence of calcium.


See Also:

REFERENCES

  1. Barton, P. J. R., Cullen, M. E., Townsend, P. J., Brand, N. J., Mullen, A. J., Norman, D. A. M., Bhavsar, P. K., Yacoub, M. H. Close physical linkage of human troponin genes: organization, sequence, and expression of the locus encoding cardiac troponin I and slow skeletal troponin T. Genomics 57: 102-109, 1999. [PubMed: 10191089, related citations] [Full Text]

  2. Fox, M. D., Carson, V. J., Feng, H.-Z., Lawlor, M. W., Gray, J. T., Brigatti, K. W., Jin, J.-P., Strauss, K. A. TNNT1 nemaline myopathy: natural history and therapeutic frontier. Hum. Molec. Genet. 27: 3272-3282, 2018. [PubMed: 29931346, images, related citations] [Full Text]

  3. Geraud, J., Dieterich, K., Rendu, J., Uro Coste, E., Dobrzynski, M., Marcorelle, P., Ioos, C., Romero, N. B., Baudou, E., Brocard, J., Coville, A.-C., Faure, J., and 10 others. Clinical phenotype and loss of the slow skeletal muscle troponin T in three new patients with recessive TNNT1 nemaline myopathy. J. Med. Genet. 58: 602-608, 2021. [PubMed: 32994279, images, related citations] [Full Text]

  4. Gonatas, N. K., Shy, G. M., Godfrey, E. H. Nemaline myopathy: the origin of nemaline structures. New Eng. J. Med. 274: 535-539, 1966. [PubMed: 5908457, related citations] [Full Text]

  5. Holling, T., Lisfeld, J., Johannsen, J., Matschke, J., Song, F., Altmeppen, H. C., Kutsche, K. Autosomal dominantly inherited myopathy likely caused by the TNNT1 variant p.(Asp65Ala). Hum. Mutat. 43: 1224-1233, 2022. [PubMed: 35510366, related citations] [Full Text]

  6. Jin, J.-P., Brotto, M. A., Hossain, M. M., Huang, Q.-Q., Brotto, L. S., Nosek, T. M., Morton, D. H., Crawford, T. O. Truncation by glu180 nonsense mutation results in complete loss of skeletal muscle troponin T in a lethal nemaline myopathy. J. Biol. Chem. 278: 26159-26165, 2003. [PubMed: 12732643, related citations] [Full Text]

  7. Johnston, J. J., Kelley, R. I., Crawford, T. O., Morton, D. H., Agarwala, R., Koch, T., Schaffer, A. A., Francomano, C. A., Biesecker, L. G. A novel nemaline myopathy in the Amish caused by a mutation in troponin T1. Am. J. Hum. Genet. 67: 814-821, 2000. [PubMed: 10952871, images, related citations] [Full Text]

  8. Konersman, C. G., Freyermuth, F., Winder, T. L., Lawlor, M. W., Lagier-Tourenne, C., Patel, S. B. Novel autosomal dominant TNNT1 mutation causing nemaline myopathy. Molec. Genet. Genomic Med. 5: 678-691, 2017. [PubMed: 29178646, images, related citations] [Full Text]

  9. Lee, S., Eum, J., Park, S., Ki, S., Hwang, B. J., Kee, Y., Chae, J. H. TNNT1 myopathy with novel compound heterozygous mutations. Neuromusc. Disord. 32: 176-184, 2022. [PubMed: 35165004, related citations] [Full Text]

  10. Martin-Jimenez, P., Fuenmayor-Fernandez de la Hoz, C. P., Hernandez-Lain, A., Arteche-Lopez, A., Quesada-Espinosa, J. F., Voth, A. H., Vesperinas, A., Olive, M., Dominguez-Gonzalez, C. Adult-onset nemaline myopathy due to a novel homozygous variant in the TNNT1 gene. Muscle Nerve 66: E13-E15, 2022. [PubMed: 35833674, related citations] [Full Text]

  11. Nadal-Ginard, B., Mahdavi, V. Molecular basis of cardiac performance: plasticity of the myocardium generated through protein isoform switches. J. Clin. Invest. 84: 1693-1700, 1989. [PubMed: 2687327, related citations] [Full Text]

  12. Novelli, G., Gennarelli, M., Rocchi, M., Dallapiccola, B. Assignment of the slow troponin T (TNNT1) gene to chromosome 19 using polymerase chain reaction. Hum. Genet. 88: 697-698, 1992. [PubMed: 1551677, related citations] [Full Text]

  13. Novelli, G., Gennarelli, M., Zelano, G., Sangiuolo, F., Rocchi, M., Dallapiccola, B. Isolation and mapping of the slow skeletal troponin T using the polymerase chain reaction. (Abstract) Cytogenet. Cell Genet. 58: 2023, 1991.

