Entry - *600417 - 5-PRIME-NUCLEOTIDASE, CYTOSOLIC II; NT5C2 - OMIM
 
* 600417

5-PRIME-NUCLEOTIDASE, CYTOSOLIC II; NT5C2


Alternative titles; symbols

NUCLEOTIDASE, 5-PRIME, CYTOSOLIC II
NUCLEOTIDASE, 5-PRIME, TYPE B; NT5B
PURINE 5-PRIME-NUCLEOTIDASE; PNT5


HGNC Approved Gene Symbol: NT5C2

Cytogenetic location: 10q24.32-q24.33     Genomic coordinates (GRCh38): 10:103,088,017-103,193,272 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
10q24.32-q24.33 Spastic paraplegia 45, autosomal recessive 613162 AR 3

TEXT

Description

Purine 5-prime-nucleotidase (EC 3.1.3.5) preferentially hydrolyzes inosine 5-prime-monophosphate (IMP) and other purine nucleotides, and is allosterically activated by various compounds, including ATP. The enzyme is exclusively located in the cytoplasmic matrix of cells and may have a critical role in the maintenance of a constant composition of intracellular purine/pyrimidine nucleotides in cooperation with other nucleotidases (summary by Oka et al., 1994).


Cloning and Expression

Oka et al. (1994) found 2 distinct but closely related types of cDNAs for purine 5-prime-nucleotidase in a chicken liver cDNA library. The encoded A- and B-type proteins were similar throughout a central core of 472 amino acids sharing 79% sequence identity. Using degenerate oligonucleotides based on identical portions of amino acid sequences between A- and B-type subunits of the chicken enzyme, Oka et al. (1994) isolated human cytosolic purine 5-prime-nucleotidase cDNA clones with PCR. The predicted open reading frame encoded a protein of 561 amino acids with a molecular mass of 64,966 Da. The deduced amino acid sequence exhibited 95% identity with the sequence of the B-type subunit of chicken enzyme. Northern blot analysis of placental polyadenylated RNA revealed a single band of 3.6 kb.


Mapping

Gross (2014) mapped the NT5C2 gene to chromosome 10q24.32-q24.33 based on an alignment of the NT5C2 sequence (GenBank BC001595) with the genomic sequence (GRCh37).


Molecular Genetics

Spastic Paraplegia 45

In affected members of the consanguineous Turkish family with spastic paraplegia-45 (SPG45; 613162) mapped to chromosome 10q24-q25 by Dursun et al. (2009), Novarino et al. (2014) identified a homozygous nonsense mutation in the NT5C2 gene (R29X; 600417.0001). In affected members of 4 other families with SPG45 recognized from a large exome sequencing project of consanguineous families with neurologic disease, they identified homozygosity for 1 other nonsense (600417.0004), 1 frameshift (600417.0002), and 2 splice site mutations (600417.0003; 600417.0005) in the NT5C2 gene.

Acute Lymphoblastic Leukemia Relapse

Heterozygous activating mutations in NT5C2 are present in about 20% of relapsed pediatric T-cell ALL (613065) and in 3 to 10% of relapsed B-precursor ALL, and an arg367-to-gln (R367Q) change is the most common NT5C2 mutation found in relapsed ALL. Tzoneva et al. (2018) used a conditional and inducible leukemia model to demonstrate that expression of NT5C2(R367Q) induces resistance to chemotherapy with 6-mercaptopurine at the cost of impaired leukemia cell growth and leukemia-initiating cell activity. The loss-of-fitness phenotype of NT5C2 +/R367Q mutant cells was associated with excess export of purines to the extracellular space and depletion of the intracellular purine-nucleotide pool. Consequently, blocking guanosine synthesis by inhibition of inosine-5-prime-monophosphate dehydrogenase (IMPDH) induced increased cytotoxicity against NT5C2-mutant leukemia lymphoblasts. Tzoneva et al. (2018) concluded that these results identified the fitness cost of NT5C2 mutation and resistance to chemotherapy as key evolutionary drivers that shape clonal evolution in relapsed ALL and supported a role for IMPDH inhibition in the treatment of ALL.


Animal Model

Johanns et al. (2019) found that NT5C2 knockout mice had reduced body weight, adipose tissue, and insulin resistance, with improved glucose tolerance compared to wildtype mice when both were fed a high fat diet. The authors provided evidence suggesting that this effect is due to increased responsiveness of skeletal muscle to insulin.


ALLELIC VARIANTS ( 5 Selected Examples):

.0001 SPASTIC PARAPLEGIA 45

SPG45, ARG29TER
  
RCV000087268...

In the 2 Turkish sibs with spastic paraplegia-45 (SPG45; 613162) reported by Dursun et al. (2009), Novarino et al. (2014) identified a homozygous c.86G-A transition in the NT5C2 gene, resulting in an arg29-to-ter (R29X) substitution.