  14. Pellerin, D., Aykanat, A., Ellezam, B., Troiano, E. C., Karamchandani, J., Dicaire, M.-J., Petitclerc, M., Robertson, R., Allard-Chamard, X., Brunet, D., Konersman, C. G., Mathieu, J., Warman Chardon, J., Gupta, V. A., Beggs, A. H., Brais, B., Chrestian, N. Novel recessive TNNT1 congenital core-rod myopathy in French Canadians. Ann. Neurol. 87: 568-583, 2020. [PubMed: 31970803, images, related citations] [Full Text]

  15. Samson, F., de Jong, P. J., Trask, B. J., Koza-Taylor, P., Speer, M. C., Potter, T., Roses, A. D., Gilbert, J. R. Assignment of the human slow skeletal troponin T gene to 19q13.4 using somatic cell hybrids and fluorescence in situ hybridization analysis. Genomics 13: 1374-1375, 1992. [PubMed: 1505979, related citations] [Full Text]

  16. Samson, F., Gilbert, J. R., Lee, J. E., Potter, T. G., Koza-Taylor, P., Speer, M. C., Bachinski, L. L., Siciliano, M. J., Roses, A. D. Isolation and localization of a human slow troponin T gene on chromosome 19q. (Abstract) Cytogenet. Cell Genet. 58: 2025, 1991.

  17. Samson, F., Lee, J. E., Hung, W. Y., Potter, T. G., Herbstreith, M., Roses, A. D., Gilbert, J. R. Isolation and localisation of a slow troponin (TNT) gene on chromosome 19 by subtraction of a cDNA muscle library using myotonic muscle cDNA. J. Neurosci. Res. 27: 441-451, 1990. [PubMed: 1706783, related citations] [Full Text]

  18. Samson, F., Mesnard, L., Mihovilovic, M., Potter, T. G., Mercadier, J.-J., Roses, A. D., Gilbert, J. R. A new human slow skeletal troponin T (TnTs) mRNA isoform derived from alternative splicing of a single gene. Biochem. Biophys. Res. Commun. 199: 841-847, 1994. [PubMed: 8135831, related citations] [Full Text]

  19. Spiro, A. J., Kennedy, C. Hereditary occurrence of nemaline myopathy. Arch. Neurol. 13: 155-159, 1965. [PubMed: 14315666, related citations] [Full Text]

  20. Trask, B., Fertitta, A., Christensen, M., Youngblom, J., Bergmann, A., Copeland, A., de Jong, P., Mohrenweiser, H., Olsen, A., Carrano, A., Tynan, K. Fluorescence in situ hybridization mapping of human chromosome 19: cytogenetic band location of 540 cosmids and 70 genes or DNA markers. Genomics 15: 133-145, 1993. [PubMed: 8432525, related citations] [Full Text]

  21. van der Pol, W. L., Leijenaar, J. F., Spliet, W. G. M., Lavrijsen, S. W., Jansen, N. J. G., Braun, K. P. J., Mulder, M., Timmers-Raaijmakers, B., Ratsma, K., Dooijes, D., van Haelst, M. M. Nemaline myopathy caused by TNNT1 mutations in a Dutch pedigree. Molec. Genet. Genomic Med. 2: 134-137, 2014. [PubMed: 24689076, images, related citations] [Full Text]

  22. Wang, X., Huang, Q.-Q., Breckenridge, M. T., Chen, A., Crawford, T. O., Morton, D. H., Jin, J.-P. Cellular fate of truncated slow skeletal muscle troponin T produced by glu180 nonsense mutation in Amish nemaline myopathy. J. Biol. Chem. 280: 13241-13249, 2005. [PubMed: 15665378, related citations] [Full Text]


Cassandra L. Kniffin - updated : 05/22/2023
Hilary J. Vernon - updated : 11/18/2020
Patricia A. Hartz - updated : 1/30/2009
Cassandra L. Kniffin - updated : 12/2/2008
Victor A. McKusick - updated : 10/19/2000
Creation Date:
Victor A. McKusick : 12/30/1989
alopez : 05/31/2023
alopez : 05/30/2023
alopez : 05/30/2023
ckniffin : 05/22/2023
carol : 11/18/2020
carol : 02/20/2014
joanna : 8/2/2013
carol : 8/2/2013
mgross : 1/30/2009
wwang : 12/8/2008
ckniffin : 12/2/2008
ckniffin : 4/4/2005
carol : 10/20/2004
carol : 10/24/2000
terry : 10/19/2000
dkim : 9/9/1998
terry : 6/3/1998
mark : 7/3/1997
joanna : 2/11/1996
jason : 6/13/1994
carol : 5/12/1993
carol : 2/11/1993
carol : 10/7/1992
carol : 9/16/1992
carol : 5/11/1992

* 191041

TROPONIN T1, SKELETAL, SLOW; TNNT1


Alternative titles; symbols

TROPONIN T


HGNC Approved Gene Symbol: TNNT1

SNOMEDCT: 1197155007;  


Cytogenetic location: 19q13.42     Genomic coordinates (GRCh38): 19:55,132,698-55,149,206 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
19q13.42 Nemaline myopathy 5A, autosomal recessive, severe infantile 605355 Autosomal recessive 3
Nemaline myopathy 5B, autosomal recessive, childhood-onset 620386 Autosomal recessive 3
Nemaline myopathy 5C, autosomal dominant 620389 Autosomal dominant 3

TEXT

Description

The TNNT1 gene encodes the slow skeletal muscle troponin. The sarcomere, the contractile element in muscle, including the myocardium, is constituted by 7 major proteins and several minor ones organized into thin and thick filaments. Each tropomyosin dimer (TPM1, 191010; TPM2, 190990) interacts with 7 actins and is associated with a troponin complex. Each complex is composed of 1 molecule of each of the 3 troponins: T, C, and I. This tropomyosin-troponin complex is responsible for the calcium sensitivity of the contractile apparatus (Nadal-Ginard and Mahdavi, 1989), and thus plays an important role in linking excitation to contraction in skeletal muscle.


Cloning and Expression

Samson et al. (1994) concluded that there are 4 isoforms of human slow skeletal troponin-T mRNA and that these probably result from combinatorial alternative splicing of a single gene.

By PCR of adult human skeletal muscle total RNA, Barton et al. (1999) cloned full-length TNNT1 and identified 3 shorter splice variants. The full-length protein contains 278 amino acids. Northern blot analysis detected TNNT1 expression in adult skeletal muscle, but not in adult heart and liver or in fetal heart.

Alternative splicing of exon 5 of the TNNT1 gene generates a high molecular weight (HMW) isoform (with exon 5) and a low molecular weight isoform (LMW) isoform (without exon 5). LMW TNNT1 has a higher binding affinity for tropomyosin and generates more calcium-activated contractile force than the HMW isoform, which enables regulation and modulation of muscle contractility. Normal adult slow muscles express mainly the HMW isoform (summary by Holling et al., 2022).


Gene Structure

Barton et al. (1999) determined that the TNNT1 gene contains 14 exons and spans more than 16 kb. The first exon is noncoding.


Mapping

Samson et al. (1990) assigned the slow skeletal isoform of troponin T (TNNT1) to chromosome 19q13.3-q13.4 using a panel of somatic cell hybrids and a cDNA clone as probe.

Novelli et al. (1991, 1992) confirmed the assignment of the TNNT1 gene to human chromosome 19 by showing the presence of a specific PCR product in hybrids retaining chromosome 19. The cardiac isoform of troponin-I (TNNI3; 191044) maps to the same region.

Samson et al. (1991) achieved regional assignment to the 19q13.2-qter region by analysis of 7 somatic cell hybrids containing different portions of chromosome 19. They excluded TNNT1 as a candidate gene for myotonic dystrophy (DM1; 160900) by the finding of obligate recombination events in family linkage studies.

Samson et al. (1992) mapped the TNNT1 gene to chromosome 19q13.4 by study of somatic cell hybrids and fluorescence in situ hybridization.

Barton et al. (1999) determined that the TNNI3 gene and the TNNT1 gene are oriented head to tail, with the TNNI3 gene 2.6 kb upstream of exon 1 of the TNNT1 gene.


Molecular Genetics

Autosomal Recessive Severe Infantile Nemaline Myopathy 5A

In Amish patients with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355), Johnston et al. (2000) identified homozygosity for a nonsense mutation in the TNNT1 gene (E180X; 191041.0001).

In a Dutch boy with NEM5A, van der Pol et al. (2014) identified compound heterozygous mutations in the TNNT1 gene: a splice site mutation (191041.0002) and an intragenic deletion (ex14del; 191041.0003). The mutations were inherited from the unaffected parents. Homozygosity for the splice site mutation was identified in 2 paternal cousins with a similar disorder who died in childhood of respiratory insufficiency.

In 3 unrelated patients with NEM5A, Geraud et al. (2021) identified homozygous or compound heterozygous mutations in the TNNT1 gene (see, e.g., 191041.0004-191041.0006). Western blot analysis of skeletal muscle from the patients showed absence of the TNNT1 protein. All died by 29 months of age.

Autosomal Recessive Childhood-Onset Nemaline Myopathy 5B

In 4 patients from 3 unrelated French Canadian families with autosomal recessive childhood-onset nemaline myopathy-5B (NEM5B; 620386), Pellerin et al. (2020) identified a homozygous missense mutation in the TNNT1 gene (L96P; 191041.0007). The mutation, which was found by next-generation sequencing and confirmed by Sanger sequencing, segregated with the disorder in 3 of the families who underwent testing. The mutation was unable to rescue the abnormal muscle phenotype in tnnt1-null zebrafish, suggesting that it causes a loss of function. The authors postulated that there was some residual activity or compensation by the low molecular weight TNNT1 isoform, resulting in a milder phenotype compared to patients with complete loss of TNNT1.