.0002 SPASTIC PARAPLEGIA 45

SPG45, 1-BP DEL, 1225A
  
RCV000087269

In 2 affected sisters from a consanguineous family (family 1290) segregating spastic paraplegia-45 (SPG45; 613162), Novarino et al. (2014) identified homozygosity for a 1-bp deletion (c.1225delA) in the NT5C2 gene, resulting in a frameshift and premature termination (Ser409Valfs436Ter). None of the unaffected family members were homozygous for this mutation.


.0003 SPASTIC PARAPLEGIA 45

SPG45, c.988-1G-T
  
RCV000087270

In 2 affected sisters from a consanguineous family (family 1549) segregating spastic paraplegia-45 (SPG45; 613162), Novarino et al. (2014) identified homozygosity for an acceptor splice site mutation (c.988-1G-T) in the NT5C2 gene. None of the unaffected family members were homozygous for this mutation.


.0004 SPASTIC PARAPLEGIA 45

SPG45, ARG149TER
  
RCV000087271

In 2 affected brothers from a consanguineous family (family 1829) segregating spastic paraplegia-45 (SPG45; 613162), Novarino et al. (2014) identified homozygosity for a c.445A-T transversion in the NT5C2 gene, resulting in an arg149-to-ter (R149X) substitution. None of the unaffected family members were homozygous for this mutation.


.0005 SPASTIC PARAPLEGIA 45

SPG45, c.175+1G-A
  
RCV000087272

In 2 affected sibs from a consanguineous family (family 659) segregating spastic paraplegia-45 (SPG45; 613162), Novarino et al. (2014) identified homozygosity for a donor splice site mutation (c.175+1G-A). None of the unaffected family members were homozygous for this mutation.


REFERENCES

  1. Dursun, U., Koroglu, C., Orhan, E.K., Ugur, S. A., Tolun, A. Autosomal recessive spastic paraplegia (SPG45) with mental retardation maps to 10q24.3-q25.1. Neurogenetics 10: 325-331, 2009. [PubMed: 19415352, related citations] [Full Text]

  2. Gross, M. B. Personal Communication. Baltimore, Md. 2/27/2014.

  3. Johanns, M., Kviklyte, S., Chuang, S.-J., Corbeels, K., Jacobs, R., Herinckx, G., Vertommen, D, Schakman, O., Duparc, T., Cani, P. D., Bouzin, C., Andersen, H., Bohlooly-Y, M., Van der Schueren, B., Oscarsson, J., Rider, M. H. Genetic deletion of soluble 5-prime-nucleotidase II reduces body weight gain and insulin resistance induced by a high-fat diet. Molec. Genet. Metab. 126: 377-387, 2019. [PubMed: 30803894, related citations] [Full Text]

  4. Novarino, G., Fenstermaker, A. G., Zaki, M. S., Hofree, M., Silhavy, J., Heiberg, A. D., Abdellateef, M., Rosti, B., Scott, E., Mansour, L., Masri, A., Kayserili, H., and 41 others. Exome sequencing links corticospinal motor neuron disease to common neurodegenerative disorders. Science 343: 506-511, 2014. [PubMed: 24482476, images, related citations] [Full Text]

  5. Oka, J., Matsumoto, A., Hosokawa, Y., Inoue, S. Molecular cloning of human cytosolic purine 5-prime-nucleotidase. Biochem. Biophys. Res. Commun. 205: 917-922, 1994. [PubMed: 7999131, related citations] [Full Text]

  6. Tzoneva, G., Dieck, C. L., Oshima, K., Ambesi-Impiombato, A., Sanchez-Martin, M., Madubata, C. J., Khiabanian, H., Yu, J., Waanders, E., Iacobucci, I., Sulis, M. L., Kato, M., and 9 others. Clonal evolution mechanisms in NT5C2 mutant-relapsed acute lymphoblastic leukaemia. Nature 553: 511-514, 2018. [PubMed: 29342136, related citations] [Full Text]


Hilary J. Vernon - updated : 05/12/2020
Ada Hamosh - updated : 08/21/2018
Matthew B. Gross - updated : 02/27/2014
Ada Hamosh - updated : 2/27/2014
Joanna S. Amberger - updated : 4/19/2002
Creation Date:
Victor A. McKusick : 2/17/1995
carol : 05/12/2020
alopez : 08/21/2018
mgross : 02/27/2014
carol : 2/27/2014
carol : 10/12/2004
joanna : 4/19/2002
joanna : 4/19/2002
mgross : 7/17/2000
carol : 2/17/1995