In 2 brothers, born of unrelated Korean parents, with NEM5B, Lee et al. (2022) identified compound heterozygosity for a missense mutation (A242P; 191041.0008) and a splice site mutation (191041.0009) in the TNNT1 gene. The mutations, which were found by whole-exome sequencing and confirmed by Sanger sequencing, were inherited from the unaffected parents. The splice site mutation was demonstrated to result in altered splicing and frameshifts, and the A242P mutation was unable to rescue the myopathic phenotype in tnnt1-null zebrafish, suggesting that both mutations caused a loss of function.

In a 51-year-old man with NEM5B, Martin-Jimenez et al. (2022) detected homozygosity for a variant in the TNNT1 gene (c.551_552delinsCA) that resulted in an arg184-to-pro (R184P) substitution. The variant was absent from gnomAD. Segregation studies and functional studies were not performed. The patient presented at age 50 years with a respiratory infection followed by respiratory arrest requiring intubation. He subsequently developed muscle weakness in all limbs which was slowly progressive and resulted in difficulty climbing stairs.

Autosomal Dominant Nemaline Myopathy 5C

In 8 affected members of a large consanguineous Ashkenazi Jewish family with autosomal dominant nemaline myopathy-5C (NEM5C; 620389) originally reported by Spiro and Kennedy (1965) and Gonatas et al. (1966), Konersman et al. (2017) identified a heterozygous missense mutation in the TNNT1 gene (E104V; 191041.0010). The mutation, which was found by Sanger sequencing of candidate genes, segregated with the disorder in the family. Holling et al. (2022) stated that the E104V mutation affects a residue just outside of the tropomyosin-binding site 1. In vitro studies in HEK293 cells transfected with the mutation showed normal expression of TNNT1 and effective binding to tropomyosin-3 (TPM3; 191030), although binding of the mutant protein to TPM3 was reduced by 35% in the presence of calcium.

In a mother and son with NEM5C, Holling et al. (2022) identified a heterozygous missense mutation in the TNNT1 gene (D65A; 191041.0011) that occurred within the tropomyosin-binding site 1 region. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the gnomAD database. In vitro studies in HEK293 cells transfected with the mutation showed normal expression of TNNT1, whereas coimmunoprecipitation studies showed significantly increased binding of the D65A variant to TPM3 compared to controls, both in the presence and absence of calcium.


Animal Model

Fox et al. (2018) described muscle features in 2 mouse models of Tnnt1 deficiency: a targeted gene knockout (Tnnt1 -/-) model and a site-specific knock-in (Tnnt1 c.505G-T) model. Quadratus femoris muscle tissue showed absence of Tnnt1, atrophy/hypotrophy of small type 1 myofibers, hypertrophy of fast type 2 fibers, and the presence of nemaline rods in both mouse models. Both models also showed impaired muscle force generation and exercise recovery in soleus muscles.

Pellerin et al. (2020) found that morpholino knockout of the tnnt1 gene in zebrafish caused reduced muscle mass, reduced birefringence, and severe truncal curvature compared to controls. Myofibers were disorganized and showed corelike intramyofibrillar areas of sarcomeric disorganization and Z-line thickening. These defects could be rescued by expression of wildtype human TNNT1 mRNA.


ALLELIC VARIANTS 11 Selected Examples):

.0001   NEMALINE MYOPATHY 5A, AUTOSOMAL RECESSIVE, SEVERE INFANTILE

TNNT1, GLU180TER
SNP: rs80358249, gnomAD: rs80358249, ClinVar: RCV000020554, RCV000024549

In affected individuals with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355), also known as Amish nemaline myopathy, Johnston et al. (2000) identified a homozygous c.579G-T transversion in exon 11 of the TNNT1 gene, resulting in a stop codon at amino acid 180 (E180X) and loss of 83 C-terminal residues. Johnston et al. (2000) had treated or obtained clinical information on 71 infants and young children from 33 nuclear Amish families with this form of nemaline myopathy. In the first months of life, affected infants had tremors with hypotonia and mild contractures of the shoulders and hips. Progressive worsening of the proximal contractures, weakness, and a pectus carinatum deformity developed before the children died of respiratory insufficiency, usually in the second year.

Variant Function

Jin et al. (2003) found complete loss of the TNNT1 protein in patients with the E180X mutation. The truncated protein results in elimination of the C-terminal T2 domain that interacts with troponin C (TNNC1; 191040), I (TNNI2; 191043), and tropomyosin (TPM1; 191010), but was predicted to retain a central tropomyosin-binding site that participates in the anchoring of troponin complex to the thin actin filament. If residual truncated protein was produced, this could potentially result in a dominant-negative effect. The significant muscle atrophy observed in the disorder was consistent with the slow Tnt isoform being involved in muscle development and growth.

In a patient with Amish nemaline myopathy due to the E180X mutation, Wang et al. (2005) detected residual mutant TNNT1 mRNA in muscle tissue but no corresponding translated mutant TNNT1 protein. In vitro functional expression studies in nonmuscle cells showed that E180X-mutant protein could be produced but was not detectable when expressed in muscle cells in vitro. The findings suggested rapid degradation of E180X-mutant protein in muscle cells, rather than a loss of nonsense mRNA to explain the absence of a dominant effect. Wang et al. (2005) postulated that inefficient incorporation of mutant TNNT1 into myofilaments results in its degradation by the muscle cell as a protective mechanism.


.0002   NEMALINE MYOPATHY 5A, AUTOSOMAL RECESSIVE, SEVERE INFANTILE

TNNT1, IVS8DS, G-A, +1
SNP: rs149559898, gnomAD: rs149559898, ClinVar: RCV001529807, RCV003230283

In a Dutch boy with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355), van der Pol et al. (2014) identified compound heterozygous mutations in the TNNT1 gene: a G-to-A transition in intron 8 (c.309+1G-A, NM_003283.4) inherited from the unaffected father, and an exon 14 deletion (ex14del; 191041.0003) inherited from the unaffected mother. Homozygosity for the c.309+1G-A mutation was identified in 2 paternal cousins with a similar disorder who died in childhood of respiratory insufficiency. The splicing defect resulted in the skipping of exon 8 with an in-frame deletion of 39 functionally important residues from the TNNT1 troponin domain (Asp65_Ile103del). The mutations were predicted to result in a loss of function, although the authors noted that the predicted transcripts could be translated into shortened proteins with some residual activity.


.0003   NEMALINE MYOPATHY 5A, AUTOSOMAL RECESSIVE, SEVERE INFANTILE

TNNT1, EX14 DEL
ClinVar: RCV003230310

For discussion of the exon 14 deletion (ex14del, NM_003283.4) in the TNNT1 gene that was found in compound heterozygous state in a patient with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355) by van der Pol et al. (2014), see 191041.0002.


.0004   NEMALINE MYOPATHY 5A, AUTOSOMAL RECESSIVE, SEVERE INFANTILE

TNNT1, GLU112TER
ClinVar: RCV003230311

In a female infant (patient 2) with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355) resulting in death at 16 months of age, Geraud et al. (2021) identified compound heterozygous mutations in the TNNT1 gene: a c.334G-T transversion (c.334G-T, NM_003283.5), resulting in a glu112-to-ter (E112X) substitution, and a C-to-A transversion in intron 4 (c.74-67C-A; 191041.0005), predicted to result in a splicing defect. Western blot analysis of skeletal muscle from the patient showed absence of the TNNT1 protein. She had a severe form of the disorder with tongue fasciculations, kyphosis, pectus carinatum, and hypotonia.


.0005   NEMALINE MYOPATHY 5A, AUTOSOMAL RECESSIVE, SEVERE INFANTILE

TNNT1, IVS4AS, C-A, -67
ClinVar: RCV003226144

For discussion of the c.74-67C-A mutation (c.74-67C-A, NM_003283.5) in intron 4 of the TNNT1 gene, resulting in a splicing defect, that was found in compound heterozygous state in a patient with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355) by Geraud et al. (2021) see 191041.0004.


.0006   NEMALINE MYOPATHY 5A, AUTOSOMAL RECESSIVE, SEVERE INFANTILE

TNNT1, GLU6TER
ClinVar: RCV003230312

In a patient (patient 3) with autosomal recessive severe infantile nemaline myopathy-5A (NEM5A; 605355) resulting in death at 6 months of age, Geraud et al. (2021) identified a homozygous c.16G-T transversion (cc.74-67C-A, NM_003283.5) in the TNNT1 gene, resulting in a glu6-to-ter (E6X) substitution. Western blot analysis of patient skeletal muscle showed absence of the TNNT1 protein.


.0007   NEMALINE MYOPATHY 5B, AUTOSOMAL RECESSIVE, CHILDHOOD-ONSET

TNNT1, LEU96PRO
ClinVar: RCV003230314

In 4 patients from 3 unrelated French Canadian families with autosomal recessive childhood-onset nemaline myopathy-5B (NEM5B; 620386), Pellerin et al. (2020) identified a homozygous c.287T-C transition (c.287T-C, NM_003283.6) in exon 8 of the TNNT1 gene, resulting in a leu96-to-pro (L96P) substitution at a conserved residue. The mutation, which was found by next-generation sequencing and confirmed by Sanger sequencing, segregated with the disorder in 3 of the families who underwent testing. The mutation was predicted to disrupt the alpha-helical secondary structure of the tropomyosin-binding site 1 and impair binding affinity. The mutant protein was likely to be misincorporated into myofilaments and undergo partial degradation, as evidenced by lower levels of the high molecular weight troponin T isoform in patient muscle; low molecular weight isoforms were retained. The mutation was unable to rescue the abnormal muscle phenotype in tnnt1-null zebrafish, suggesting that it causes a loss of function. The authors postulated that there was some residual activity or compensation by the low molecular weight isoform, resulting in a milder phenotype compared to patients with complete loss of TNNT1.