* 600417

5-PRIME-NUCLEOTIDASE, CYTOSOLIC II; NT5C2


Alternative titles; symbols

NUCLEOTIDASE, 5-PRIME, CYTOSOLIC II
NUCLEOTIDASE, 5-PRIME, TYPE B; NT5B
PURINE 5-PRIME-NUCLEOTIDASE; PNT5


HGNC Approved Gene Symbol: NT5C2

Cytogenetic location: 10q24.32-q24.33     Genomic coordinates (GRCh38): 10:103,088,017-103,193,272 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
10q24.32-q24.33 Spastic paraplegia 45, autosomal recessive 613162 Autosomal recessive 3

TEXT

Description

Purine 5-prime-nucleotidase (EC 3.1.3.5) preferentially hydrolyzes inosine 5-prime-monophosphate (IMP) and other purine nucleotides, and is allosterically activated by various compounds, including ATP. The enzyme is exclusively located in the cytoplasmic matrix of cells and may have a critical role in the maintenance of a constant composition of intracellular purine/pyrimidine nucleotides in cooperation with other nucleotidases (summary by Oka et al., 1994).


Cloning and Expression

Oka et al. (1994) found 2 distinct but closely related types of cDNAs for purine 5-prime-nucleotidase in a chicken liver cDNA library. The encoded A- and B-type proteins were similar throughout a central core of 472 amino acids sharing 79% sequence identity. Using degenerate oligonucleotides based on identical portions of amino acid sequences between A- and B-type subunits of the chicken enzyme, Oka et al. (1994) isolated human cytosolic purine 5-prime-nucleotidase cDNA clones with PCR. The predicted open reading frame encoded a protein of 561 amino acids with a molecular mass of 64,966 Da. The deduced amino acid sequence exhibited 95% identity with the sequence of the B-type subunit of chicken enzyme. Northern blot analysis of placental polyadenylated RNA revealed a single band of 3.6 kb.


Mapping

Gross (2014) mapped the NT5C2 gene to chromosome 10q24.32-q24.33 based on an alignment of the NT5C2 sequence (GenBank BC001595) with the genomic sequence (GRCh37).


Molecular Genetics

Spastic Paraplegia 45

In affected members of the consanguineous Turkish family with spastic paraplegia-45 (SPG45; 613162) mapped to chromosome 10q24-q25 by Dursun et al. (2009), Novarino et al. (2014) identified a homozygous nonsense mutation in the NT5C2 gene (R29X; 600417.0001). In affected members of 4 other families with SPG45 recognized from a large exome sequencing project of consanguineous families with neurologic disease, they identified homozygosity for 1 other nonsense (600417.0004), 1 frameshift (600417.0002), and 2 splice site mutations (600417.0003; 600417.0005) in the NT5C2 gene.

Acute Lymphoblastic Leukemia Relapse

Heterozygous activating mutations in NT5C2 are present in about 20% of relapsed pediatric T-cell ALL (613065) and in 3 to 10% of relapsed B-precursor ALL, and an arg367-to-gln (R367Q) change is the most common NT5C2 mutation found in relapsed ALL. Tzoneva et al. (2018) used a conditional and inducible leukemia model to demonstrate that expression of NT5C2(R367Q) induces resistance to chemotherapy with 6-mercaptopurine at the cost of impaired leukemia cell growth and leukemia-initiating cell activity. The loss-of-fitness phenotype of NT5C2 +/R367Q mutant cells was associated with excess export of purines to the extracellular space and depletion of the intracellular purine-nucleotide pool. Consequently, blocking guanosine synthesis by inhibition of inosine-5-prime-monophosphate dehydrogenase (IMPDH) induced increased cytotoxicity against NT5C2-mutant leukemia lymphoblasts. Tzoneva et al. (2018) concluded that these results identified the fitness cost of NT5C2 mutation and resistance to chemotherapy as key evolutionary drivers that shape clonal evolution in relapsed ALL and supported a role for IMPDH inhibition in the treatment of ALL.


Animal Model

Johanns et al. (2019) found that NT5C2 knockout mice had reduced body weight, adipose tissue, and insulin resistance, with improved glucose tolerance compared to wildtype mice when both were fed a high fat diet. The authors provided evidence suggesting that this effect is due to increased responsiveness of skeletal muscle to insulin.


ALLELIC VARIANTS 5 Selected Examples):

.0001   SPASTIC PARAPLEGIA 45

SPG45, ARG29TER
SNP: rs587777173, ClinVar: RCV000087268, RCV001310575

In the 2 Turkish sibs with spastic paraplegia-45 (SPG45; 613162) reported by Dursun et al. (2009), Novarino et al. (2014) identified a homozygous c.86G-A transition in the NT5C2 gene, resulting in an arg29-to-ter (R29X) substitution.