Variant Function

Holling et al. (2022) found that expression of the L96P mutation was increased compared to controls when transfected into HEK293 cells. Coimmunoprecipitation studies showed significantly reduced interaction (by 88%) of the L96P mutant with tropomyosin-3 (TPM3; 191030), and this interaction was abrogated in the presence of calcium.


.0008   NEMALINE MYOPATHY 5B, AUTOSOMAL RECESSIVE, CHILDHOOD-ONSET

TNNT1, ALA242PRO
ClinVar: RCV003230315

In 2 brothers, born of unrelated Korean parents, with autosomal recessive childhood-onset nemaline myopathy-5B (NEM5B; 620386), Lee et al. (2022) identified compound heterozygous mutations in the TNNT1 gene: a c.724G-C transversion (c.724G-C, NM_003283) in exon 12, resulting in an ala242-to-pro (A242P) substitution in the conserved troponin I-binding domain, and a G-to-A transition in intron 11 (c.611+1G-A; 191041.0009), resulting in a splicing defect and frameshifts. The mutations, which were found by whole-exome sequencing and confirmed by Sanger sequencing, were inherited from the unaffected parents. The splice site mutation was found once in the heterozygous state in a Korean individual in gnomAD (1 of 249,306 alleles, frequency of 4.01 x 10(-6)). The A242P mutation was unable to rescue the myopathic phenotype in tnnt1-null zebrafish, suggesting that this mutation causes a loss of function.


.0009   NEMALINE MYOPATHY 5B, AUTOSOMAL RECESSIVE, CHILDHOOD-ONSET

TNNT1, IVS11DS, G-A, +1
ClinVar: RCV003230313

For discussion of the G-to-A transition in intron 11 of the TNNT1 gene (c.611+1G-A, NM_003283), resulting in a splicing defect and frameshifts, that was found in compound heterozygous state in 2 brothers with autosomal recessive childhood-onset nemaline myopathy-5B (NEM5B; 620386) by Lee et al. (2022), see 191041.0008.


.0010   NEMALINE MYOPATHY 5C, AUTOSOMAL DOMINANT

TNNT1, GLU104VAL
SNP: rs2085441049, ClinVar: RCV001209655, RCV003230280

In 8 affected members of a large consanguineous Ashkenazi Jewish family with autosomal dominant nemaline myopathy-5C (NEM5C; 620389) originally reported by Spiro and Kennedy (1965) and Gonatas et al. (1966), Konersman et al. (2017) identified a heterozygous c.311A-T transversion (c.311A-T, NM_003283.4) in exon 9 of the TNNT1 gene, resulting in a glu104-to-val (E104V) substitution at a highly conserved residue in the tropomyosin-binding site 1 region. The mutation, which was found by Sanger sequencing of candidate genes, segregated with the disorder in the family. It was not present in public databases, including the Exome Variant Server, 1000 Genomes Project, and ExAC. Muscle samples from 2 patients showed normal levels of the TNNT1 protein, although low molecular weight (LMW) TNNT1 transcripts and protein levels were increased compared to controls, suggesting a compensatory mechanism. The authors postulated that the mutation could affect the affinity of TNNT1 for tropomyosin.

Variant Function

Holling et al. (2022) stated that the E104V mutation affects a residue just outside of the tropomyosin-binding site 1. In vitro studies in HEK293 cells transfected with the mutation showed normal expression of TNNT1 and effective binding to tropomyosin-3 (TPM3; 191030), although binding of the mutant protein to TPM3 was reduced by 35% in the presence of calcium.


.0011   NEMALINE MYOPATHY 5C, AUTOSOMAL DOMINANT

TNNT1, ASP65ALA
ClinVar: RCV003230316

In a mother and son with autosomal dominant nemaline myopathy-5C (NEM5C; 620389), Holling et al. (2022) identified a heterozygous c.194A-C transversion (c.194A-C, NM_003283.6) in exon 8 of the TNNT1 gene, resulting in an asp65-to-ala (D65A) substitution in the tropomyosin-binding site 1 region. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the gnomAD database. Genetic studies of the mother's parents showed that the mutation occurred de novo in the mother. The total amount of TNNT1 protein in the mother's skeletal muscle was similar to controls. Muscle biopsy from the mother showed type 1 fiber hypotrophy, type 2 fiber hypertrophy, increased amounts of fast skeletal fiber myosin-2a (MYH2; 160740), and increased actin levels, which may be compensatory mechanisms. In vitro studies in HEK293 cells transfected with the mutation showed normal expression of TNNT1, and coimmunoprecipitation studies showed significantly increased binding to tropomyosin-3 (TPM3; 191030), both in the presence and absence of calcium.


See Also:

Trask et al. (1993)

REFERENCES

  1. Barton, P. J. R., Cullen, M. E., Townsend, P. J., Brand, N. J., Mullen, A. J., Norman, D. A. M., Bhavsar, P. K., Yacoub, M. H. Close physical linkage of human troponin genes: organization, sequence, and expression of the locus encoding cardiac troponin I and slow skeletal troponin T. Genomics 57: 102-109, 1999. [PubMed: 10191089] [Full Text: https://doi.org/10.1006/geno.1998.5702]

  2. Fox, M. D., Carson, V. J., Feng, H.-Z., Lawlor, M. W., Gray, J. T., Brigatti, K. W., Jin, J.-P., Strauss, K. A. TNNT1 nemaline myopathy: natural history and therapeutic frontier. Hum. Molec. Genet. 27: 3272-3282, 2018. [PubMed: 29931346] [Full Text: https://doi.org/10.1093/hmg/ddy233]

  3. Geraud, J., Dieterich, K., Rendu, J., Uro Coste, E., Dobrzynski, M., Marcorelle, P., Ioos, C., Romero, N. B., Baudou, E., Brocard, J., Coville, A.-C., Faure, J., and 10 others. Clinical phenotype and loss of the slow skeletal muscle troponin T in three new patients with recessive TNNT1 nemaline myopathy. J. Med. Genet. 58: 602-608, 2021. [PubMed: 32994279] [Full Text: https://doi.org/10.1136/jmedgenet-2019-106714]

  4. Gonatas, N. K., Shy, G. M., Godfrey, E. H. Nemaline myopathy: the origin of nemaline structures. New Eng. J. Med. 274: 535-539, 1966. [PubMed: 5908457] [Full Text: https://doi.org/10.1056/NEJM196603102741002]

  5. Holling, T., Lisfeld, J., Johannsen, J., Matschke, J., Song, F., Altmeppen, H. C., Kutsche, K. Autosomal dominantly inherited myopathy likely caused by the TNNT1 variant p.(Asp65Ala). Hum. Mutat. 43: 1224-1233, 2022. [PubMed: 35510366] [Full Text: https://doi.org/10.1002/humu.24397]

  6. Jin, J.-P., Brotto, M. A., Hossain, M. M., Huang, Q.-Q., Brotto, L. S., Nosek, T. M., Morton, D. H., Crawford, T. O. Truncation by glu180 nonsense mutation results in complete loss of skeletal muscle troponin T in a lethal nemaline myopathy. J. Biol. Chem. 278: 26159-26165, 2003. [PubMed: 12732643] [Full Text: https://doi.org/10.1074/jbc.M303469200]

  7. Johnston, J. J., Kelley, R. I., Crawford, T. O., Morton, D. H., Agarwala, R., Koch, T., Schaffer, A. A., Francomano, C. A., Biesecker, L. G. A novel nemaline myopathy in the Amish caused by a mutation in troponin T1. Am. J. Hum. Genet. 67: 814-821, 2000. [PubMed: 10952871] [Full Text: https://doi.org/10.1086/303089]

  8. Konersman, C. G., Freyermuth, F., Winder, T. L., Lawlor, M. W., Lagier-Tourenne, C., Patel, S. B. Novel autosomal dominant TNNT1 mutation causing nemaline myopathy. Molec. Genet. Genomic Med. 5: 678-691, 2017. [PubMed: 29178646] [Full Text: https://doi.org/10.1002/mgg3.325]

  9. Lee, S., Eum, J., Park, S., Ki, S., Hwang, B. J., Kee, Y., Chae, J. H. TNNT1 myopathy with novel compound heterozygous mutations. Neuromusc. Disord. 32: 176-184, 2022. [PubMed: 35165004] [Full Text: https://doi.org/10.1016/j.nmd.2021.12.003]

  10. Martin-Jimenez, P., Fuenmayor-Fernandez de la Hoz, C. P., Hernandez-Lain, A., Arteche-Lopez, A., Quesada-Espinosa, J. F., Voth, A. H., Vesperinas, A., Olive, M., Dominguez-Gonzalez, C. Adult-onset nemaline myopathy due to a novel homozygous variant in the TNNT1 gene. Muscle Nerve 66: E13-E15, 2022. [PubMed: 35833674] [Full Text: https://doi.org/10.1002/mus.27678]

  11. Nadal-Ginard, B., Mahdavi, V. Molecular basis of cardiac performance: plasticity of the myocardium generated through protein isoform switches. J. Clin. Invest. 84: 1693-1700, 1989. [PubMed: 2687327] [Full Text: https://doi.org/10.1172/JCI114351]

  12. Novelli, G., Gennarelli, M., Rocchi, M., Dallapiccola, B. Assignment of the slow troponin T (TNNT1) gene to chromosome 19 using polymerase chain reaction. Hum. Genet. 88: 697-698, 1992. [PubMed: 1551677] [Full Text: https://doi.org/10.1007/BF02265301]

  13. Novelli, G., Gennarelli, M., Zelano, G., Sangiuolo, F., Rocchi, M., Dallapiccola, B. Isolation and mapping of the slow skeletal troponin T using the polymerase chain reaction. (Abstract) Cytogenet. Cell Genet. 58: 2023, 1991.