.0002   SPASTIC PARAPLEGIA 45

SPG45, 1-BP DEL, 1225A
SNP: rs886037656, ClinVar: RCV000087269

In 2 affected sisters from a consanguineous family (family 1290) segregating spastic paraplegia-45 (SPG45; 613162), Novarino et al. (2014) identified homozygosity for a 1-bp deletion (c.1225delA) in the NT5C2 gene, resulting in a frameshift and premature termination (Ser409Valfs436Ter). None of the unaffected family members were homozygous for this mutation.


.0003   SPASTIC PARAPLEGIA 45

SPG45, c.988-1G-T
SNP: rs886037657, ClinVar: RCV000087270

In 2 affected sisters from a consanguineous family (family 1549) segregating spastic paraplegia-45 (SPG45; 613162), Novarino et al. (2014) identified homozygosity for an acceptor splice site mutation (c.988-1G-T) in the NT5C2 gene. None of the unaffected family members were homozygous for this mutation.


.0004   SPASTIC PARAPLEGIA 45

SPG45, ARG149TER
SNP: rs587777174, ClinVar: RCV000087271

In 2 affected brothers from a consanguineous family (family 1829) segregating spastic paraplegia-45 (SPG45; 613162), Novarino et al. (2014) identified homozygosity for a c.445A-T transversion in the NT5C2 gene, resulting in an arg149-to-ter (R149X) substitution. None of the unaffected family members were homozygous for this mutation.


.0005   SPASTIC PARAPLEGIA 45

SPG45, c.175+1G-A
SNP: rs886037658, ClinVar: RCV000087272

In 2 affected sibs from a consanguineous family (family 659) segregating spastic paraplegia-45 (SPG45; 613162), Novarino et al. (2014) identified homozygosity for a donor splice site mutation (c.175+1G-A). None of the unaffected family members were homozygous for this mutation.


REFERENCES

  1. Dursun, U., Koroglu, C., Orhan, E.K., Ugur, S. A., Tolun, A. Autosomal recessive spastic paraplegia (SPG45) with mental retardation maps to 10q24.3-q25.1. Neurogenetics 10: 325-331, 2009. [PubMed: 19415352] [Full Text: https://doi.org/10.1007/s10048-009-0191-3]

  2. Gross, M. B. Personal Communication. Baltimore, Md. 2/27/2014.

  3. Johanns, M., Kviklyte, S., Chuang, S.-J., Corbeels, K., Jacobs, R., Herinckx, G., Vertommen, D, Schakman, O., Duparc, T., Cani, P. D., Bouzin, C., Andersen, H., Bohlooly-Y, M., Van der Schueren, B., Oscarsson, J., Rider, M. H. Genetic deletion of soluble 5-prime-nucleotidase II reduces body weight gain and insulin resistance induced by a high-fat diet. Molec. Genet. Metab. 126: 377-387, 2019. [PubMed: 30803894] [Full Text: https://doi.org/10.1016/j.ymgme.2019.01.017]

  4. Novarino, G., Fenstermaker, A. G., Zaki, M. S., Hofree, M., Silhavy, J., Heiberg, A. D., Abdellateef, M., Rosti, B., Scott, E., Mansour, L., Masri, A., Kayserili, H., and 41 others. Exome sequencing links corticospinal motor neuron disease to common neurodegenerative disorders. Science 343: 506-511, 2014. [PubMed: 24482476] [Full Text: https://doi.org/10.1126/science.1247363]

  5. Oka, J., Matsumoto, A., Hosokawa, Y., Inoue, S. Molecular cloning of human cytosolic purine 5-prime-nucleotidase. Biochem. Biophys. Res. Commun. 205: 917-922, 1994. [PubMed: 7999131] [Full Text: https://doi.org/10.1006/bbrc.1994.2752]

  6. Tzoneva, G., Dieck, C. L., Oshima, K., Ambesi-Impiombato, A., Sanchez-Martin, M., Madubata, C. J., Khiabanian, H., Yu, J., Waanders, E., Iacobucci, I., Sulis, M. L., Kato, M., and 9 others. Clonal evolution mechanisms in NT5C2 mutant-relapsed acute lymphoblastic leukaemia. Nature 553: 511-514, 2018. [PubMed: 29342136] [Full Text: https://doi.org/10.1038/nature25186]


Contributors:
Hilary J. Vernon - updated : 05/12/2020
Ada Hamosh - updated : 08/21/2018
Matthew B. Gross - updated : 02/27/2014
Ada Hamosh - updated : 2/27/2014
Joanna S. Amberger - updated : 4/19/2002

Creation Date:
Victor A. McKusick : 2/17/1995

Edit History:
carol : 05/12/2020
alopez : 08/21/2018
mgross : 02/27/2014
carol : 2/27/2014
carol : 10/12/2004
joanna : 4/19/2002
joanna : 4/19/2002
mgross : 7/17/2000
carol : 2/17/1995