  14. Pellerin, D., Aykanat, A., Ellezam, B., Troiano, E. C., Karamchandani, J., Dicaire, M.-J., Petitclerc, M., Robertson, R., Allard-Chamard, X., Brunet, D., Konersman, C. G., Mathieu, J., Warman Chardon, J., Gupta, V. A., Beggs, A. H., Brais, B., Chrestian, N. Novel recessive TNNT1 congenital core-rod myopathy in French Canadians. Ann. Neurol. 87: 568-583, 2020. [PubMed: 31970803] [Full Text: https://doi.org/10.1002/ana.25685]

  15. Samson, F., de Jong, P. J., Trask, B. J., Koza-Taylor, P., Speer, M. C., Potter, T., Roses, A. D., Gilbert, J. R. Assignment of the human slow skeletal troponin T gene to 19q13.4 using somatic cell hybrids and fluorescence in situ hybridization analysis. Genomics 13: 1374-1375, 1992. [PubMed: 1505979] [Full Text: https://doi.org/10.1016/0888-7543(92)90077-6]

  16. Samson, F., Gilbert, J. R., Lee, J. E., Potter, T. G., Koza-Taylor, P., Speer, M. C., Bachinski, L. L., Siciliano, M. J., Roses, A. D. Isolation and localization of a human slow troponin T gene on chromosome 19q. (Abstract) Cytogenet. Cell Genet. 58: 2025, 1991.

  17. Samson, F., Lee, J. E., Hung, W. Y., Potter, T. G., Herbstreith, M., Roses, A. D., Gilbert, J. R. Isolation and localisation of a slow troponin (TNT) gene on chromosome 19 by subtraction of a cDNA muscle library using myotonic muscle cDNA. J. Neurosci. Res. 27: 441-451, 1990. [PubMed: 1706783] [Full Text: https://doi.org/10.1002/jnr.490270403]

  18. Samson, F., Mesnard, L., Mihovilovic, M., Potter, T. G., Mercadier, J.-J., Roses, A. D., Gilbert, J. R. A new human slow skeletal troponin T (TnTs) mRNA isoform derived from alternative splicing of a single gene. Biochem. Biophys. Res. Commun. 199: 841-847, 1994. [PubMed: 8135831] [Full Text: https://doi.org/10.1006/bbrc.1994.1305]

  19. Spiro, A. J., Kennedy, C. Hereditary occurrence of nemaline myopathy. Arch. Neurol. 13: 155-159, 1965. [PubMed: 14315666] [Full Text: https://doi.org/10.1001/archneur.1965.00470020045006]

  20. Trask, B., Fertitta, A., Christensen, M., Youngblom, J., Bergmann, A., Copeland, A., de Jong, P., Mohrenweiser, H., Olsen, A., Carrano, A., Tynan, K. Fluorescence in situ hybridization mapping of human chromosome 19: cytogenetic band location of 540 cosmids and 70 genes or DNA markers. Genomics 15: 133-145, 1993. [PubMed: 8432525] [Full Text: https://doi.org/10.1006/geno.1993.1021]

  21. van der Pol, W. L., Leijenaar, J. F., Spliet, W. G. M., Lavrijsen, S. W., Jansen, N. J. G., Braun, K. P. J., Mulder, M., Timmers-Raaijmakers, B., Ratsma, K., Dooijes, D., van Haelst, M. M. Nemaline myopathy caused by TNNT1 mutations in a Dutch pedigree. Molec. Genet. Genomic Med. 2: 134-137, 2014. [PubMed: 24689076] [Full Text: https://doi.org/10.1002/mgg3.52]

  22. Wang, X., Huang, Q.-Q., Breckenridge, M. T., Chen, A., Crawford, T. O., Morton, D. H., Jin, J.-P. Cellular fate of truncated slow skeletal muscle troponin T produced by glu180 nonsense mutation in Amish nemaline myopathy. J. Biol. Chem. 280: 13241-13249, 2005. [PubMed: 15665378] [Full Text: https://doi.org/10.1074/jbc.M413696200]


Contributors:
Cassandra L. Kniffin - updated : 05/22/2023
Hilary J. Vernon - updated : 11/18/2020
Patricia A. Hartz - updated : 1/30/2009
Cassandra L. Kniffin - updated : 12/2/2008
Victor A. McKusick - updated : 10/19/2000

Creation Date:
Victor A. McKusick : 12/30/1989

Edit History:
alopez : 05/31/2023
alopez : 05/30/2023
alopez : 05/30/2023
ckniffin : 05/22/2023
carol : 11/18/2020
carol : 02/20/2014
joanna : 8/2/2013
carol : 8/2/2013
mgross : 1/30/2009
wwang : 12/8/2008
ckniffin : 12/2/2008
ckniffin : 4/4/2005
carol : 10/20/2004
carol : 10/24/2000
terry : 10/19/2000
dkim : 9/9/1998
terry : 6/3/1998
mark : 7/3/1997
joanna : 2/11/1996
jason : 6/13/1994
carol : 5/12/1993
carol : 2/11/1993
carol : 10/7/1992
carol : 9/16/1992
carol : 5/11/1992